1
|
Lin J, Wei Y, Gu X, Liu M, Wang M, Zhou R, Zou D, Yin L, Zhou C, Hu D. Nanotherapeutics-mediated restoration of pancreatic homeostasis and intestinal barrier for the treatment of severe acute pancreatitis. J Control Release 2025; 377:93-105. [PMID: 39542256 DOI: 10.1016/j.jconrel.2024.11.022] [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: 08/12/2024] [Revised: 10/03/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
Severe acute pancreatitis (SAP) is an inflammatory disease of the pancreas accompanied with intestinal injury, and effective therapeutic modalities are still highly lacking. Herein, a facile and effective nanotherapeutics (pHA@IBNCs) is developed to alleviate pancreatic inflammation and restore intestinal barrier for SAP treatment. Epigallocatechin gallate (EGCG, an anti-oxidant), interleukin-22 (IL-22, an anti-inflammatory and epithelial barrier-protecting cytokine), and bovine serum albumin (a framework protein), are assembled via non-covalent interactions to form nanocomplexes (IBNCs). Then, phenylboronic acid-modified hyaluronic acid (pHA) is synthesized and coated onto IBNCs via formation of the reversible boronate ester bonds to obtain pHA@IBNCs. Upon intravenous injection, pHA@IBNCs could efficiently accumulate at the lesion sites of sodium taurocholate (STC)-induced SAP mice, based on their prolonged blood circulation time and pHA-mediated targeting of activated intestinal epithelial cells and macrophages. Inside the inflammatory microenvironment, over-produced reactive oxygen species (ROS) trigger the shedding of the pHA layer and release of the drug payloads. Thereby, EGCG cooperates with IL-22 to attenuate pancreatitis and restore the intestinal barrier by scavenging ROS, suppressing pro-inflammatory cytokines secretion, and promoting the repair of intestinal epithelia. Such a nano-therapeutic approach targeting multiple pathological events may serve as a promising paradigm for the effective management of SAP.
Collapse
Affiliation(s)
- Juanhui Lin
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Yuansong Wei
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Xiaxian Gu
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Miaoru Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Mengru Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Renxiang Zhou
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Duowu Zou
- Department of Gastroenterology, Ruijin Hosptial, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lichen Yin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China.
| | - Chunhua Zhou
- Department of Gastroenterology, Ruijin Hosptial, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Duanmin Hu
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China.
| |
Collapse
|
2
|
Wang Y, Deng M, Wu Y, Zheng C, Zhang F, Guo C, Zhang B, Hu C, Kong Q, Wang Y. A multifunctional mitochondria-protective gene delivery platform promote intervertebral disc regeneration. Biomaterials 2024; 317:123067. [PMID: 39742837 DOI: 10.1016/j.biomaterials.2024.123067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/09/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025]
Abstract
Intervertebral disc degeneration (IDD) is a deleterious condition driven by localized inflammation and the associated disruption of the normal homeostatic balance between anabolism and catabolism, contributing to progressive functional abnormalities within the nucleus pulposus (NP). Despite our prior evidence demonstrating that a miR-21 inhibitor can have regenerative effects that counteract the progression of IDD, its application for IDD treatment remains limited by the inadequacy of current local delivery systems. Here, an injectable tannic acid (TA)-loaded hydrogel gene delivery system was developed and used for the encapsulation of a multifunctional mitochondria-protecting gene nanocarrier (PHs). This engineered platform was designed for the sustained on-demand delivery of both miR-21 inhibitor and ss-31 (mitochondrial-targeted peptide) constructs to the NP. This prepared hydrogel could be implanted into the intervertebral disc using a minimally invasive approach whereupon it was able to rapidly release TA. Sustained PHs release was then achieved as appropriate through a mechanism mediated by the activity of MMP-2. Following the targeted uptake of PHs by degenerated NP cells, the subsequent release of encapsulated miR-21 inhibitor suppressed apoptotic cell death and modulated the metabolism of the extracellular matrix (ECM) by targeting the Spry1 gene. At the same time, ss-31 was able to target damaged mitochondria and alleviate inflammatory activity via the suppression of mitochondrial ROS-NLRP3-IL-1β/Caspase1 pathway activity. Synergistic ECM regeneration and anti-inflammatory effects were sufficient to provide therapeutic benefits in an in vivo model of IDD. Together, these results thus highlight this hydrogel-based gene delivery platform as a promising novel approach to the treatment of IDD.
Collapse
Affiliation(s)
- Yu Wang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Mingyan Deng
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ye Wu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Cheng Zheng
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, China
| | - Fanjun Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, China
| | - Chuan Guo
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bo Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, China
| | - Cheng Hu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, China
| | - Qingquan Kong
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
3
|
Zhou Q, Huang G, Si J, Wu Y, Jin S, Ji Y, Ge Z. Potent Covalent Organic Framework Nanophotosensitizers with Staggered Type I/II Motifs for Photodynamic Immunotherapy of Hypoxic Tumors. ACS NANO 2024. [PMID: 39698912 DOI: 10.1021/acsnano.4c14555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Photodynamic therapy (PDT) using oxygen-dependent type II photosensitizers is frequently limited by the hypoxic microenvironment of solid tumors. Type I photosensitizers show oxygen-independent reactive oxygen species (ROS) generation upon light irradiation but still face the challenges of aggregation-caused quenching (ACQ) and low efficiency to produce ROS. Herein, we first prepare an efficient type I photosensitizer from a perylene derivative via intramolecular donor-acceptor binding and sulfur substitution, which significantly enhance intersystem crossing between singlet and triplet states and electron transfer capability. After reaction with a type II photosensitizer, the covalent organic framework (COF) nanophotosensitizer is formed with alternated type I and II photosensitizer motifs in the same layer and staggered AB stacking between layers to avoid ACQ. The nanophotosensitizer exhibits high-efficiency generation of singlet oxygen (1O2) and superoxide anion radicals (O2•-) via type I and II mechanism under normoxia upon exposure to light irradiation. Under hypoxia, massive O2•- can be produced continuously. The potent ROS generation capability results in efficient cellular apoptosis and immunogenic cell death (ICD) efficiently. After combination with immune checkpoint inhibitors, tumor immunosuppressive microenvironment is reversed, which effectively ablates bulky hypoxic primary tumors and suppresses metastases via photodynamic immunotherapy. The COF nanophotosensitizers with staggered type I and II photosensitizer motifs represent a promising strategy to boost photodynamic immunotherapy of hypoxic tumors.
Collapse
Affiliation(s)
- Qinghao Zhou
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Guopu Huang
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Jiale Si
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Youshen Wu
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, 28 West Xianning Road, Xi'an 710049, Shaanxi, China
| | - Shangbin Jin
- School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Yuanyuan Ji
- Department of Geriatric General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| | - Zhishen Ge
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
- Department of Geriatric General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China
| |
Collapse
|
4
|
Li L, Feng C, Zhang W, Qi L, Liu B, Wang H, Li C, Li Z, Tu C, Zhou W. Mitigation of Cisplatin-Induced Nephrotoxicity and Augmentation of Anticancer Potency via Tea Polyphenol Nanoparticles' Codelivery of siRNA from CRISPR/Cas9 Screened Targets. ACS APPLIED MATERIALS & INTERFACES 2024; 16:59721-59737. [PMID: 39460726 DOI: 10.1021/acsami.4c10171] [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: 10/28/2024]
Abstract
Cisplatin, a frontline chemotherapeutic agent against cancer, faces challenges in clinical application due to significant toxicities and suboptimal efficacy. Renal toxicity, a dose-limiting factor of cisplatin, results from multifactorial processes including cisplatin-induced cellular pyroptosis, oxidative damage, and inflammatory responses. Our findings reveal that Tea Polyphenols Nanoparticles (TPNs) derived from Epigallocatechin gallate (EGCG) effectively could address these diverse mechanisms, comprehensively alleviating cisplatin-induced nephrotoxicity. Leveraging TPNs as carriers, chemical conjugation enables the encapsulation of tetravalent cisplatin prodrug, extending its systemic half-life, enhancing tumor tissue accumulation, while simultaneously mitigating renal toxicity. Concurrently, employing a CRISPR/Cas9 kinase library, we identified CSNK2A1 as a target sensitizing tumor cells to cisplatin, enabling specific siRNA sequences to augment cisplatin susceptibility, thereby minimizing the dosage requirement. Benefiting from the versatile carrier properties of TPNs to codeliver cisplatin prodrug and anti-CSNK2A1 siRNA, we developed a codelivery system, Pt-TPNs/siRNA. Pt-TPNs/siRNA not only enhances the anticancer effects but also mitigates cisplatin-induced renal toxicity, achieving efficacy while reducing toxicity. Mechanistic and safety assessments of these nanoparticles were conducted at both cellular and animal levels, opening new avenues for improved clinical utilization of cisplatin.
Collapse
Affiliation(s)
- Lingjiao Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
- Loudi Central Hospital, Loudi 417000, China
| | - Chengyao Feng
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Wenchao Zhang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Lin Qi
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Binfeng Liu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Hua Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Chenbei Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Zhihong Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha 410011, China
- Shenzhen Research Institute of Central South University, Guangdong 518063, China
- FuRong Laboratory, Changsha 410078, Hunan China
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha 410011, China
- Shenzhen Research Institute of Central South University, Guangdong 518063, China
- Hunan Engineering Research Center of Artificial Intelligence-based Medical Equipment, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| |
Collapse
|
5
|
Yuan T, Liu W, Wang T, Ye F, Zhang J, Gu Z, Xu J, Li Y. Natural Polyphenol Delivered Methylprednisolone Achieve Targeted Enrichment for Acute Spinal Cord Injury Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2404815. [PMID: 39105462 DOI: 10.1002/smll.202404815] [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: 06/12/2024] [Revised: 07/05/2024] [Indexed: 08/07/2024]
Abstract
The strong anti-inflammatory effect of methylprednisolone (MP) is a necessary treatment for various severe cases including acute spinal cord injury (SCI). However, concerns have been raised regarding adverse effects from MP, which also severely limits its clinical application. Natural polyphenols, due to their rich phenolic hydroxyl chemical properties, can form dynamic structures without additional modification, achieving targeted enrichment and drug release at the disease lesion, making them a highly promising carrier. Considering the clinical application challenges of MP, a natural polyphenolic platform is employed for targeted and efficient delivery of MP, reducing its systemic side effects. Both in vitro and SCI models demonstrated polyphenols have multiple advantages as carriers for delivering MP: (1) Achieved maximum enrichment at the injured site in 2 h post-administration, which met the desires of early treatment for diseases; (2) Traceless release of MP; (3) Reducing its side effects; (4) Endowed treatment system with new antioxidative properties, which is also an aspect that needs to be addressed for diseases treatment. This study highlighted a promising prospect of the robust delivery system based on natural polyphenols can successfully overcome the barrier of MP treatment, providing the possibility for its widespread clinical application.
Collapse
Affiliation(s)
- Taoyang Yuan
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Weijie Liu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Tianyou Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Feng Ye
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jianhua Zhang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Zhipeng Gu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Jianguo Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yiwen Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| |
Collapse
|
6
|
Fan Y, Ou Y, Xiao T, Wei Z, Zhu L, Zhu C, Ma Y, Qu S, Zhou W. Coordination-Driven Nanomedicine Mitigates One-Lung Ventilation-Induced Lung Injury via Radicals Scavenging and Cell Pyroptosis Inhibition. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401056. [PMID: 39115137 DOI: 10.1002/smll.202401056] [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: 02/08/2024] [Revised: 07/21/2024] [Indexed: 11/21/2024]
Abstract
One-lung ventilation (OLV) during thoracic surgery often leads to post-operative complications, yet effective pharmacological interventions are lacking. This study reports a baicalin-based metal-coordination nanomedicine with disulfiram (DSF) co-loading to address one-lung ventilation-induced lung injury and reperfusion injury (OLV-LIRI). Baicalin, known for its robust antioxidant properties, suffers from poor water solubility and stability. Leveraging nanotechnology, baicalin's coordination is systematically explored with seven common metal ions, designing iron/copper-mediated binary coordination nanoparticles to overcome these limitations. The self-assembled nanoparticles, primarily formed through metal coordination and π-π stacking forces, encapsulated DSF, ensuring high colloidal stability in diverse physiological matrices. Upon a single-dose administration via endotracheal intubation, the nanoparticles efficiently accumulate in lung tissues and swiftly penetrate the pulmonary mucosa. Intracellularly, baicalin exhibits free radical scavenging activity to suppress inflammation. Concurrently, the release of Cu2+ and DSF enables the in situ generation of CuET, a potent inhibitor of cell pyroptosis. Harnessing these multifaceted mechanisms, the nanoparticles alleviate lung injury symptoms without notable toxic side effects, suggesting a promising preventive strategy for OLV-LIRI.
Collapse
Affiliation(s)
- Yujie Fan
- Department of Anesthesiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), Changsha, Hunan, 410007, China
| | - Yangqin Ou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Ting Xiao
- Department of Anesthesiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), Changsha, Hunan, 410007, China
| | - Ziye Wei
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - LiLing Zhu
- Department of Anesthesiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), Changsha, Hunan, 410007, China
| | - Chenghao Zhu
- Department of Anesthesiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), Changsha, Hunan, 410007, China
| | - Yiran Ma
- Hunan Prize Life Science Research Institute Co., LTD. No. 101 WenYi Road, Changsha, Hunan, 410008, China
| | - Shuangquan Qu
- Department of Anesthesiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital), Changsha, Hunan, 410007, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
- Key Laboratory of Biological Nanotechnology, NHC. No. 87 XiangYa Road, Changsha, Hunan, 410008, China
| |
Collapse
|
7
|
Chi X, Chen T, Luo F, Zhao R, Li Y, Hu S, Li Y, Jiang W, Chen L, Wu D, Du Y, Hu J. Targeted no-releasing L-arginine-induced hesperetin self-assembled nanoparticles for ulcerative colitis intervention. Acta Biomater 2024:S1742-7061(24)00628-7. [PMID: 39461688 DOI: 10.1016/j.actbio.2024.10.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/07/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024]
Abstract
Overproduction of reactive oxygen species (ROS) plays a crucial role in initiating and advancing ulcerative colitis (UC), and the persistent cycle between ROS and inflammation accelerates disease development. Therefore, developing strategies that can effectively scavenge ROS and provide targeted intervention are crucial for the management of UC. In this study, we synthesized natural carrier-free nanoparticles (HST-Arg NPs) using the Mannich reaction and π-π stacking for the intervention of UC. HST-Arg NPs are an oral formulation that exhibit good antioxidant capabilities and gastrointestinal stability. Benefiting from the negatively charged characteristics, HST-Arg NPs can specifically accumulate in positively charged inflamed regions of the colon. Furthermore, in the oxidative microenvironment of colonic inflammation, HST-Arg NPs respond to ROS by releasing nitric oxide (NO). In mice model of UC induced by dextran sulfate sodium (DSS), HST-Arg NPs significantly mitigated colonic injury by modulating oxidative stress, lowering pro-inflammatory cytokines, and repairing intestinal barrier integrity. In summary, this convenient and targeted oral nanoparticle can effectively scavenge ROS at the site of inflammation and achieve gas intervention, offering robust theoretical support for the development of subsequent oral formulations in related inflammatory interventions. STATEMENT OF SIGNIFICANCE: Nanotechnology has been extensively explored in the biomedical field, but the application of natural carrier-free nanotechnology in this area remains relatively rare. In this study, we developed a natural nanoparticle system based on hesperetin (HST), L-arginine (L-Arg), and vanillin (VA) to scavenge ROS and alleviate inflammation. In the context of ulcerative colitis (UC), the synthesized nanoparticles exhibited excellent intervention effects, effectively protecting the colon from damage. Consequently, these nanoparticles provide a promising and precise nutritional intervention strategy by addressing both oxidative stress and inflammatory pathways simultaneously, demonstrating significant potential for application.
Collapse
Affiliation(s)
- Xuesong Chi
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Tao Chen
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Fengxian Luo
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Runan Zhao
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China; College of Biosystems Engineering and Food Science, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China
| | - Yangjing Li
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Shumeng Hu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China; College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China
| | - Yanfei Li
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Wen Jiang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - LiHang Chen
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Di Wu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Yinan Du
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Jiangning Hu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China.
| |
Collapse
|
8
|
Wu J, Wang H, Gao P, Ouyang S. Pyroptosis: Induction and inhibition strategies for immunotherapy of diseases. Acta Pharm Sin B 2024; 14:4195-4227. [PMID: 39525577 PMCID: PMC11544194 DOI: 10.1016/j.apsb.2024.06.026] [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: 03/13/2024] [Revised: 05/15/2024] [Accepted: 06/20/2024] [Indexed: 11/16/2024] Open
Abstract
Cell death is a central process for organismal health. Pyroptosis, namely pyroptotic cell death, is recognized as a critical type that disrupts membrane and triggers pro-inflammatory cytokine secretion via gasdermins, providing a robust form of cytolysis. Meanwhile, along with the thorough research, a great deal of evidence has demonstrated the dual effects of pyroptosis in host defense and inflammatory diseases. More importantly, the recent identification of abundant gasdermin-like proteins in bacteria and fungi suggests an ancient origin of pyroptosis-based regulated cell death in the life evolution. In this review, we bring a general overview of pyroptosis pathways focusing on gasdermin structural biology, regulatory mechanisms, and recent progress in induction and inhibition strategies for disease treatment. We look forward to providing an insightful perspective for readers to comprehend the frame and challenges of the pyroptosis field, and to accelerating its clinical application.
Collapse
Affiliation(s)
- Junjun Wu
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, the Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Hong Wang
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, the Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Pu Gao
- CAS Key Laboratory of Infection and Immunity, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Songying Ouyang
- Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, the Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| |
Collapse
|
9
|
Zhang X, Wu Y, Gong H, Xiong Y, Chen Y, Li L, Zhi B, Lv S, Peng T, Zhang H. A Multifunctional Herb-Derived Glycopeptide Hydrogel for Chronic Wound Healing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400516. [PMID: 38686688 DOI: 10.1002/smll.202400516] [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: 01/22/2024] [Revised: 04/12/2024] [Indexed: 05/02/2024]
Abstract
Chronic wounds constitute an increasingly prevalent global healthcare issue, characterized by recurring bacterial infections, pronounced oxidative stress, compromised functionality of immune cells, unrelenting inflammatory reactions, and deficits in angiogenesis. In response to these multifaceted challenges, the study introduced a stimulus-responsive glycopeptide hydrogel constructed by oxidized Bletilla striata polysaccharide (OBSP), gallic acid-grafted ε-Polylysine (PLY-GA), and paeoniflorin-loaded micelles (MIC@Pae), called OBPG&MP. The hydrogel emulates the structure of glycoprotein fibers of the extracellular matrix (ECM), exhibiting exceptional injectability, self-healing, and biocompatibility. It adapts responsively to the inflammatory microenvironment of chronic wounds, sequentially releasing therapeutic agents to eradicate bacterial infection, neutralize reactive oxygen species (ROS), modulate macrophage polarization, suppress inflammation, and encourage vascular regeneration and ECM remodeling, playing a critical role across the inflammatory, proliferative, and remodeling phases of wound healing. Both in vitro and in vivo studies confirmed the efficacy of OBPG&MP hydrogel in regulating the wound microenvironment and enhancing the regeneration and remodeling of chronic wound skin tissue. This research supports the vast potential for herb-derived multifunctional hydrogels in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Xinyi Zhang
- Department of Radiology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan, 610081, China
| | - Ye Wu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Heng Gong
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yan Xiong
- Department of Radiology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan, 610081, China
| | - Yu Chen
- Department of Radiology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan, 610081, China
| | - Lin Li
- Department of Radiology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan, 610081, China
| | - Biao Zhi
- Department of Radiology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan, 610081, China
| | - Saiqun Lv
- Department of Radiology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan, 610081, China
| | - Tao Peng
- Department of Radiology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan, 610081, China
| | - Hui Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- Med-X Center for Manufacturing, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
10
|
Pan Z, Xu K, Huang G, Hu H, Yang H, Shen H, Qiu K, Wang C, Xu T, Yu X, Fang J, Wang J, Lin Y, Dai J, Zhong Y, Song H, Zhu S, Wang S, Zhou Z, Sun C, Tang Z, Liao S, Yang G, You Z, Dai X, Mao Z. Pyroptotic-Spatiotemporally Selective Delivery of siRNA against Pyroptosis and Autoimmune Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407115. [PMID: 39081086 DOI: 10.1002/adma.202407115] [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: 05/19/2024] [Revised: 07/09/2024] [Indexed: 10/04/2024]
Abstract
Small-interfering RNAs (siRNAs) offer promising prospects for treating pyroptosis-related autoimmune diseases. However, poor stability and off-target effects during in vivo transportation hinder their practical clinical applications. Precision delivery and adaptive release of siRNAs into inflamed tissues and immune cells could unleash their full therapeutic potential. This study establishes a pyroptotic-spatiotemporally selective siRNA delivery system (PMRC@siGSDME) that selectively targets inflammatory tissues, responds to pyroptosis, and exhibits remarkable therapeutic efficacy against various autoimmune diseases. Novel hybrid nanovesicles (NVs) are designed as a combination of pyroptotic macrophage membranes (PMs) and R8-cardiolipin-containing nanovesicles (RC-NVs). Evidence provides that PM-derived proteins involved in cell-cell interactions and membrane trafficking may contribute to the specificity of NVs to inflammatory tissue. In addition, cardiolipin anchored in the hybrid NVs increases its affinity for activated gasdermin E (GSDME) and achieves pyroptosis-adaptive release of siGSDME for the spatiotemporally selective suppression of immune responses. More importantly, PMRC@siGSDME displays significant anti-inflammatory and therapeutic effects in multiple mouse autoimmune disease models, including arthritis and inflammatory bowel disease (IBD). Collectively, an innovative siRNA delivery strategy precisely tailored for pyroptotic cells has been developed, paving the way for new treatments for autoimmune inflammatory diseases with minimal side effects and wide clinical applicability.
Collapse
Affiliation(s)
- Zongyou Pan
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Kaiwang Xu
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Guanrui Huang
- Department of Orthopedic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Haoran Hu
- Department of Pathology, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong, 999077, China
| | - Huang Yang
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Haotian Shen
- Department of Orthopedic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Kaijie Qiu
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Canlong Wang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Tengjing Xu
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Xinning Yu
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Jinhua Fang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Jiajie Wang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Yunting Lin
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Jiacheng Dai
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Yuting Zhong
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Hongyun Song
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Sunan Zhu
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Siheng Wang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Zhuxing Zhou
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Chuyue Sun
- Department of Otorhinolaryngology, Head and Neck Surgery, Taihe Hospital of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Fuyang, 236000, China
| | - Zhaopeng Tang
- Department of Orthopedic Surgery, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, 730050, China
| | - Shiyao Liao
- Center for Plastic & Reconstructive Surgery, Department of Orthopedics, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, 310014, China
| | - Guang Yang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Zhiyuan You
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Xuesong Dai
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310009, China
| | - Zhengwei Mao
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
11
|
Zhao L, Cheng H, Tong Z, Cai J. Nanoparticle-mediated cell pyroptosis: a new therapeutic strategy for inflammatory diseases and cancer. J Nanobiotechnology 2024; 22:504. [PMID: 39175020 PMCID: PMC11340130 DOI: 10.1186/s12951-024-02763-3] [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: 06/26/2024] [Accepted: 08/12/2024] [Indexed: 08/24/2024] Open
Abstract
Pyroptosis, a lytic form of cell death mediated by the gasdermin family, is characterized by cell swelling and membrane rupture. Inducing pyroptosis in cancer cells can enhance antitumor immune responses and is a promising strategy for cancer therapy. However, excessive pyroptosis may trigger the development of inflammatory diseases due to immoderate and continuous inflammatory reactions. Nanomaterials and nanobiotechnology, renowned for their unique advantages and diverse structures, have garnered increasing attention owing to their potential to induce pyroptosis in diseases such as cancer. A nano-delivery system for drug-induced pyroptosis in cancer cells can overcome the limitations of small molecules. Furthermore, nanomedicines can directly induce and manipulate pyroptosis. This review summarizes and discusses the latest advancements in nanoparticle-based treatments with pyroptosis among inflammatory diseases and cancer, focusing on their functions and mechanisms and providing valuable insights into selecting nanodrugs for pyroptosis. However, the clinical application of these strategies still faces challenges owing to a limited understanding of nanobiological interactions. Finally, future perspectives on the emerging field of pyroptotic nanomaterials are presented.
Collapse
Affiliation(s)
- Lin Zhao
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China
| | - Haipeng Cheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China
| | - Zhongyi Tong
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China
| | - Jing Cai
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, 410011, China.
| |
Collapse
|
12
|
Hammad AM, Alzaghari LF, Alfaraj M, Lux V, Sunoqrot S. Green Tea Polyphenol Nanoparticles Reduce Anxiety Caused by Tobacco Smoking Withdrawal in Rats by Suppressing Neuroinflammation. TOXICS 2024; 12:598. [PMID: 39195700 PMCID: PMC11360476 DOI: 10.3390/toxics12080598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024]
Abstract
Repeated exposure to tobacco smoke causes neuroinflammation and neuroplasticity, which correlates with smoking withdrawal-induced anxiety. The purpose of this study was to investigate the anticipated involvement of antioxidant-rich nanoparticles (NPs) prepared by oxidation-triggered polymerization of green tea catechins in impacting these effects in a rat model of tobacco smoke exposure. Exposure to tobacco smoke was carried out for 2 h a day, 5 days a week, for a total of 36 days. Weekly behavioral tests were conducted prior to recommencing the exposure. Following a 20-day exposure period, rats were administered either distilled water or green tea (GT) NPs (20 mg/kg, orally) for an additional 16 days. Our findings revealed that tobacco smoke exposure induced anxiety-like behavior indicative of withdrawal, and this effect was alleviated by GT NPs. Tobacco smoke exposure caused a marked increase in the relative mRNA and protein expression of nuclear factor-kappa B (NF-κB) and reduced the relative mRNA and protein expression of brain-derived neurotrophic factor (BDNF) in the hippocampus (HIP) and hypothalamus (HYP) brain subregions. The intervention of GT NPs effectively inhibited these effects. Our findings demonstrate the potent protective role of GT NPs in reducing withdrawal-induced anxiety-like behavior, neuroinflammation, and neuroplasticity triggered by tobacco smoke exposure.
Collapse
Affiliation(s)
- Alaa M. Hammad
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Lujain F. Alzaghari
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Malek Alfaraj
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Vanessa Lux
- Department of Genetic Psychology, Ruhr University Bochum, 44801 Bochum, Germany;
| | - Suhair Sunoqrot
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| |
Collapse
|
13
|
Liu D, Sun S, Qiao H, Xin Q, Zhou S, Li L, Song N, Zhang L, Chen Q, Tian F, Mu X, Zhang S, Zhang J, Guo M, Wang H, Zhang XD, Zhang R. Ce 12V 6 Clusters with Multi-Enzymatic Activities for Sepsis Treatment. Adv Healthc Mater 2024:e2401581. [PMID: 39129228 DOI: 10.1002/adhm.202401581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/22/2024] [Indexed: 08/13/2024]
Abstract
Artificial enzymes, especially nanozymes, have attracted wide attention due to their controlled catalytic activity, selectivity, and stability. The rising Cerium-based nanozymes exhibit unique SOD-like activity, and Vanadium-based nanozymes always hold excellent GPx-like activity. However, most inflammatory diseases involve polymerase biocatalytic processes that require multi-enzyme activities. The nanocomposite can fulfill multi-enzymatic activity simultaneously, but large nanoparticles (>10 nm) cannot be excreted rapidly, leading to biosafety challenges. Herein, atomically precise Ce12V6 clusters with a size of 2.19 nm are constructed. The Ce12V6 clusters show excellent glutathione peroxidase (GPx) -like activity with a significantly lower Michaelis-Menten constant (Km, 0.0125 mM versus 0.03 mM of natural counterpart) and good activities mimic superoxide dismutase (SOD) and peroxidase (POD). The Ce12V6 clusters exhibit the ability to scavenge the ROS including O2 ·- and H2O2 via the cascade reactions of multi-enzymatic activities. Further, the Ce12V6 clusters modulate the proinflammatory cytokines including tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β) and consequently rescue the multi-organ failure in the lipopolysaccharide (LPS)-induced sepsis mouse model. With excellent biocompatibility, the Ce12V6 clusters show promise in the treatment of sepsis.
Collapse
Affiliation(s)
- Di Liu
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Si Sun
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Huanhuan Qiao
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Qi Xin
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin Third Central Hospital, Tianjin, 300170, China
| | - Sufei Zhou
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Lingxia Li
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Nan Song
- Department of Physics, School of Science, Tianjin Chengjian University, Tianjin, 300384, 18, China
| | - Lijie Zhang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Qi Chen
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Fangzhen Tian
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Xiaoyu Mu
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Shaofang Zhang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Jing Zhang
- Department of Cardiology Tianjin Chest Hospital, Tianjin University, Tianjin, 300222, China
| | - Meili Guo
- Department of Physics, School of Science, Tianjin Chengjian University, Tianjin, 300384, 18, China
| | - Hao Wang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
| | - Xiao-Dong Zhang
- Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, 300072, China
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin, 300350, China
| | - Ruiping Zhang
- The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
14
|
Wu Z, Zhang P, Yue J, Wang Q, Zhuang P, Jehan S, Fan L, Xue J, Zhou W, Wang H. Tea polyphenol nanoparticles enable targeted siRNA delivery and multi-bioactive therapy for abdominal aortic aneurysms. J Nanobiotechnology 2024; 22:471. [PMID: 39118143 PMCID: PMC11308685 DOI: 10.1186/s12951-024-02756-2] [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: 03/31/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening vascular disease, while there is a lack of pharmaceutical interventions to halt AAA progression presently. To address the multifaceted pathology of AAA, this work develops a novel multifunctional gene delivery system to simultaneously deliver two siRNAs targeting MMP-2 and MMP-9. The system (TPNs-siRNA), formed through the oxidative polymerization and self-assembly of epigallocatechin gallate (EGCG), efficiently encapsulates siRNAs during self-assembly. TPNs-siRNA safeguards siRNAs from biological degradation, facilitates intracellular siRNA transfection, promotes lysosomal escape, and releases siRNAs to silence MMP-2 and MMP-9. Additionally, TPNs, serving as a multi-bioactive material, mitigates oxidative stress and inflammation, fosters M1-to-M2 repolarization of macrophages, and inhibits cell calcification and apoptosis. In experiments with AAA mice, TPNs-siRNA accumulated and persisted in aneurysmal tissue after intravenous delivery, demonstrating that TPNs-siRNA can be significantly distributed in macrophages and VSMCs relevant to AAA pathogenesis. Leveraging the carrier's intrinsic multi-bioactive properties, the targeted siRNA delivery by TPNs exhibits a synergistic effect for enhanced AAA therapy. Furthermore, TPNs-siRNA is gradually metabolized and excreted from the body, resulting in excellent biocompatibility. Consequently, TPNs emerges as a promising multi-bioactive nanotherapy and a targeted delivery nanocarrier for effective AAA therapy.
Collapse
Affiliation(s)
- Zhen Wu
- Department of Vascular and Interventional Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Peng Zhang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Jie Yue
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Qingshan Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Peipei Zhuang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Shah Jehan
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Liyuan Fan
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Jiarun Xue
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Haiyang Wang
- Department of Vascular and Interventional Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
15
|
Cai K, Jiang H, Zou Y, Song C, Cao K, Chen S, Wu Y, Zhang Z, Geng D, Zhang N, Liu B, Sun G, Tang M, Li Z, Zhang Y, Sun Y, Zhang Y. Programmed death of cardiomyocytes in cardiovascular disease and new therapeutic approaches. Pharmacol Res 2024; 206:107281. [PMID: 38942341 DOI: 10.1016/j.phrs.2024.107281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024]
Abstract
Cardiovascular diseases (CVDs) have a complex pathogenesis and pose a major threat to human health. Cardiomyocytes have a low regenerative capacity, and their death is a key factor in the morbidity and mortality of many CVDs. Cardiomyocyte death can be regulated by specific signaling pathways known as programmed cell death (PCD), including apoptosis, necroptosis, autophagy, pyroptosis, and ferroptosis, etc. Abnormalities in PCD can lead to the development of a variety of cardiovascular diseases, and there are also molecular-level interconnections between different PCD pathways under the same cardiovascular disease model. Currently, the link between programmed cell death in cardiomyocytes and cardiovascular disease is not fully understood. This review describes the molecular mechanisms of programmed death and the impact of cardiomyocyte death on cardiovascular disease development. Emphasis is placed on a summary of drugs and potential therapeutic approaches that can be used to treat cardiovascular disease by targeting and blocking programmed cell death in cardiomyocytes.
Collapse
Affiliation(s)
- Kexin Cai
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Haoyue Jiang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Yuanming Zou
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Chunyu Song
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Kexin Cao
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Shuxian Chen
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Yanjiao Wu
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Zhaobo Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Danxi Geng
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China
| | - Bo Liu
- The first hospital of China Medical University, Department of cardiac surgery, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Guozhe Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Man Tang
- Department of clinical pharmacology, College of Pharmacy, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Zhao Li
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Yixiao Zhang
- Department of Urology Surgery, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China; Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| |
Collapse
|
16
|
Wang F, Fan Y, Liu Y, Lou X, Sutrisno L, Peng S, Li J. Oxygen-carrying semiconducting polymer nanoprodrugs induce sono-pyroptosis for deep-tissue tumor treatment. EXPLORATION (BEIJING, CHINA) 2024; 4:20230100. [PMID: 39175882 PMCID: PMC11335461 DOI: 10.1002/exp.20230100] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/20/2023] [Indexed: 08/24/2024]
Abstract
Sonodynamic therapy (SDT) has been explored for cancer therapy, especially for deep tumors due to its low tissue penetration restriction. The therapeutic efficacy of SDT is limited due to the complicated tumor microenvironment. This study reports the construction of oxygen-carrying semiconducting polymer nanoprodrugs (OSPNpro) for deep tumor treatment via combining amplified SDT with pyroptosis. An oxygen carrier perfluorohexane, sonodynamic semiconducting polymer as the sonosensitizer, and reactive oxygen species (ROS)-responsive prodrug are co-loaded into a nanoparticle system, leading to the formation of these polymer nanoprodrugs. Such OSPNpro show an effective accumulation in tumor tissues after systemic administration, in which they deliver oxygen to relieve tumor hypoxia microenvironment and thus mediate amplified SDT via producing ROS under ultrasound (US) irradiation, even when the tumors are covered with a 2-cm chicken breast tissue. In addition, the ROS-responsive prodrugs are activated by the generated ROS to trigger pyroptosis of tumor cells. Such a sono-pyroptosis induces a strong antitumor immunity with obviously higher level infiltrations of effector immune cells into tumors. Therefore, OSPNpro-based combinational therapy can greatly inhibit the growth of 2-cm chicken breast tissue-covered deep tumors and suppress tumor metastasis. This study offers a prodrug nanoplatform for treatment of deep tumor via sono-pyroptosis strategy.
Collapse
Affiliation(s)
- Fengshuo Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Biological Science and Medical EngineeringDonghua UniversityShanghaiChina
| | - Yongliang Fan
- Department of Cardiovascular SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yue Liu
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Biological Science and Medical EngineeringDonghua UniversityShanghaiChina
| | - Xiangxin Lou
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Biological Science and Medical EngineeringDonghua UniversityShanghaiChina
| | - Linawati Sutrisno
- World Premier International (WPI) Research Center for Materials Nanoarchitectonics (MANA)National Institute for Materials Science (NIMS)TsukubaJapan
| | - Shaojun Peng
- Zhuhai Institute of Translational MedicineZhuhai Precision Medical CenterZhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University)ZhuhaiGuangdongChina
| | - Jingchao Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Biological Science and Medical EngineeringDonghua UniversityShanghaiChina
| |
Collapse
|
17
|
Yang Q, Sun X, Ding Q, Qi M, Liu C, Li T, Shi F, Wang L, Li C, Kim JS. An ATP-responsive metal-organic framework against periodontitis via synergistic ion-interference-mediated pyroptosis. Natl Sci Rev 2024; 11:nwae225. [PMID: 39071842 PMCID: PMC11275458 DOI: 10.1093/nsr/nwae225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/08/2024] [Accepted: 06/11/2024] [Indexed: 07/30/2024] Open
Abstract
Periodontitis involves hyperactivated stromal cells that recruit immune cells, exacerbating inflammation. This study presents an ATP-responsive metal-organic framework (Mg/Zn-MOF) designed for periodontitis treatment, utilizing ion interference to modulate immune responses and prevent tissue destruction. Addressing the challenges of synergistic ion effects and targeted delivery faced by traditional immunomodulatory nanomaterials, the Mg/Zn-MOF system is activated by extracellular ATP-a pivotal molecule in periodontitis pathology-ensuring targeted ion release. Magnesium and zinc ions released from the framework synergistically inhibit membrane pore formation by attenuating Gasdermin D (GSDMD) expression and activation. This action curtails pyroptosis, lactate dehydrogenase and IL-1β release, thwarting the onset of inflammatory cascades. Mechanistically, Mg/Zn-MOF intervenes in both the NLRP3/Caspase-1/GSDMD and Caspase-11/GSDMD pathways to mitigate pyroptosis. In vivo assessments confirm its effectiveness in diminishing inflammatory cell infiltration and preserving collagen integrity, thereby safeguarding against periodontal tissue damage and bone loss. This investigation highlights the promise of ion-interference strategies in periodontitis immunotherapy, representing a significant stride in developing targeted therapeutic approaches.
Collapse
Affiliation(s)
- Qijing Yang
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Xiaolin Sun
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Qihang Ding
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
- Department of Chemistry, Korea University, Seoul 02841, South Korea
| | - Manlin Qi
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Chengyu Liu
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Tingxuan Li
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Fangyu Shi
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Lin Wang
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Chunyan Li
- Department of Prosthodontics, Jilin Provincial Engineering Laboratory of Intelligent Oral Treatment Technology, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, South Korea
| |
Collapse
|
18
|
Ha W, Ma R, Kang JY, Iradukunda Y, Shi YP. Green and shape-tunable synthesis of ellagic acid crystalline particles by tannic acid for neuroprotection against oxidative stress. Biomater Sci 2024; 12:3610-3621. [PMID: 38842122 DOI: 10.1039/d4bm00380b] [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: 06/07/2024]
Abstract
Oxidative stress (OS) plays an important role in the emergence and prevention of neurodegenerative diseases, such as Alzheimer's disease (AD). Excess reactive oxygen species (ROS) accumulated in a neuronal cell can lead to OS, producing cell injury and death. Seeking nanoantioxidants against AD-related oxidative stress has attracted a lot of attention, especially those potential antioxidant agents derived from natural polyphenols. However, the transformation of abundant plant polyphenols to antioxidative biomaterials against OS is still challenging. In this work, we report a new method to transform amorphous tannic acid (TA) into tailorable shaped ellagic acid (EA) crystalline particles without using an organic solvent. EA crystalline particles were generated from TA, which underwent a chemical transformation, in situ metal phenolic coordination and acid-induced assembly process, and the size and shape could be controlled by varying the amount of acid. As-prepared EA crystalline particles showed excellent stability in water and lysosomal mimicking fluid and possess unique fluorescence properties and a strong response in mass spectrometry, which is beneficial for their imaging analysis in cells and tissues. More importantly, EA particles have shown significant H2O2-related ROS scavenging ability, a high cellular uptake capacity, an excellent neuroprotective effect in PC12 cells, a high drug loading capacity and BBB permeability to enter the brain. Our study suggested that the EA crystalline particles show great potential for OS-mediated AD treatment.
Collapse
Affiliation(s)
- Wei Ha
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicines of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, P. R. China.
| | - Rui Ma
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicines of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, P. R. China.
| | - Jing-Yan Kang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicines of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, P. R. China.
| | - Yves Iradukunda
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicines of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, P. R. China.
| | - Yan-Ping Shi
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicines of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Lanzhou 730000, P. R. China.
| |
Collapse
|
19
|
Zhou H, He J, Liu R, Cheng J, Yuan Y, Mao W, Zhou J, He H, Liu Q, Tan W, Shuai C, Deng Y. Microenvironment-responsive metal-phenolic network release platform with ROS scavenging, anti-pyroptosis, and ECM regeneration for intervertebral disc degeneration. Bioact Mater 2024; 37:51-71. [PMID: 38515609 PMCID: PMC10954684 DOI: 10.1016/j.bioactmat.2024.02.036] [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: 12/14/2023] [Revised: 02/13/2024] [Accepted: 02/29/2024] [Indexed: 03/23/2024] Open
Abstract
Intervertebral disc degeneration (IVDD) can be caused by aging, injury, and genetic factors. The pathological changes associated with IVDD include the excessive accumulation of reactive oxygen species (ROS), cellular pyroptosis, and extracellular matrix (ECM) degradation. There are currently no approved specific molecular therapies for IVDD. In this study, we developed a multifunctional and microenvironment-responsive metal-phenolic network release platform, termed TMP@Alg-PBA/PVA, which could treat (IL-1β)-induced IVDD. The metal-phenolic network (TA-Mn-PVP, TMP) released from this platform targeted mitochondria to efficiently scavenge ROS and reduce ECM degradation. Pyroptosis was suppressed through the inhibition of the IL-17/ERK signaling pathway. These findings demonstrate the versatility of the platform. And in a rat model of IVDD, TMP@Alg-PBA/PVA exhibited excellent therapeutic effects by reducing the progression of the disease. TMP@Alg-PBA/PVA, therefore, presents clinical potential for the treatment of IVDD.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
- Department of Joint Surgery and Sports Medicine, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Jinpeng He
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Renfeng Liu
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Jun Cheng
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Yuhao Yuan
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Wanpu Mao
- Department of Joint Surgery and Sports Medicine, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Jun Zhou
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Honghui He
- Department of Joint Surgery and Sports Medicine, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421002, China
| | - Qianqi Liu
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Wei Tan
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Cijun Shuai
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Youwen Deng
- Department of Spine Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| |
Collapse
|
20
|
Fan X, Han J, Zhong L, Zheng W, Shao R, Zhang Y, Shi S, Lin S, Huang Z, Huang W, Cai X, Ye B. Macrophage-Derived GSDMD Plays an Essential Role in Atherosclerosis and Cross Talk Between Macrophages via the Mitochondria-STING-IRF3/NF-κB Axis. Arterioscler Thromb Vasc Biol 2024; 44:1365-1378. [PMID: 38695170 DOI: 10.1161/atvbaha.123.320612] [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: 12/19/2023] [Accepted: 04/16/2024] [Indexed: 05/24/2024]
Abstract
BACKGROUND Macrophages play a crucial role in atherosclerotic plaque formation, and the death of macrophages is a vital factor in determining the fate of atherosclerosis. GSDMD (gasdermin D)-mediated pyroptosis is a programmed cell death, characterized by membrane pore formation and inflammatory factor release. METHODS ApoE-/- and Gsdmd-/- ApoE-/- mice, bone marrow transplantation, and AAV (adeno-associated virus serotype 9)-F4/80-shGSDMD (shRNA-GSDMD) were used to examine the effect of macrophage-derived GSDMD on atherosclerosis. Single-cell RNA sequencing was used to investigate the changing profile of different cellular components and the cellular localization of GSDMD during atherosclerosis. RESULTS First, we found that GSDMD is activated in human and mouse atherosclerotic plaques and Gsdmd-/- attenuates the atherosclerotic lesion area in high-fat diet-fed ApoE-/- mice. We performed single-cell RNA sequencing of ApoE-/- and Gsdmd-/- ApoE-/- mouse aortas and showed that GSDMD is principally expressed in atherosclerotic macrophages. Using bone marrow transplantation and AAV-F4/80-shGSDMD, we identified the potential role of macrophage-derived GSDMD in aortic pyroptosis and atherosclerotic injuries in vivo. Mechanistically, GSDMD contributes to mitochondrial perforation and mitochondrial DNA leakage and subsequently activates the STING (stimulator of interferon gene)-IRF3 (interferon regulatory factor 3)/NF-κB (nuclear factor kappa B) axis. Meanwhile, GSDMD regulates the STING pathway activation and macrophage migration via cytokine secretion. Inhibition of GSDMD with GSDMD-specific inhibitor GI-Y1 (GSDMD inhibitor Y1) can effectively alleviate the progression of atherosclerosis. CONCLUSIONS Our study has provided a novel macrophage-derived GSDMD mechanism in the promotion of atherosclerosis and demonstrated that GSDMD can be a potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Xiaoxi Fan
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Jibo Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Zhejiang, China (J.H.)
| | - Lingfeng Zhong
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Wenyuan Zheng
- Key Laboratory of Precision Medicine for Atherosclerosis Disease of Zhejiang Province, Department of Cardiology, Affiliated First Hospital of Ningbo University, Zhejiang, China (W.Z., B.Y.)
| | - Ruiyin Shao
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Yucong Zhang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Si Shi
- First School of Medicine, Wenzhou Medical University, Zhejiang, China (S.S.)
| | - Shuang Lin
- Department of Cardiology, Ningbo Medical Center Li Huili Hospital, Zhejiang, China (S.L.)
| | - Zhouqing Huang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Weijian Huang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Xueli Cai
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
| | - Bozhi Ye
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Zhejiang, China (X.F., L.Z., R.S., Y.Z., Z.H., W.H., X.C., B.Y.)
- Key Laboratory of Precision Medicine for Atherosclerosis Disease of Zhejiang Province, Department of Cardiology, Affiliated First Hospital of Ningbo University, Zhejiang, China (W.Z., B.Y.)
| |
Collapse
|
21
|
Wang M, Liao J, Lin W, Jiang L, Peng K, Su X, Li H, Wang H, Wang Y. YL-109 attenuates sepsis-associated multiple organ injury through inhibiting the ERK/AP-1 axis and pyroptosis by upregulating CHIP. Biomed Pharmacother 2024; 175:116633. [PMID: 38670049 DOI: 10.1016/j.biopha.2024.116633] [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: 02/07/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Sepsis is a severe inflammatory disorder that can lead to life-threatening multiple organ injury. Lipopolysaccharide (LPS)-induced inflammation is the leading cause of multiple organ failure in sepsis. This study aimed to explore the effect of a novel agent, 2-(4-hydroxy-3-methoxyphenyl)-benzothiazole (YL-109), on LPS-induced multiple organ injury and the molecular mechanisms underlying these processes. The results showed that YL-109 protected against LPS-induced high mortality, cardiac dysfunction, pulmonary and intestinal injury through inhibiting the proinflammatory response, NLRP3 expression and pyroptosis-associated indicators in mouse tissues. YL-109 suppressed LPS-initiated cytokine release, pyroptosis and pyroptosis-related protein expression in HL-1, IEC-6 and MLE-12 cells, which was consistent with the results of the in vivo experiments. Mechanistically, YL-109 reduces phosphorylated ERK (extracellular signal-regulated kinase) levels and NF-κB activation, which are achieved through upregulating CHIP (carboxy terminus of Hsc70-interacting protein) expression, thereby inhibiting c-Jun and c-Fos activation as well as NLRP3 expression. As an E3 ligase, CHIP overexpression obviously promoted the degradation of phosphorylated ERK and inhibited the expression of NF-κB-mediated NLRP3 in cells stimulated with LPS. The protective effects of YL-109 against cardiac, pulmonary and intestinal damage, inflammation and pyroptosis caused by LPS were eliminated in CHIP knockout mice. Our results not only reveal the protective effect and molecular mechanism of YL-109 against LPS-mediated organs damage but also provide additional insights into the effect of CHIP on negatively regulating pyroptosis and inflammatory pathways.
Collapse
Affiliation(s)
- Miao Wang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Jia Liao
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Wan Lin
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Lucen Jiang
- Department of Pathology, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Kangli Peng
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xingyu Su
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Hang Li
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Huadong Wang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Yiyang Wang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
22
|
Zhao P, Hu HZ, Chen XT, Jiang QY, Yu XZ, Cen XL, Lin SQ, Mai SQ, Pang WL, Chen JX, Zhang Q. Mild hyperthermia enhanced synergistic uric acid degradation and multiple ROS elimination for an effective acute gout therapy. J Nanobiotechnology 2024; 22:275. [PMID: 38778401 PMCID: PMC11112921 DOI: 10.1186/s12951-024-02539-9] [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: 02/18/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Acute gouty is caused by the excessive accumulation of Monosodium Urate (MSU) crystals within various parts of the body, which leads to a deterioration of the local microenvironment. This degradation is marked by elevated levels of uric acid (UA), increased reactive oxygen species (ROS) production, hypoxic conditions, an upsurge in pro-inflammatory mediators, and mitochondrial dysfunction. RESULTS In this study, we developed a multifunctional nanoparticle of polydopamine-platinum (PDA@Pt) to combat acute gout by leveraging mild hyperthermia to synergistically enhance UA degradation and anti-inflammatory effect. Herein, PDA acts as a foundational template that facilitates the growth of a Pt shell on the surface of its nanospheres, leading to the formation of the PDA@Pt nanomedicine. Within this therapeutic agent, the Pt nanoparticle catalyzes the decomposition of UA and actively breaks down endogenous hydrogen peroxide (H2O2) to produce O2, which helps to alleviate hypoxic conditions. Concurrently, the PDA component possesses exceptional capacity for ROS scavenging. Most significantly, Both PDA and Pt shell exhibit absorption in the Near-Infrared-II (NIR-II) region, which not only endow PDA@Pt with superior photothermal conversion efficiency for effective photothermal therapy (PTT) but also substantially enhances the nanomedicine's capacity for UA degradation, O2 production and ROS scavenging enzymatic activities. This photothermally-enhanced approach effectively facilitates the repair of mitochondrial damage and downregulates the NF-κB signaling pathway to inhibit the expression of pro-inflammatory cytokines. CONCLUSIONS The multifunctional nanomedicine PDA@Pt exhibits exceptional efficacy in UA reduction and anti-inflammatory effects, presenting a promising potential therapeutic strategy for the management of acute gout.
Collapse
Affiliation(s)
- Pei Zhao
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Office of Clinical Trial of Drug, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510663, Guangdong, China
| | - Hua-Zhong Hu
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Office of Clinical Trial of Drug, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510663, Guangdong, China
| | - Xiao-Tong Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Qi-Yun Jiang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Office of Clinical Trial of Drug, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510663, Guangdong, China
| | - Xue-Zhao Yu
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Office of Clinical Trial of Drug, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510663, Guangdong, China
| | - Xiao-Lin Cen
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Office of Clinical Trial of Drug, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510663, Guangdong, China
| | - Shi-Qing Lin
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Office of Clinical Trial of Drug, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510663, Guangdong, China
| | - Sui-Qing Mai
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Office of Clinical Trial of Drug, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510663, Guangdong, China
| | - Wei-Lin Pang
- School of Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jin-Xiang Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Qun Zhang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Office of Clinical Trial of Drug, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510663, Guangdong, China.
| |
Collapse
|
23
|
Hu Q, Li J, Wang T, Xu X, Duan Y, Jin Y. Polyphenolic Nanoparticle-Modified Probiotics for Microenvironment Remodeling and Targeted Therapy of Inflammatory Bowel Disease. ACS NANO 2024; 18:12917-12932. [PMID: 38720520 DOI: 10.1021/acsnano.4c00830] [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: 05/22/2024]
Abstract
Inflammatory bowel diseases (IBDs) refer to multifaceted disorders in the intestinal microenvironment and microbiota homeostasis. In view of the broad bioactivity and high compatibility of polyphenols, there is considerable interest in developing a polyphenol-based collaborative platform to remodel the IBD microenvironment and regulate microbiota. Here, we demonstrated the coordination assembly of nanostructured polyphenols to modify probiotics and simultaneously deliver drugs for IBD treatment. Inspired by the distinctive structure of tannic acid (TA), we fabricated nanostructured pBDT-TA by using a self-polymerizable aromatic dithiol (BDT) and TA, which exhibited excellent antioxidant and anti-inflammatory capability in vitro. We thus coated pBDT-TA and sodium alginate (SA) to the surface of Escherichia coli Nissle 1917 layer by layer to construct the collaborative platform EcN@SA-pBDT-TA. The modified probiotics showed improved resistance to oxidative and inflammatory stress, which resulted in superior colon accumulation and retention in IBD model mice. Further, EcN@SA-pBDT-TA could alleviate dextran sulfate sodium (DSS)-induced colitis by controlling the inflammatory response, repairing intestinal barriers, and modulating gut microbiota. Importantly, EcN@SA-pBDT-TA-mediated IBD drug delivery could achieve an improved therapeutic effect in DSS model mice. Given the availability and functionality of polyphenol and prebiotics, we expected that nanostructured polyphenol-modified probiotics provided a solution to develop a collaborative platform for IBD treatment.
Collapse
Affiliation(s)
- Qinglian Hu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Jingyu Li
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Tong Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Xiangchi Xu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yuxuan Duan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| |
Collapse
|
24
|
Wang H, Tang C, Xiang Y, Zou C, Hu J, Yang G, Zhou W. Tea polyphenol-derived nanomedicine for targeted photothermal thrombolysis and inflammation suppression. J Nanobiotechnology 2024; 22:146. [PMID: 38566213 PMCID: PMC10988797 DOI: 10.1186/s12951-024-02446-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: 11/02/2023] [Accepted: 03/28/2024] [Indexed: 04/04/2024] Open
Abstract
Thrombotic diseases impose a significant global health burden, and conventional drug-based thrombolytic therapies are encumbered by the risk of bleeding complications. In this study, we introduce a novel drug-free nanomedicine founded on tea polyphenols nanoparticles (TPNs), which exhibits multifaceted capabilities for localized photothermal thrombolysis. TPNs were synthesized through a one-pot process under mild conditions, deriving from the monomeric epigallocatechin-3-gallate (EGCG). Within this process, indocyanine green (ICG) was effectively encapsulated, exploiting multiple intermolecular interactions between EGCG and ICG. While both TPNs and ICG inherently possessed photothermal potential, their synergy significantly enhanced photothermal conversion and stability. Furthermore, the nanomedicine was functionalized with cRGD for targeted delivery to activated platelets within thrombus sites, eliciting robust thrombolysis upon laser irradiation across diverse thrombus types. Importantly, the nanomedicine's potent free radical scavenging abilities concurrently mitigated vascular inflammation, thus diminishing the risk of disease recurrence. In summary, this highly biocompatible multifunctional nanomaterial holds promise as a comprehensive approach that combines thrombolysis with anti-inflammatory actions, offering precision in thrombosis treatment.
Collapse
Affiliation(s)
- Hui Wang
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Cui Tang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Yuxia Xiang
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Chan Zou
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Jianming Hu
- First Department of Pathology, Affiliated Hospital, Shihezi University, Shihezi, Xinjiang Uygur Autonomous Region, 832002, China
| | - Guoping Yang
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China.
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China.
- National-Local Joint Engineering Laboratory of Drug Clinical Evaluation Technology, Changsha, Hunan, 410000, China.
- Hunan Engineering Research Center for Optimization of Drug Formulation and Early Clinical Evaluation, Changsha, Hunan, 410013, China.
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, China.
- Academician Workstation, Changsha Medical University, Changsha, 410219, China.
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, Affiliated Hospital, Shihezi University, Shihezi, Xinjiang, 832002, China.
| |
Collapse
|
25
|
Wang Y, Zhao X, Zhou X, Dai J, Hu X, Piao Y, Zu G, Xiao J, Shi K, Liu Y, Li Y, Shi L. A supramolecular hydrogel dressing with antibacterial, immunoregulation, and pro-regeneration ability for biofilm-associated wound healing. J Control Release 2024; 368:740-755. [PMID: 38499092 DOI: 10.1016/j.jconrel.2024.03.024] [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: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024]
Abstract
Chronic wound treatment has emerged as a significant healthcare concern worldwide due to its substantial economic burden and the limited effectiveness of current treatments. Effective management of biofilm infections, regulation of excessive oxidative stress, and promotion of tissue regeneration are crucial for addressing chronic wounds. Hydrogel stands out as a promising candidate for chronic wound treatment. However, its clinical application is hindered by the difficulty in designing and fabricating easily and conveniently. To overcome these obstacles, we present a supermolecular G-quadruplex hydrogel with the desired multifunction via a dynamic covalent strategy and Hoogsteen-type hydrogen bonding. The G-quadruplex hydrogel is made from the self-assembly of guanosine, 2-formylphenyboronic acid, polyethylenimine, and potassium chloride, employing dynamic covalent strategy and Hoogsteen-type hydrogen bonding. In the acidic/oxidative microenvironment associated with bacterial infections, the hydrogel undergoes controlled degradation, releasing the polyethylenimine domain, which effectively eliminates bacteria. Furthermore, nanocomplexes comprising guanosine monophosphate and manganese sulfate are incorporated into the hydrogel skeleton, endowing it with the ability to scavenge reactive oxygen species and modulate macrophages. Additionally, the integration of basic fibroblast growth factor into the G-quadruplex skeleton through dynamic covalent bonds facilitates controlled tissue regeneration. In summary, the facile preparation process and the incorporation of multiple functionalities render the G-quadruplex hydrogel a highly promising candidate for advanced wound dressing. It holds great potential to transition from laboratory research to clinical practice, addressing the pressing needs of chronic wound management.
Collapse
Affiliation(s)
- Yumeng Wang
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xinghong Zhao
- Center for Sustainable Antimicrobials, Department of Pharmacy, Sichuan Agricultural University, Chengdu 611130, China
| | - Xingjian Zhou
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Juqin Dai
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaowen Hu
- Wenzhou Institute, University of Chinese Academy of Sciences, Zhejiang 325001, China
| | - Yinzi Piao
- Wenzhou Institute, University of Chinese Academy of Sciences, Zhejiang 325001, China
| | - Guangyue Zu
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Jian Xiao
- School of Pharmaceutical Sciences, Key Laboratory of Biotechnology and Pharmaceutical Engineering, The Center of Wound Healing and Regenerative Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Keqing Shi
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Yong Liu
- Wenzhou Institute, University of Chinese Academy of Sciences, Zhejiang 325001, China; State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China.
| | - Yuanfeng Li
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, and Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
26
|
Bhat AA, Riadi Y, Afzal M, Bansal P, Kaur H, Deorari M, Ali H, Shahwan M, Almalki WH, Kazmi I, Alzarea SI, Dureja H, Singh SK, Dua K, Gupta G. Exploring ncRNA-mediated pathways in sepsis-induced pyroptosis. Pathol Res Pract 2024; 256:155224. [PMID: 38452584 DOI: 10.1016/j.prp.2024.155224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/08/2024] [Accepted: 02/21/2024] [Indexed: 03/09/2024]
Abstract
Sepsis, a potentially fatal illness caused by an improper host response to infection, remains a serious problem in the world of healthcare. In recent years, the role of ncRNA has emerged as a pivotal aspect in the intricate landscape of cellular regulation. The exploration of ncRNA-mediated regulatory networks reveals their profound influence on key molecular pathways orchestrating pyroptotic responses during septic conditions. Through a comprehensive analysis of current literature, we navigate the diverse classes of ncRNAs, including miRNAs, lncRNAs, and circRNAs, elucidating their roles as both facilitators and inhibitors in the modulation of pyroptotic processes. Furthermore, we highlight the potential diagnostic and therapeutic implications of targeting these ncRNAs in the context of sepsis, aiming to cover the method for novel and effective strategies to mitigate the devastating consequences of septic pathogenesis. As we unravel the complexities of this regulatory axis, a deeper understanding of the intricate crosstalk between ncRNAs and pyroptosis emerges, offering promising avenues for advancing our approach to sepsis intervention. The intricate pathophysiology of sepsis is examined in this review, which explores the dynamic interaction between ncRNAs and pyroptosis, a highly regulated kind of programmed cell death.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Yassine Riadi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, Uttar Pradesh 247341, India; Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, Jharkhand 831001, India
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Moyad Shahwan
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman 3467, United Arab Emirates; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, Ajman 3469, United Arab Emirates
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Aljouf 72341, Saudi Arabia
| | - Hairsh Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo-NSW 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology, Sydney, Ultimo-NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology, Sydney, Ultimo-NSW 2007, Australia
| | - Gaurav Gupta
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, Ajman 3469, United Arab Emirates; School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India.
| |
Collapse
|
27
|
Chen Q, Qian Q, Xu H, Zhou H, Chen L, Shao N, Zhang K, Chen T, Tian H, Zhang Z, Jones M, Kwan KYH, Sewell M, Shen S, Wang X, Khan MA, Makvandi P, Jin S, Zhou Y, Wu A. Mitochondrial-Targeted Metal-Phenolic Nanoparticles to Attenuate Intervertebral Disc Degeneration: Alleviating Oxidative Stress and Mitochondrial Dysfunction. ACS NANO 2024; 18:8885-8905. [PMID: 38465890 DOI: 10.1021/acsnano.3c12163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
As intervertebral disc degeneration (IVDD) proceeds, the dysfunctional mitochondria disrupt the viability of nucleus pulposus cells, initiating the degradation of the extracellular matrix. To date, there is a lack of effective therapies targeting the mitochondria of nucleus pulposus cells. Here, we synthesized polygallic acid-manganese (PGA-Mn) nanoparticles via self-assembly polymerization of gallic acid in an aqueous medium and introduced a mitochondrial targeting peptide (TP04) onto the nanoparticles using a Schiff base linkage, resulting in PGA-Mn-TP04 nanoparticles. With a size smaller than 50 nm, PGA-Mn-TP04 possesses pH-buffering capacity, avoiding lysosomal confinement and selectively accumulating within mitochondria through electrostatic interactions. The rapid electron exchange between manganese ions and gallic acid enhances the redox capability of PGA-Mn-TP04, effectively reducing mitochondrial damage caused by mitochondrial reactive oxygen species. Moreover, PGA-Mn-TP04 restores mitochondrial function by facilitating the fusion of mitochondria and minimizing their fission, thereby sustaining the vitality of nucleus pulposus cells. In the rat IVDD model, PGA-Mn-TP04 maintained intervertebral disc height and nucleus pulposus tissue hydration. It offers a nonoperative treatment approach for IVDD and other skeletal muscle diseases resulting from mitochondrial dysfunction, presenting an alternative to traditional surgical interventions.
Collapse
Affiliation(s)
- Qizhu Chen
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Qiuping Qian
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Hongbo Xu
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Hao Zhou
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Linjie Chen
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Nannan Shao
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Kai Zhang
- Ninth People's Hospital, Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
| | - Tao Chen
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Haijun Tian
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhiguang Zhang
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Morgan Jones
- Spine Unit, The Royal Orthopaedic Hospital, Bristol Road South, Northfield, Birmingham B31 2AP, U.K
| | - Kenny Yat Hong Kwan
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Mathew Sewell
- Spine Unit, The Royal Orthopaedic Hospital, Bristol Road South, Northfield, Birmingham B31 2AP, U.K
| | - Shuying Shen
- Department of Orthopaedics, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310013, China
| | - Xiangyang Wang
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Moonis Ali Khan
- Chemistry Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, Zhejiang, China
- Centre of Research Impact and Outcome, Chitkara University, Rajpura-140401, Punjab, India
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai-600077, India
| | - Shengwei Jin
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yunlong Zhou
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Aimin Wu
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| |
Collapse
|
28
|
Gao Q, Yan Y, Zhang J, Li X, Wang J, Feng Y, Li P, Wang H, Zhang Y, He L, Shan Z, Li B. Autologous cryo-shocked neutrophils enable targeted therapy of sepsis via broad-spectrum neutralization of pro-inflammatory cytokines and endotoxins. Front Chem 2024; 12:1359946. [PMID: 38449477 PMCID: PMC10914999 DOI: 10.3389/fchem.2024.1359946] [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: 12/22/2023] [Accepted: 01/25/2024] [Indexed: 03/08/2024] Open
Abstract
Background: Sepsis is a life-threatening disease characterized by multiple organ failure due to excessive activation of the inflammatory response and cytokine storm. Despite recent advances in the clinical use of anti-cytokine biologics, sepsis treatment efficacy and improvements in mortality remain unsatisfactory, largely due to the mechanistic complexity of immune regulation and cytokine interactions. Methods: In this study, a broad-spectrum anti-inflammatory and endotoxin neutralization strategy was developed based on autologous "cryo-shocked" neutrophils (CS-Neus) for the management of sepsis. Neutrophils were frozen to death using a novel liquid nitrogen "cryo-shock" strategy. The CS-Neus retained the source cell membrane structure and functions related to inflammatory site targeting, broad-spectrum inflammatory cytokines, and endotoxin (LPS) neutralizing properties. This strategy aimed to disable harmful pro-inflammatory functions of neutrophils, such as cytokine secretion. Autologous cell-based therapy strategies were employed to avoid immune rejection and enhance treatment safety. Results: In both LPS-induced sepsis mouse models and clinical patient-derived blood samples, CS-Neus treatment significantly ameliorated cytokine storms by removing inflammatory cytokines and endotoxin. The therapy showed notable anti-inflammatory therapeutic effects and improved the survival rate of mice. Discussion: The results of this study demonstrate the potential of autologous "cryo-shocked" neutrophils as a promising therapeutic approach for managing sepsis. By targeting inflammatory organs and exhibiting anti-inflammatory activity, CS-Neus offer a novel strategy to combat the complexities of sepsis treatment. Further research and clinical trials are needed to validate the efficacy and safety of this approach in broader populations and settings.
Collapse
Affiliation(s)
- Qiuxia Gao
- School of Inspection, Ningxia Medical University, Yinchuan, Ningxia, China
- Institute of Translational Medicine, The First People’s Hospital of Foshan, Foshan, Guangdong, China
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Yan Yan
- Department of Critical Care Medicine, Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jie Zhang
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of Education, Xizang Minzu University, Xianyang, Shaanxi, China
| | - Xiaoxue Li
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Jiamei Wang
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Yi Feng
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Peiran Li
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Huanhuan Wang
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Yunlong Zhang
- Department of Critical Care Medicine, Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lingjie He
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Zhiyan Shan
- Department of Histology and Embryology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Bin Li
- School of Inspection, Ningxia Medical University, Yinchuan, Ningxia, China
- Institute of Translational Medicine, The First People’s Hospital of Foshan, Foshan, Guangdong, China
| |
Collapse
|
29
|
Li X, Xing D, Bai Y, Du Y, Lang S, Li K, Xiang J, Liu G, Liu S. Injectable hydrogel with antimicrobial and anti-inflammatory properties for postoperative tumor wound care. Biomed Mater 2024; 19:025028. [PMID: 38290161 DOI: 10.1088/1748-605x/ad2408] [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: 09/07/2023] [Accepted: 01/30/2024] [Indexed: 02/01/2024]
Abstract
Clinically, tumor removal surgery leaves irregularly shaped wounds that are susceptible to bacterial infection and further lead to excessive inflammation. Injectable hydrogel dressings with antimicrobial and anti-inflammatory properties have been recognized as an effective strategy to care for postoperative tumor wounds and prevent recurrence in recent years. In this work, we constructed a hydrogel network by ionic bonding interactions between quaternized chitosan (QCS) and epigallocatechin gallate (EGCG)-Zn complexes which were coordinated by EGCG and zinc ions. Because of the synergistic effect of QCS and EGCG-Zn, the hydrogel exhibited outstanding antimicrobial capacity (>99.9% inhibition), which could prevent infections caused byEscherichia coli and Staphylococcus aureus. In addition, the hydrogel was able to inhibit the growth of mice breast cancer cells (56.81% survival rate within 72 h) and reduce inflammation, which was attributed to the sustained release of EGCG. The results showed that the hydrogel was effective in inhibiting tumor recurrence and accelerating wound closure when applied to the postoperative tumor wounds. This study provided a simple and reliable strategy for postoperative tumor wound care using antimicrobial and anti-inflammatory injectable dressings, confirming their great potential in the field of postoperative wound dressings.
Collapse
Affiliation(s)
- Xinyun Li
- Department of Oncology, Dazhou Integrated Traditional Chinese Medicine and Western Medicine Hospital, Dazhou Second People's Hospital, Dazhou, Sichuan 635000, People's Republic of China
| | - Dandan Xing
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Yangjing Bai
- West China School of Nursing, Sichuan University/Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Yangrui Du
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Shiying Lang
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Kaijun Li
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Jun Xiang
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Gongyan Liu
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Shan Liu
- Department of Endocrinology, Yueyang Central Hospital, Yueyang 414100, People's Republic of China
| |
Collapse
|
30
|
Xu Y, An X, Liu L, Cao X, Wu Z, Jia W, Sun J, Wang H, Huo J, Sun Z, Zhen M, Wang C, Bai C. Self-Cascade Redox Modulator Trilogically Renovates Intestinal Microenvironment for Mitigating Endotoxemia. ACS NANO 2024; 18:2131-2148. [PMID: 38198697 DOI: 10.1021/acsnano.3c09397] [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: 01/12/2024]
Abstract
Endotoxemia is a life-threatening multiple organ failure disease caused by bacterial endotoxin infection. Unfortunately, current single-target therapy strategies have failed to prevent the progression of endotoxemia. Here, we reported that alanine fullerene redox modulator (AFRM) remodeled the intestinal microenvironment for multiple targets endotoxemia mitigation by suppressing inflammatory macrophages, inhibiting macrophage pyroptosis, and repairing epithelial cell barrier integrity. Specifically, AFRM exhibited broad-spectrum and self-cascade redox regulation properties with superoxide dismutase (SOD)-like enzyme, peroxidase (POD)-like enzyme activity, and hydroxyl radical (•OH) scavenging ability. Guided by proteomics, we demonstrated that AFRM regulated macrophage redox homeostasis and down-regulated LPS/TLR4/NF-κB and MAPK/ERK signaling pathways to suppress inflammatory hyperactivation. Of note, AFRM could attenuate inflammation-induced macrophage pyroptosis via inhibiting the activation of gasdermin D (GSDMD). In addition, our results revealed that AFRM could restore extracellular matrix and cell-tight junction proteins and protect the epithelial cell barrier integrity by regulating extracellular redox homeostasis. Consequently, AFRM inhibited systemic inflammation and potentiated intestinal epithelial barrier damage repair during endotoxemia in mice. Together, our work suggested that fullerene based self-cascade redox modulator has the potential in the management of endotoxemia through synergistically remodeling the inflammation and epithelial barriers in the intestinal microenvironment.
Collapse
Affiliation(s)
- Yuan Xu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xin An
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinran Cao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhanfeng Wu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wang Jia
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiacheng Sun
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haoyu Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiawei Huo
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zihao Sun
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingming Zhen
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunru Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunli Bai
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
31
|
Wang M, Fu Q. Nanomaterials for Disease Treatment by Modulating the Pyroptosis Pathway. Adv Healthc Mater 2024; 13:e2301266. [PMID: 37354133 DOI: 10.1002/adhm.202301266] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/06/2023] [Indexed: 06/26/2023]
Abstract
Pyroptosis differs significantly from apoptosis and cell necrosis as an alternative mode of programmed cell death. Its occurrence is mediated by the gasdermin protein, leading to characteristic outcomes including cell swelling, membrane perforation, and release of cell contents. Research underscores the role of pyroptosis in the etiology and progression of many diseases, making it a focus of research intervention as scientists explore ways to regulate pyroptosis pathways in disease management. Despite numerous reviews detailing the relationship between pyroptosis and disease mechanisms, few delve into recent advancements in nanomaterials as a mechanism for modulating the pyroptosis pathway to mitigate disease effects. Therefore, there is an urgent need to fill this gap and elucidate the path for the use of this promising technology in the field of disease treatment. This review article delves into recent developments in nanomaterials for disease management through pyroptosis modulation, details the mechanisms by which drugs interact with pyroptosis pathways, and highlights the promise that nanomaterial research holds in driving forward disease treatment.
Collapse
Affiliation(s)
- Mengzhen Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, P. R. China
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, P. R. China
| |
Collapse
|
32
|
Wang J, Gan L, Li F, Li Q, Wu T, Wu Z, Chen P, Scicluna BP, Feng X, Gu J, Han W, Li N, Lei L. Tracheal epithelial cell-exosome-derived MiR-21-5p inhibits alveolar macrophage pyroptosis to resist pulmonary bacterial infection through PIK3CD-autophagy pathway. Life Sci 2024; 336:122340. [PMID: 38092143 DOI: 10.1016/j.lfs.2023.122340] [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: 03/30/2023] [Revised: 06/06/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023]
Abstract
AIMS Structural cells play an important role in regulating immune cells during infection. Our aim was to determine whether structural porcine tracheal epithelial cells (PTECs) can regulate alveolar macrophages (AMs) to prevent bacterial pneumonia, explore the underlying mechanism(s) and therapeutic target. MATERIALS AND METHODS Actinobacillus pleuropneumoniae (APP) was used as the model strain for infection studies. Small RNA sequencing was used to identify differentially abundant exosome-derived miRNAs. The role of PTECs exosome-derived miR-21-5p in regulating AMs autophagy, pyroptosis, reactive oxygen species (ROS) was determined using RT-qPCR, western-blotting, flow cytometry, immunohistochemistry. Luciferase reporter assays were conducted to identify potential binding targets of miR-21-5p. The universality of miR-21-5p action on resistance to bacterial pulmonary infection was demonstrated using Klebsiella pneumoniae or Staphylococcus aureus in vitro and in vivo infection models. KEY FINDINGS MiR-21-5p was enriched in PETCs-derived exosomes, which protected AMs against pulmonary bacterial infection. Mechanistically, miR-21-5p targeted PIK3CD, to promote autophagy of AMs, which reduced the pyroptosis induced by APP infection via inhibiting the over-production of ROS, which in turn suppressed the over-expression of pro-inflammatory cytokines, and increased bacterial clearance. Importantly, the protective effect and mechanism of miR-21-5p were universal as they also occurred upon challenge with Klebsiella pneumoniae and Staphylococcus aureus. SIGNIFICANCE Our data reveals miR-21-5p can promote pulmonary resistance to bacterial infection by inhibiting pyroptosis of alveolar macrophages through the PIK3CD-autophagy-ROS pathway, suggesting PIK3CD may be a potential therapeutic target for bacterial pneumonia.
Collapse
Affiliation(s)
- Jun Wang
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Lin Gan
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Fengyang Li
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Qin Li
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Tong Wu
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zengshuai Wu
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Peiru Chen
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Brendon P Scicluna
- Department of Applied Biomedical Science, Faculty of Health Sciences, Mater Dei Hospital, University of Malta, Msida, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Xin Feng
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jingmin Gu
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Wenyu Han
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Na Li
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Liancheng Lei
- State Key Laboratory for Zoonotic Diseases, College of Veterinary Medicine, Jilin University, Changchun, China.
| |
Collapse
|
33
|
Zhang Y, Li J, Jing Q, Chen Z, Wang K, Sun C. An Erythrocyte Membrane-Derived Nanosystem for Efficient Reversal of Endothelial Injury in Sepsis. Adv Healthc Mater 2024; 13:e2302320. [PMID: 37883686 DOI: 10.1002/adhm.202302320] [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: 07/21/2023] [Revised: 10/19/2023] [Indexed: 10/28/2023]
Abstract
Sepsis is caused by a disordered host immune in response to infection and endothelial cells perform a crucial role in boosting immunity reaction in the pathophysiology of sepsis and septic organ failure. The aim of this study is to construct a novel erythrocyte membrane-derived nanosystems to reverse endothelial damage in sepsis. Herein, an innovative nanometer calcium metal-organic framework (Ca-MOF) is generated for the first time by using chelidonic acid as a ligand and calcium chloride as an ion donor for anti-inflammation. Then, zoliflodacin is loaded into Ca-MOF (CMZ) to sterilize and nanoscale erythrocyte membrane vesicles are prepared by modification with a γ3 peptide on the surface (γ3-RM) for precise targeting. Finally, γ3-RM camouflages the nanocore CMZ, to form novel erythrocyte membrane-camouflaged nanoparticle γ3-RCMZ. The superior performance of novel nanosystem results from its suitable biocompatibility, nontoxicity, specific targeting, and anti-inflammatory and bactericidal effects. Its anti-inflammatory mechanism mainly involves inhibiting the Caspase1-nuclear factor kappa-B (Caspase1-NF-κB) pathway and oxidative stress reduction to alleviate endothelial damage. Moreover, the findings have revealed for the first time that the bactericidal drug zoliflodacin also has anti-inflammatory effects in vivo and in vitro. Therefore, the novel nanosystem (γ3-RCMZ) provides a new nanotherapy strategy for sepsis treatment.
Collapse
Affiliation(s)
- Yao Zhang
- Department of Emergency, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Jian Li
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Qi Jing
- Department of Emergency, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Ziying Chen
- Department of Emergency, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Kai Wang
- Department of Emergency, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Chuanzheng Sun
- Department of Emergency, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| |
Collapse
|
34
|
Liu AB, Li SJ, Yu YY, Zhang JF, Ma L. Current insight on the mechanisms of programmed cell death in sepsis-induced myocardial dysfunction. Front Cell Dev Biol 2023; 11:1309719. [PMID: 38161332 PMCID: PMC10754983 DOI: 10.3389/fcell.2023.1309719] [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: 10/15/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024] Open
Abstract
Sepsis is a clinical syndrome characterized by a dysregulated host response to infection, leading to life-threatening organ dysfunction. It is a high-fatality condition associated with a complex interplay of immune and inflammatory responses that can cause severe harm to vital organs. Sepsis-induced myocardial injury (SIMI), as a severe complication of sepsis, significantly affects the prognosis of septic patients and shortens their survival time. For the sake of better administrating hospitalized patients with sepsis, it is necessary to understand the specific mechanisms of SIMI. To date, multiple studies have shown that programmed cell death (PCD) may play an essential role in myocardial injury in sepsis, offering new strategies and insights for the therapeutic aspects of SIMI. This review aims to elucidate the role of cardiomyocyte's programmed death in the pathophysiological mechanisms of SIMI, with a particular focus on the classical pathways, key molecules, and signaling transduction of PCD. It will explore the role of the cross-interaction between different patterns of PCD in SIMI, providing a new theoretical basis for multi-target treatments for SIMI.
Collapse
Affiliation(s)
- An-Bu Liu
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Shu-Jing Li
- Department of Pediatrics Medical, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yuan-Yuan Yu
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jun-Fei Zhang
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Lei Ma
- Department of Emergency Medical, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
35
|
Yi Z, Ran Y, Chen X, Tong Q, Ma L, Tan Y, Ma X, Li X. Tea polyphenol carrier-enhanced dexamethasone nanomedicines for inflammation-targeted treatment of rheumatoid arthritis. J Mater Chem B 2023; 11:11505-11518. [PMID: 38038124 DOI: 10.1039/d3tb02316h] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease characterized by synovial inflammation, cartilage damage and bone erosion. In the progression of RA, the inflammatory mediators including ROS, NO, TNF-α, and IL-6 play important roles in the aggravation of inflammation. Hence, reducing the generation and release of inflammatory mediators is of great importance. However, the high dose and frequent administration of clinical anti-inflammatory drugs such as glucocorticoids (GCs) usually lead to severe side effects. The development of nanotechnology provides a promising strategy to overcome these issues. Here, polyphenol-based nanoparticles with inherent anti-oxidative and anti-inflammatory activities were developed and used as a kind of nanocarrier to deliver dexamethasone (Dex). The in vitro experiments confirmed that the nanoparticles and drugs could act synergistically for suppressing inflammatory mediators in the LPS/INF-γ-induced inflammatory cell model. After intravenous administration, the Dex-loaded nanoparticles with good biosafety showed effective accumulation in inflamed joints and improved therapeutic efficacy by inducing anesis of synovial inflammation and cartilage destruction over free Dex in a collagen-induced arthritis (CIA) mouse model. The results demonstrated that polyphenol-based nanoparticles with therapeutic functions may serve as an innovative platform to synergize with chemotherapeutic agents for enhanced treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Zeng Yi
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yaqin Ran
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xiangyu Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Qiulan Tong
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Lei Ma
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yunfei Tan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xiaomin Ma
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xudong Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|
36
|
Chen X, Zhang L, Zeng H, Meng W, Liu G, Zhang W, Zhao P, Zhang Q, Chen M, Chen J. Manganese-Based Immunomodulatory Nanocomposite with Catalase-Like Activity and Microwave-Enhanced ROS Elimination Ability for Efficient Rheumatoid Arthritis Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2304610. [PMID: 37632302 DOI: 10.1002/smll.202304610] [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: 06/01/2023] [Revised: 08/02/2023] [Indexed: 08/27/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease commonly associated with the accumulation of hyperactive immune cells (HICs), particularly macrophages of pro-inflammatory (M1) phenotype, accompanied by the elevated level of reactive oxygen species (ROS), decreased pH and O2 content in joint synovium. In this work, an immunomodulatory nanosystem (IMN) is developed for RA therapy by modulating and restoring the function of HICs in inflamed tissues. Manganese tetraoxide nanoparticles (Mn3 O4 ) nanoparticles anchored on UiO-66-NH2 are designed, and then the hybrid is coated with Mn-EGCG film, further wrapped with HA to obtain the final nanocomposite of UiO-66-NH2 @Mn3 O4 /Mn-EGCG@HA (termed as UMnEH). When UMnEH diffuses to the inflammatory site of RA synovium, the stimulation of microwave (MW) irradiation and low pH trigger the slow dissociation of Mn-EGCG film. Then the endogenously overexpressed hydrogen peroxide (H2 O2 ) disintegrates the exposed Mn3 O4 NPs to promote ROS scavenging and O2 generation. Assisted by MW irradiation, the elevated O2 content in the RA microenvironment down-regulates the expression of hypoxia-inducible factor-1α (HIF-1α). Coupled with the clearance of ROS, it promotes the re-polarization of M1 phenotype macrophages into anti-inflammatory (M2) phenotype macrophages. Therefore, the multifunctional UMnEH nanoplatform, as the IMN, exhibits a promising potential to modulate and restore the function of HICs and has an exciting prospect in the treatment of RA.
Collapse
Affiliation(s)
- Xiaotong Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Lianying Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Haifeng Zeng
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Wei Meng
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Guijiang Liu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Wenhua Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Pei Zhao
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Office of Clinical Trial of Drug, Guangzhou, Guangdong, 510663, China
| | - Qun Zhang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Office of Clinical Trial of Drug, Guangzhou, Guangdong, 510663, China
| | - Ming Chen
- The People's Hospital of Gaozhou, Maoming, Guangdong, 525200, China
| | - Jinxiang Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| |
Collapse
|
37
|
Zhu Q, Han Y, Wang X, Jia R, Zhang J, Liu M, Zhang W. Hypoxia exacerbates intestinal injury and inflammatory response mediated by myeloperoxidase during Salmonella Typhimurium infection in mice. Gut Pathog 2023; 15:62. [PMID: 38037141 PMCID: PMC10688069 DOI: 10.1186/s13099-023-00586-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 11/21/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND High-altitude exposure can cause oxidative stress damage in the intestine, which leads to increased intestinal permeability and bacterial translocation, resulting in local and systemic inflammation. Control of infection is critically dependent on the host's ability to kill pathogens with reactive oxygen species (ROS). Myeloperoxidase (MPO) targets ROS in pathogens. This study aimed to investigate the effects of hypoxia on the colonic mucosal barrier and myeloperoxidase (MPO)-mediated innate immune response in the colon. METHODS AND RESULTS Genetically engineered mice were exposed to a hypobaric oxygen chamber for 3 days and an inflammation model was established using Salmonella Typhimurium infection. We found that hypoxic exposure caused the development of exacerbated bacterial colitis and enhanced bacterial dissemination in MPO-deficient mice. Infection and disease severity were associated with significantly increased Ly6G+ neutrophil and F4/80+ macrophage counts in infected tissues, which is consistent with elevated proinflammatory cytokines and chemoattractant molecules. Hypoxia restrained antioxidant ability and MPO deficiency aggravated the respiratory burst in the colon. CONCLUSION Hypoxia can damage the colonic mucosa. MPO mediates the innate immune response and regulates the mucosal and systemic inflammatory responses to Salmonella infection during hypoxia.
Collapse
Affiliation(s)
- Qinfang Zhu
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Ying Han
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Xiaozhou Wang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Ruhan Jia
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Jingxuan Zhang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Meiheng Liu
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Wei Zhang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China.
| |
Collapse
|
38
|
Guo J, Miao Y, Nie F, Gao F, Li H, Wang Y, Liu Q, Zhang T, Yang X, Liu L, Fan H, Wang Q, Qiao H. Zn-Shik-PEG nanoparticles alleviate inflammation and multi-organ damage in sepsis. J Nanobiotechnology 2023; 21:448. [PMID: 38001490 PMCID: PMC10675904 DOI: 10.1186/s12951-023-02224-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 11/19/2023] [Indexed: 11/26/2023] Open
Abstract
Sepsis is defined as a life-threatening organ dysfunction caused by excessive formation of reactive oxygen species (ROS) and dysregulated inflammatory response. Previous studies have reported that shikonin (Shik) possess prominent anti-inflammatory and antioxidant effects and holds promise as a potential therapeutic drug for sepsis. However, the poor water solubility and the relatively high toxicity of shikonin hamper its clinical application. To address this challenge, we constructed Zn2+-shikonin nanoparticles, hereafter Zn-Shik-PEG NPs, based on an organic-inorganic hybridization strategy of metal-polyphenol coordination to improve the aqueous solubility and biosafety of shikonin. Mechanistic studies suggest that Zn-Shik-PEG NPs could effectively clear intracellular ROS via regulating the Nrf2/HO-1 pathway, meanwhile Zn-Shik-PEG NPs could inhibit NLRP3 inflammasome-mediated activation of inflammation and apoptosis by regulating the AMPK/SIRT1 pathway. As a result, the Zn-Shik-PEG NPs demonstrated excellent therapeutic efficacies in lipopolysaccharide (LPS) as well as cecal ligation puncture (CLP) induced sepsis model. These findings suggest that Zn-Shik-PEG NPs may have therapeutic potential for the treatment of other ROS-associated and inflammatory diseases.
Collapse
Affiliation(s)
- Jie Guo
- Shaanxi Collaborative Innovation Center of TCM Technologies and Devices, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Yuqing Miao
- Shaanxi Collaborative Innovation Center of TCM Technologies and Devices, Shaanxi University of Chinese Medicine, Xianyang, 712046, China.
| | - Fayi Nie
- Shaanxi Collaborative Innovation Center of TCM Technologies and Devices, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Fei Gao
- Shaanxi Collaborative Innovation Center of TCM Technologies and Devices, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Hua Li
- Shaanxi Collaborative Innovation Center of TCM Technologies and Devices, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Yuan Wang
- Shaanxi Key Laboratory of Integrated Acupuncture and Drugs, College of Acupuncture and Tuina, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Qi Liu
- Shaanxi Key Laboratory of Integrated Acupuncture and Drugs, College of Acupuncture and Tuina, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Tingbin Zhang
- Center for Health Science and Engineering, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300130, China
| | - Xiaohang Yang
- Shaanxi Collaborative Innovation Center of TCM Technologies and Devices, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Li Liu
- Shaanxi Collaborative Innovation Center of TCM Technologies and Devices, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Haiming Fan
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, 710069, China
| | - Qiang Wang
- Shaanxi Key Laboratory of Integrated Acupuncture and Drugs, College of Acupuncture and Tuina, Shaanxi University of Chinese Medicine, Xianyang, 712046, China.
| | - Haifa Qiao
- Shaanxi Collaborative Innovation Center of TCM Technologies and Devices, Shaanxi University of Chinese Medicine, Xianyang, 712046, China.
- Shaanxi Key Laboratory of Integrated Acupuncture and Drugs, College of Acupuncture and Tuina, Shaanxi University of Chinese Medicine, Xianyang, 712046, China.
| |
Collapse
|
39
|
Zhu R, Qin F, Zheng X, Fang S, Ding J, Wang D, Liang L. Single-molecule lipopolysaccharides identification and the interplay with biomolecules via nanopore readout. Biosens Bioelectron 2023; 240:115641. [PMID: 37657310 DOI: 10.1016/j.bios.2023.115641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/03/2023]
Abstract
Lipopolysaccharides (LPS) are the major constituent on the cell envelope of all gram-negative bacteria. They are ubiquitous in air, and are toxic inflammatory stimulators for urinary disorders and sepsis. The reported optical, thermal, and electrochemical sensors via the intermolecular interplay of LPS with proteins and aptamers are generally complicated methods. We demonstrate the single-molecule nanopore approach for LPS identification in distinct bacteria as well as the serotypes discrimination. With a 4 nm nanopore, we achieve a detection limit of 10 ng/mL. Both the antibiotic polymyxin B (PMB) and DNA aptamer display specific binding to LPS. The identification of LPS in both human serum and tap water show good performance with nanopore platforms. Our work shows a highly-sensitive and easy-to-handle scheme for clinical and environmental biomarkers determination and provides a promising screening tool for early warning of contamination in water and medical supplies.
Collapse
Affiliation(s)
- Rui Zhu
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences & Chongqing School, University of Chinese Academy of Science, Chongqing, 400714, PR China; Chongqing Jiaotong University, Chongqing, 400014, PR China
| | - Fupeng Qin
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences & Chongqing School, University of Chinese Academy of Science, Chongqing, 400714, PR China
| | - Xinchuan Zheng
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences & Chongqing School, University of Chinese Academy of Science, Chongqing, 400714, PR China
| | - Shaoxi Fang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences & Chongqing School, University of Chinese Academy of Science, Chongqing, 400714, PR China
| | - Jianjun Ding
- Southwest University, Chongqing, 400715, PR China
| | - Deqiang Wang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences & Chongqing School, University of Chinese Academy of Science, Chongqing, 400714, PR China.
| | - Liyuan Liang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences & Chongqing School, University of Chinese Academy of Science, Chongqing, 400714, PR China.
| |
Collapse
|
40
|
Zhou H, Qian Q, Chen Q, Chen T, Wu C, Chen L, Zhang Z, Wu O, Jin Y, Wang X, Guo Z, Sun J, Zhang J, Shen S, Wang X, Jones M, Khan MA, Makvandi P, Zhou Y, Wu A. Enhanced Mitochondrial Targeting and Inhibition of Pyroptosis with Multifunctional Metallopolyphenol Nanoparticles in Intervertebral Disc Degeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023:e2308167. [PMID: 37953455 DOI: 10.1002/smll.202308167] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/29/2023] [Indexed: 11/14/2023]
Abstract
Intervertebral disc degeneration (IVDD) is a significant contributor to low back pain, characterized by excessive reactive oxygen species generation and inflammation-induced pyroptosis. Unfortunately, there are currently no specific molecules or materials available to effectively delay IVDD. This study develops a multifunctional full name of PG@Cu nanoparticle network (PG@Cu). A designed pentapeptide, bonded on PG@Cu nanoparticles via a Schiff base bond, imparts multifunctionality to the metal polyphenol particles (PG@Cu-FP). PG@Cu-FP exhibits enhanced escape from lysosomal capture, enabling efficient targeting of mitochondria to scavenge excess reactive oxygen species. The scavenging activity against reactive oxygen species originates from the polyphenol-based structures within the nanoparticles. Furthermore, Pyroptosis is effectively blocked by inhibiting Gasdermin mediated pore formation and membrane rupture. PG@Cu-FP successfully reduces the activation of the nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 inflammasome by inhibiting Gasdermin protein family (Gasdermin D, GSDMD) oligomerization, leading to reduced expression of Nod-like receptors. This multifaceted approach demonstrates higher efficiency in inhibiting Pyroptosis. Experimental results confirm that PG@Cu-FP preserves disc height, retains water content, and preserves tissue structure. These findings highlight the potential of PG@Cu-FP in improving IVDD and provide novel insights for future research in IVDD treatments.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Qiuping Qian
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Qizhu Chen
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Tao Chen
- Department of Orthopaedics, Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education Tongji Hospital, Tongji University School of Medicine, School of Life Science and Technology, Tongji University, Shanghai, 200065, China
| | - Chenyu Wu
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Linjie Chen
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Zhiguang Zhang
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Ouqiang Wu
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yuxin Jin
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xinzhou Wang
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Zhenyu Guo
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Jing Sun
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Jun Zhang
- Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou, 551700, China
| | - Shuying Shen
- Department of Orthopaedics, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Xiangyang Wang
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Morgan Jones
- Spine Unit, The Royal Orthopaedic Hospital, Bristol Road South, Northfield, Birmingham, B31 2AP, United Kingdom
| | - Moonis Ali Khan
- Chemistry Department, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, 324000, China
| | - Yunlong Zhou
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, China
| | - Aimin Wu
- Department of Orthopaedics, Key Laboratory of Structural Malformations in Children of Zhejiang Province, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| |
Collapse
|
41
|
Wang W, He Z. Gasdermins in sepsis. Front Immunol 2023; 14:1203687. [PMID: 38022612 PMCID: PMC10655013 DOI: 10.3389/fimmu.2023.1203687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Sepsis is a hyper-heterogeneous syndrome in which the systemic inflammatory response persists throughout the course of the disease and the inflammatory and immune responses are dynamically altered at different pathogenic stages. Gasdermins (GSDMs) proteins are pore-forming executors in the membrane, subsequently mediating the release of pro-inflammatory mediators and inflammatory cell death. With the increasing research on GSDMs proteins and sepsis, it is believed that GSDMs protein are one of the most promising therapeutic targets in sepsis in the future. A more comprehensive and in-depth understanding of the functions of GSDMs proteins in sepsis is important to alleviate the multi-organ dysfunction and reduce sepsis-induced mortality. In this review, we focus on the function of GSDMs proteins, the molecular mechanism of GSDMs involved in sepsis, and the regulatory mechanism of GSDMs-mediated signaling pathways, aiming to provide novel ideas and therapeutic strategies for the diagnosis and treatment of sepsis.
Collapse
Affiliation(s)
- Wenhua Wang
- Department of Intensive Care Unit, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhihui He
- Department of Intensive Care Unit, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, Hunan, China
| |
Collapse
|
42
|
Wu J, Cai J, Tang Y, Lu B. The noncanonical inflammasome-induced pyroptosis and septic shock. Semin Immunol 2023; 70:101844. [PMID: 37778179 DOI: 10.1016/j.smim.2023.101844] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 09/10/2023] [Accepted: 09/22/2023] [Indexed: 10/03/2023]
Abstract
Sepsis remains one of the most common and lethal conditions globally. Currently, no proposed target specific to sepsis improves survival in clinical trials. Thus, an in-depth understanding of the pathogenesis of sepsis is needed to propel the discovery of effective treatment. Recently attention to sepsis has intensified because of a growing recognition of a non-canonical inflammasome-triggered lytic mode of cell death termed pyroptosis upon sensing cytosolic lipopolysaccharide (LPS). Although the consequences of activation of the canonical and non-canonical inflammasome are similar, the non-canonical inflammasome formation requires caspase-4/5/11, which enzymatically cleave the pore-forming protein gasdermin D (GSDMD) and thereby cause pyroptosis. The non-canonical inflammasome assembly triggers such inflammatory cell death by itself; or leverages a secondary activation of the canonical NLRP3 inflammasome pathway. Excessive cell death induced by oligomerization of GSDMD and NINJ1 leads to cytokine release and massive tissue damage, facilitating devastating consequences and death. This review summarized the updated mechanisms that initiate and regulate non-canonical inflammasome activation and pyroptosis and highlighted various endogenous or synthetic molecules as potential therapeutic targets for treating sepsis.
Collapse
Affiliation(s)
- Junru Wu
- Department of Cardiology, The 3rd Xiangya Hospital, Central South University, Changsha 410000, PR China
| | - Jingjing Cai
- Department of Cardiology, The 3rd Xiangya Hospital, Central South University, Changsha 410000, PR China
| | - Yiting Tang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410000, PR China
| | - Ben Lu
- Department of Critical Care Medicine and Hematology, The 3rd Xiangya Hospital, Central South University, Changsha 410000, PR China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha 410000, PR China.
| |
Collapse
|
43
|
He W, Fu D, Gai Y, Liu X, Yang C, Ye Z, Chen X, Liu J, Chang B. An infection-microenvironment-targeted and responsive peptide-drug nanosystem for sepsis emergency by suppressing infection and inflammation. Asian J Pharm Sci 2023; 18:100869. [PMID: 38161786 PMCID: PMC10755722 DOI: 10.1016/j.ajps.2023.100869] [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: 07/25/2023] [Revised: 10/13/2023] [Accepted: 11/19/2023] [Indexed: 01/03/2024] Open
Abstract
Sepsis is a life-threatening emergency that causes millions of deaths every year due to severe infection and inflammation. Nevertheless, current therapeutic regimens are inadequate to promptly address the vast diversity of potential pathogens. Omiganan, an antimicrobial peptide, has shown promise for neutralizing endotoxins and eliminating diverse pathogens. However, its clinical application is hindered by safety and stability concerns. Herein, we present a nanoscale drug delivery system (Omi-hyd-Dex@HA NPs) that selectively targets infectious microenvironments (IMEs) and responds to specific stimuli for efficient intervention in sepsis. The system consists of omiganan-dexamethasone conjugates linked by hydrazone bonds which self-assemble into nanoparticles coated with a hyaluronic acid (HA). The HA coating not only facilitates IMEs-targeting through interaction with intercellular-adhesion-molecule-1 on inflamed endotheliocytes, but also improves the biosafety of the nanosystem and enhances drug accumulation in primary infection sites triggered by hyaluronidase. The nanoparticles release dual drugs in IMEs through pH-sensitive cleavage of hydrazone bonds to eradicate pathogens and suppress inflammation. In multiple tissue infection and sepsis animal models, Omi-hyd-Dex@HA NPs exhibited rapid source control and comprehensive inflammation reduction, thereby preventing subsequent fatal complications and significantly improving survival outcomes. The bio-responsive and self-delivering nanosystem offers a promising strategy for systemic sepsis treatment in emergencies.
Collapse
Affiliation(s)
- Wei He
- The Second Clinical College, The Second Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang 550003, China
| | - Daan Fu
- Department of Anesthesiology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yongkang Gai
- Department of Nuclear Medicine, Hubei Province Key Laboratory of Molecular Imaging, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xingxin Liu
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu 610065, China
| | - Chang Yang
- Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, China
| | - Zhilan Ye
- Department of Geriatrics, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xu Chen
- The Second Clinical College, The Second Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang 550003, China
| | - Jia Liu
- Research Center for Tissue Engineering and Regenerative Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bingcheng Chang
- The Second Clinical College, The Second Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang 550003, China
| |
Collapse
|
44
|
Weng X, Luo X, Dai X, Lv Y, Zhang S, Bai X, Bao X, Wang Y, Zhao C, Zeng M, Hu S, Li J, Jia H, Yu B. Apigenin inhibits macrophage pyroptosis through regulation of oxidative stress and the NF-κB pathway and ameliorates atherosclerosis. Phytother Res 2023; 37:5300-5314. [PMID: 37526050 DOI: 10.1002/ptr.7962] [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: 11/12/2022] [Revised: 06/21/2023] [Accepted: 07/02/2023] [Indexed: 08/02/2023]
Abstract
Pyroptosis plays an important role in inflammatory diseases such as viral hepatitis and atherosclerosis. Apigenin exhibits various bioactivities, particularly anti-inflammation, but its effect on pyroptosis remains unclear. The aim of this study is to investigate the effect of apigenin on pyroptosis and explore its potential against inflammatory diseases. THP-1 macrophages treated by lipopolysaccharides/adenosine 5'-triphosphate were used as the in vitro pyroptosis model. Western blot was used to detect the expression of NLRP3 inflammasome components and key regulators. Immunofluorescence was used to observe ROS production and intracellular location of p65. The potential of apigenin against inflammatory diseases was evaluated using atherosclerotic mice. Plaque progression was observed by pathological staining. Immunofluorescence was used to observe the expression of NLRP3 inflammasome components in plaques. The results showed that apigenin inhibited NLRP3 inflammasome activation. Apigenin reduced ROS overproduction and inhibited p65 nuclear translocation. Additionally, apigenin decreased the expression of NLRP3 inflammasome components in the plaque. Plaque progression was inhibited by apigenin. In conclusion, apigenin exhibited a preventive effect on macrophage pyroptosis by reducing oxidative stress and inhibiting the NF-κB pathway. Apigenin may alleviate atherosclerosis at least partially by inhibiting macrophage pyroptosis. These findings suggest apigenin to be a promising therapeutic agent for inflammatory diseases.
Collapse
Affiliation(s)
- Xiuzhu Weng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, People's Republic of China
| | - Xing Luo
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, People's Republic of China
| | - Xinyu Dai
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, People's Republic of China
| | - Ying Lv
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, People's Republic of China
| | - Shan Zhang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, People's Republic of China
| | - Xiaoxuan Bai
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, People's Republic of China
| | - Xiaoyi Bao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, People's Republic of China
| | - Ying Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, People's Republic of China
| | - Chen Zhao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, People's Republic of China
| | - Ming Zeng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, People's Republic of China
| | - Sining Hu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, People's Republic of China
| | - Ji Li
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, People's Republic of China
| | - Haibo Jia
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, People's Republic of China
| | - Bo Yu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases, Harbin, People's Republic of China
| |
Collapse
|
45
|
Zhong L, Han J, Fan X, Huang Z, Su L, Cai X, Lin S, Chen X, Huang W, Dai S, Ye B. Novel GSDMD inhibitor GI-Y1 protects heart against pyroptosis and ischemia/reperfusion injury by blocking pyroptotic pore formation. Basic Res Cardiol 2023; 118:40. [PMID: 37782407 DOI: 10.1007/s00395-023-01010-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 10/03/2023]
Abstract
Activation of gasdermin D (GSDMD) and its concomitant cardiomyocyte pyroptosis are critically involved in multiple cardiac pathological conditions. Pharmacological inhibition or gene knockout of GSDMD could protect cardiomyocyte from pyroptosis and dysfunction. Thus, seeking and developing highly potent GSDMD inhibitors probably provide an attractive strategy for treating diseases targeting GSDMD. Through structure-based virtual screening, pharmacological screening and subsequent pharmacological validations, we preliminarily identified GSDMD inhibitor Y1 (GI-Y1) as a selective GSDMD inhibitor with cardioprotective effects. Mechanistically, GI-Y1 binds to GSDMD and inhibits lipid- binding and pyroptotic pore formation of GSDMD-N by targeting the Arg7 residue. Importantly, we confirmed the cardioprotective effect of GI-Y1 on myocardial I/R injury and cardiac remodeling by targeting GSDMD. More extensively, GI-Y1 also inhibited the mitochondrial binding of GSDMD-N and its concomitant mitochondrial dysfunction. The findings of this study identified a new drug (GI-Y1) for the treatment of cardiac disorders by targeting GSDMD, and provide a new tool compound for pyroptosis research.
Collapse
Affiliation(s)
- Lingfeng Zhong
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jibo Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Xiaoxi Fan
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhouqing Huang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lan Su
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xueli Cai
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shuang Lin
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xudong Chen
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weijian Huang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Shanshan Dai
- The Key Laboratory of Emergency and Disaster Medicine of Wenzhou, Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Bozhi Ye
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
46
|
Jin H, Zhao Y, Yao Y, Zhao J, Luo R, Fan S, Wei Y, Ouyang S, Peng W, Zhang Y, Pi J, Huang G. Therapeutic effects of tea polyphenol-loaded nanoparticles coated with platelet membranes on LPS-induced lung injury. Biomater Sci 2023; 11:6223-6235. [PMID: 37529873 DOI: 10.1039/d3bm00802a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
Patients with ALI (acute lung injury)/ARDS (acute respiratory distress syndrome) are often septic and with poor prognosis, which leads to a high mortality rate of 25-40%. Despite the advances in medicine, there are no effective pharmacological therapies for ALI/ARDS due to the short systemic circulation and poor specificity in the lungs. To address this problem, we prepared TP-loaded nanoparticles (TP-NPs) through the emulsification-and-evaporation method, and then the platelet membrane vesicles were extracted and coated onto the surface of the NPs to constitute the biomimetic PM@TP-NPs. In a LPS-induced ALI mouse model, PM@TP-NPs showed good biocompatibility and biosafety, which was evidenced by no significant toxic effect on cell viability and no hemolysis of red blood cells. In ALI mice, the PM@TP-NPs showed favorable anti-inflammation and enhanced therapeutic activity of TPs compared to the free drug. Administration of PM@TP-NPs effectively inhibited lung vascular injury, evidenced by the decreased lung vascular permeability, reduced pro-inflammatory cytokine burden, evidenced by decreased inflammatory cell (macrophages, neutrophils, etc.) infiltration in the bronchoalveolar lavage fluid (BALF) and lung tissues, and inhibited the secretion of pro-inflammatory cytokines and NLRP3 inflammasome activation. ALI/ARDS is defined by damage to the alveolar epithelium and endothelium; thus, effective intervention targeting pulmonary vascular endothelial cells (VECs) is crucial for the treatment of respiratory diseases. For further determination of the targeting of PM cloaked NPs, healthy mice were also administered with the same NPs. Interestingly, the PM cloaked NPs only showed highly efficient targeting to the inflamed lungs and VECs, but no accumulation in healthy lungs and VECs. The data demonstrated that this biomimetic nanoplatform could be used as a potential strategy for personalized therapies in the treatment of inflammatory diseases, such as ALI/ARDS, and even COVID-19-associated pneumonia.
Collapse
Affiliation(s)
- Hua Jin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, China.
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Yue Zhao
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, China.
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Yinlian Yao
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Jin Zhao
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Renxing Luo
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, China.
- School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Shilong Fan
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Yanlan Wei
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Suidong Ouyang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, China.
| | - Wanqing Peng
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Yumin Zhang
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China
| | - Jiang Pi
- School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Gonghua Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
47
|
Bao Y, Ge Y, Wu M, Mao Z, Ye J, Tong W. Record-High Ultrasound-Sensitive NO Nanogenerators for Cascade Tumor Pyroptosis and Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302278. [PMID: 37400368 PMCID: PMC10502831 DOI: 10.1002/advs.202302278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/30/2023] [Indexed: 07/05/2023]
Abstract
Pyroptosis is a pro-inflammatory cell death that is associated with innate immunity promotion against tumors. Excess nitric oxide (NO)-triggered nitric stress has potential to induce pyroptosis, but the precise delivery of NO is challenging. Ultrasound (US)-responsive NO production has dominant priority due to its deep penetration, low side effects, noninvasion, and local activation manner. In this work, US-sensitive NO donor N-methyl-N-nitrosoaniline (NMA) with thermodynamically favorable structure is selected and loaded into hyaluronic acid (HA)-modified hollow manganese dioxide nanoparticles (hMnO2 NPs) to fabricate hMnO2 @HA@NMA (MHN) nanogenerators (NGs). The obtained NGs have a record-high NO generation efficiency under US irradiation and can release Mn2+ after targeting the tumor sites. Later on, cascade tumor pyroptosis and cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING)-based immunotherapy is achieved and tumor growth is effectively inhibited.
Collapse
Affiliation(s)
- Yuheng Bao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationMinistry of EducationDepartment of Polymer Science and EngineeringZhejiang UniversityHangzhouZhejiang310027China
| | - Yanni Ge
- Eye CenterThe Second Affiliated HospitalSchool of MedicineZhejiang UniversityZhejiang Provincial Key Laboratory of OphthalmologyZhejiang Provincial Clinical Research Center for Eye DiseasesZhejiang Provincial Engineering Institute on Eye DiseasesHangzhouZhejiang310009China
| | - Mengjie Wu
- Stomatology HospitalSchool of StomatologyZhejiang University School of MedicineZhejiang Provincial Clinical Research Center for Oral DiseasesKey Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang UniversityHangzhouZhejiang310058China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationMinistry of EducationDepartment of Polymer Science and EngineeringZhejiang UniversityHangzhouZhejiang310027China
| | - Juan Ye
- Eye CenterThe Second Affiliated HospitalSchool of MedicineZhejiang UniversityZhejiang Provincial Key Laboratory of OphthalmologyZhejiang Provincial Clinical Research Center for Eye DiseasesZhejiang Provincial Engineering Institute on Eye DiseasesHangzhouZhejiang310009China
| | - Weijun Tong
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationMinistry of EducationDepartment of Polymer Science and EngineeringZhejiang UniversityHangzhouZhejiang310027China
| |
Collapse
|
48
|
Guo X, Shen P, Shao R, Hong T, Liu W, Shen Y, Su F, Wang Q, He B. Efferocytosis-inspired nanodrug treats sepsis by alleviating inflammation and secondary immunosuppression. Biomed Mater 2023; 18:055020. [PMID: 37567216 DOI: 10.1088/1748-605x/acef9a] [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: 06/10/2023] [Accepted: 08/10/2023] [Indexed: 08/13/2023]
Abstract
Uncontrolled inflammation storm induced by sepsis may lead to severe organ dysfunction and secondary immunosuppression, which is one of the main reasons for high mortality and prolonged hospitalization of septic patients. However, there is a lack of effective treatments for it at present. Here, we report an efferocytosis-inspired nanodrug (BCN@M) to treat sepsis and secondary immunosuppression via regulating the macrophage function. Bioactive molecular curcumin was loaded with bovine serum albumin and then coated with the damaged erythrocyte membrane derived from septic mice. It was found that the septic erythrocytes promoted the efferocytosis signal and BCN@M uptake efficiency by macrophages. The well-constructed BCN@M nanodrug reduced the hyperinflammation in sepsis and restored the bacterial clearance ability of macrophage in the secondary immunosuppression state. This study highlights BCN@M as an efferocytosis-inspired nanodrug to alleviate hyperinflammation and secondary immunosuppression of sepsis.
Collapse
Affiliation(s)
- Xiaoyu Guo
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Centre for Cardiopulmonary Translational Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Department of Anesthesiology, Huadong Hospital affiliated to Fudan University, Shanghai, People's Republic of China
| | - Peiming Shen
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Rongjiao Shao
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Centre for Cardiopulmonary Translational Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Ting Hong
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Department of Anesthesiology, Huadong Hospital affiliated to Fudan University, Shanghai, People's Republic of China
| | - Weizhuo Liu
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Centre for Cardiopulmonary Translational Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yi Shen
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Centre for Cardiopulmonary Translational Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Fan Su
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Centre for Cardiopulmonary Translational Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Qinlan Wang
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Bin He
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
49
|
Wen X, Wan F, Wu Y, Liu L, Liu Y, Zhong R, Chen L, Zhang H. Caffeic acid supplementation ameliorates intestinal injury by modulating intestinal microbiota in LPS-challenged piglets. Food Funct 2023; 14:7705-7717. [PMID: 37547959 DOI: 10.1039/d3fo02286b] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
During weaning, piglets are susceptible to intestinal injuries caused by a range of infections, which result in serious economic losses for pig producers. Caffeic acid (CA) is a plant-derived phenolic acid that exhibits potential as a dietary supplement for enhancing intestinal health. There is, however, limited information available about the potential benefits of CA supplementation on intestinal injury and growth performance in piglets. A 28-day study was conducted to examine the effectiveness of CA supplementation in protecting against intestinal injury induced by intraperitoneal injection of Escherichia coli lipopolysaccharide (LPS) in piglets. Twenty-four piglets (7.43 ± 0.79 kg body weight; Duroc × Landrace × Large White; barrows) were randomly divided into 4 groups: the control group, the LPS group, the LPS + CA group, and the CA group. Piglets were administered with LPS or saline on d21 and d28 of the experiment. Supplementation with CA improved intestinal barrier function in LPS-challenged piglets by enhancing intestinal morphology and integrity, as well as increasing the expression of Claudin-1 and ZO-1. Meanwhile, CA supplementation improved the systemic and colonic inflammation responses, oxidative stress, and apoptosis induced by LPS. CA supplementation improved the alpha diversity and structure of the intestinal microbiota by increasing the abundance of beneficial microbiota. Additionally, it was found that it improves metabolic disorders of colonic bile acids (BAs) and short-chain fatty acids (SCFAs) in LPS-challenged piglets, including an increase in primary BAs and isovalerate. In conclusion, CA supplementation could enhance intestinal integrity and barrier function by modifying intestinal microbiota and its metabolites, which could lead to a reduction in inflammatory responses and oxidative stress and ultimately enhanced growth performance in piglets.
Collapse
Affiliation(s)
- Xiaobin Wen
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - Fan Wan
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
- State Key Laboratory of Grassland Agro-Ecosystem, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, China
| | - You Wu
- College of Biological Science and Engineering, Beijing University of Agriculture, Beijing 102206, China
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Lei Liu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - Yueping Liu
- College of Biological Science and Engineering, Beijing University of Agriculture, Beijing 102206, China
| | - Ruqing Zhong
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - Liang Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
50
|
Chen H, Guo L, Ding J, Zhou W, Qi Y. A General and Efficient Strategy for Gene Delivery Based on Tea Polyphenols Intercalation and Self-Polymerization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302620. [PMID: 37349886 PMCID: PMC10460882 DOI: 10.1002/advs.202302620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/19/2023] [Indexed: 06/24/2023]
Abstract
Gene therapy that employs therapeutic nucleic acids to modulate gene expression has shown great promise for diseases therapy, and its clinical application relies on the development of effective gene vector. Herein a novel gene delivery strategy by just using natural polyphenol (-)-epigallocatechin-3-O-gallate (EGCG) as raw material is reported. EGCG first intercalates into nucleic acids to yield a complex, which then oxidizes and self-polymerizes to form tea polyphenols nanoparticles (TPNs) for effective nucleic acids encapsulation. This is a general method to load any types of nucleic acids with single or double strands and short or long sequences. Such TPNs-based vector achieves comparable gene loading capacity to commonly used cationic materials, but showing lower cytotoxicity. TPNs can effectively penetrate inside cells, escape from endo/lysosomes, and release nucleic acids in response to intracellular glutathione to exert biological functions. To demonstrate the in vivo application, an anti-caspase-3 small interfering ribonucleic acid is loaded into TPNs to treat concanavalin A-induced acute hepatitis, and excellent therapeutic efficacy is obtained in combination with the intrinsic activities of TPNs vector. This work provides a simple, versatile, and cost-effective gene delivery strategy. Given the biocompatibility and intrinsic biofunctions, this TPNs-based gene vector holds great potential to treat various diseases.
Collapse
Affiliation(s)
- Hao Chen
- Department of PathologyZhanjiang Central HospitalGuangdong Medical UniversityZhanjiangGuangdong524000China
- Department of PathologyShihezi University School of MedicineShiheziXinjiang832002China
| | - Lina Guo
- Department of PharmaceuticsXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Jinsong Ding
- Department of PharmaceuticsXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Wenhu Zhou
- Department of PharmaceuticsXiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410013China
| | - Yan Qi
- Department of PathologyZhanjiang Central HospitalGuangdong Medical UniversityZhanjiangGuangdong524000China
- Department of PathologyShihezi University School of MedicineShiheziXinjiang832002China
| |
Collapse
|