1
|
Zhang M, Cui Y, Liu P, Mo R, Wang H, Li Y, Wu Y. Oat β-(1 → 3, 1 → 4)-d-glucan alleviates food allergy-induced colonic injury in mice by increasing Lachnospiraceae abundance and butyrate production. Carbohydr Polym 2024; 344:122535. [PMID: 39218555 DOI: 10.1016/j.carbpol.2024.122535] [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: 01/19/2024] [Revised: 07/18/2024] [Accepted: 07/21/2024] [Indexed: 09/04/2024]
Abstract
Oat β-(1 → 3, 1 → 4)-d-glucan (OBG), a linear polysaccharide primarily found in oat bran, has been demonstrated to possess immunomodulatory properties and regulate gut microbiota. This study aimed to investigate the impact of low molecular weight (Mw) OBG (155.2 kDa) on colonic injury and allergic symptoms induced by food allergy (FA), and to explore its potential mechanism. In Experiment 1, results indicated that oral OBG improved colonic inflammation and epithelial barrier, and significantly relieved allergy symptoms. Importantly, the OBG supplement altered the gut microbiota composition, particularly increasing the abundance of Lachnospiraceae and its genera, and promoted the production of short-chain fatty acids, especially butyrate. However, in Experiment 2, the gut microbial depletion eliminated these protective effects of OBG on the colon in allergic mice. Further, in Experiment 3, fecal microbiota transplantation and sterile fecal filtrate transfer directly validated the role of OBG-mediated gut microbiota and its metabolites in relieving FA and its induced colonic injury. Our findings suggest that low Mw OBG can alleviate FA-induced colonic damage by increasing Lachnospiraceae abundance and butyrate production, and provide novel insights into the health benefits and mechanisms of dietary polysaccharide intervention for FA.
Collapse
Affiliation(s)
- Mingrui Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Yingyue Cui
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Pan Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Ruixia Mo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Haotian Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Yingying Li
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Yi Wu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
2
|
An K, Shi B, Lv X, Liu Y, Xia Z. T-2 toxin triggers lipid metabolism disorder and oxidative stress in liver of ducks. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 286:117169. [PMID: 39405967 DOI: 10.1016/j.ecoenv.2024.117169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024]
Abstract
T-2 toxin (T-2) is a highly toxic mycotoxin that threatens organism health, yet its hepatoxicity on ducks remains unknown. The present study aimed to assess the hepatoxicity and redox reactions induced by T-2 in ducks. Sixty 7-day-old ducklings were divided into 4 groups and exposed to 0, 200, 400 and 800 μg/kg bodyweight of T-2 through oral gavage for 2 weeks. The growth performance, liver histopathology, biochemical indicators, antioxidant capacity and hepatic damage-related genes of ducks were analyzed. The results revealed that 800 µg/kg T-2 inhibited the growth and feed intake of ducks, whereas liver index increased with the elevation of T-2 concentration. Histological examinations exhibited that T-2 caused hepatic cord disappeared and severe steatosis. Moreover, serum AST, ALT and TG were substantially higher in 400 μg/kg group, while γ-GT and ALB were reduced under 800 μg/kg T-2 exposure. In addition, significant increase of malondialdehyde (MDA) in liver, decrease of hepatic total antioxidant capacity (T-AOC) and serum glutathione peroxidase (GPx) were observed in all T-2 groups. Furthermore, T-2 disrupted lipid metabolism and oxidative stress-related genes expression in liver. The transcript level of fatty acid binding protein 1 (FABP1) was markedly raised in all T-2 groups, and hepatic acyl-CoA oxidase 1 (ACOX1) was significantly raised in 200 and 400 μg/kg T-2 groups. Under 800 μg/kg T-2, significant induction of hypoxia inducible factor-1 alpha (HIF-1α), and downregulated peroxisome proliferator-activated receptor (PPAR)-alpha, carnitine palmitoyl transferase 1A (CPT1A), peroxisome proliferator-activated receptor gamma coactivator 1alpha (PGC-1α), GPx1, catalase (CAT) mRNA levels were observed. Therefore, we conclude that T-2 caused liver injury through lipid metabolism disruption and oxidative stress in ducks, which reinforces understanding about the hepatoxicity mechanisms of T-2 and provides new targets for detoxication and prevention.
Collapse
Affiliation(s)
- Keying An
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Bozhi Shi
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Xueze Lv
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; Beijing General Station of Animal Husbandry, Beijing 100107, China
| | - Yanhan Liu
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; Shandong Provincial Center for Animal Disease Control, Jinan 250100, China
| | - Zhaofei Xia
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
3
|
Zhao M, Yang J, Liang J, Shi R, Song W. Emerging nanozyme therapy incorporated into dental materials for diverse oral pathologies. Dent Mater 2024; 40:1710-1728. [PMID: 39107224 DOI: 10.1016/j.dental.2024.07.025] [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: 12/07/2023] [Revised: 06/25/2024] [Accepted: 07/24/2024] [Indexed: 08/09/2024]
Abstract
OBJECTIVE Nanozyme materials combine the advantages of natural enzymes and artificial catalysis, and have been widely applied in new technologies for dental materials and oral disease treatment. Based on the role of reactive oxygen species (ROS) and oxidative stress pathways in the occurrence and therapy of oral diseases, a comprehensive review was conducted on the methods and mechanisms of nanozymes and their dental materials in treating different oral diseases. METHODS This review is based on literature surveys from PubMed and Web of Science databases, as well as reviews of relevant researches and publications on nanozymes in the therapy of oral diseases and oral tumors in international peer-reviewed journals. RESULTS Given the unique function of nanozymes in the generation and elimination of ROS, they play an important role in the occurrence, development, and treatment of different oral diseases. The application of nanozymes in dental materials and oral disease treatment was introduced, including the latest advances in their use for dental caries, pulpitis, jaw osteomyelitis, periodontitis, oral mucosal diseases, temporomandibular joint disorders, and oral tumors. Future approaches were also summarized and proposed based on the characteristics of these diseases. SIGNIFICANCE This review will guide biomedical researchers and oral clinicians to understand the mechanisms and applications of nanozymes in the therapy of oral diseases, promoting further development in the field of dental materials within the oral medication. It is anticipated that more suitable therapeutic agents or dental materials encapsulating nanozymes, specifically designed for the oral environment and simpler for clinical utilization, will emerge in the forthcoming future.
Collapse
Affiliation(s)
- Menghan Zhao
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, China; Department of Orthodontics, School and Hospital of Stomatology, Jilin University, China
| | - Jin Yang
- College of Basic Medical Sciences, Jilin University, China
| | - Jiangyi Liang
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, China
| | - Ruixin Shi
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, China.
| | - Wei Song
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, China.
| |
Collapse
|
4
|
Yang W, Yuan H, Sun H, Hu T, Xu Y, Qiu Y, Li Y. Co-Mn Complex Oxide Nanoparticles as Potential Reactive Oxygen Species Scavenging Agents for Pulmonary Fibrosis Treatment. Molecules 2024; 29:5106. [PMID: 39519747 PMCID: PMC11547682 DOI: 10.3390/molecules29215106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/14/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and age-related lung disease that has few treatment options. Reactive oxygen species (ROS) play an important role in the introduction and development of IPF. In the present study, we developed multifunctional Cobalt (Co)-Manganese (Mn) complex oxide nanoparticles (Co-MnNPs), which can scavenge multiple types of ROS. Benefiting from ROS scavenging activities and good biosafety, Co-MnNPs can suppress canonical and non-canonical TGF-β pathways and, thus, inhibit the activation of fibroblasts and the productions of extracellular matrix. Furthermore, the scavenging of ROS by Co-MnNPs reduce the LPS-induced expressions of pro-inflammatory factors in macrophages, by suppressing NF-κB signaling pathway. Therefore, Co-MnNPs can reduce the excessive extracellular matrix deposition and inflammatory responses in lungs and, thus, alleviate pulmonary fibrosis induced by bleomycin (BLM) in mice. Taken together, this work offers an anti-fibrotic agent for treatment of IPF and other ROS-related diseases.
Collapse
Affiliation(s)
- Wuhao Yang
- College of Materials, Xiamen University, Xiamen 361005, China;
| | - Hui Yuan
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; (H.Y.); (H.S.); (T.H.)
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institutes, Chinese Academy of Sciences, Xiamen 361021, China
| | - Hao Sun
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; (H.Y.); (H.S.); (T.H.)
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institutes, Chinese Academy of Sciences, Xiamen 361021, China
| | - Ting Hu
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; (H.Y.); (H.S.); (T.H.)
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institutes, Chinese Academy of Sciences, Xiamen 361021, China
| | - Yaping Xu
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen Medical College, Xiamen 361023, China;
| | - Yan Qiu
- School of Medicine, Xiamen University, Xiamen 361102, China
- Xiamen Key Laboratory of Chiral Drugs, Xiamen 361102, China
| | - Yuhang Li
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; (H.Y.); (H.S.); (T.H.)
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institutes, Chinese Academy of Sciences, Xiamen 361021, China
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen Medical College, Xiamen 361023, China;
| |
Collapse
|
5
|
Yang Y, Zhang C, Lin L, Li Q, Wang M, Zhang Y, Yu Y, Hu D, Wang X. Multifunctional MnGA Nanozymes for the Treatment of Ulcerative Colitis by Reducing Intestinal Inflammation and Regulating the Intestinal Flora. ACS APPLIED MATERIALS & INTERFACES 2024; 16:56884-56901. [PMID: 39401179 DOI: 10.1021/acsami.4c14291] [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: 10/25/2024]
Abstract
In ulcerative colitis (UC), the formation of an inflammatory environment is due to the combined effects of excess production of reactive oxygen species (ROS) and reactive nitrogen species (RNS), overproduction of proinflammatory cytokines, and disruption of immune system function. There are many kinds of traditional drugs for the clinical treatment of UC, but long-term drug use can cause toxic side effects and drug resistance and can also reduce patient compliance and other drawbacks. Hence, in light of the clinical challenges associated with UC, including the limitations of existing treatments, intense adverse reactions and the development of resistance to medications, no novel therapeutic agents that offer effective relief and maintain a high level of biosafety are urgently needed. Although many anti-inflammatory nanomedicines have been developed by researchers, the development of efficient and nontoxic nanomedicines is still a major challenge in clinical medicine. Using the natural product gallic acid and the metal compound manganese chloride, a highly effective and nontoxic multifunctional nanoenzyme was developed for the treatment of UC. Nanozymes can effectively eliminate ROS and RNS to reduce the inflammation of intestinal epithelial cells caused by oxidation, facilitate the restoration of the intestinal epithelial barrier through the upregulation of tight junction protein expression, and balance the intestinal microbiota to maintain the stability of the intestinal environment. Using a rodent model designed to mimic UC, we monitored body weight, colon length, the spleen index, and the degree of tissue damage and demonstrated that manganese gallate (MnGA) nanoparticles can reduce intestinal inflammation by clearing ROS and active nitrogen. Intestinal flora sequencing revealed that MnGA nanoparticles could regulate the intestinal flora, promote the growth of beneficial bacteria and decrease the levels of detrimental bacteria within the intestinal tract in a mouse model of UC. Thus, MnGA nanoparticles can maintain the balance of the intestinal flora. This study demonstrated that MnGA nanoparticles are excellent antioxidant and effective anti-inflammatory agents, have good biosafety, and can effectively treat UC.
Collapse
Affiliation(s)
- Yan Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
- Department of Gastroenterology, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230011, China
| | - Cong Zhang
- Department of Gastroenterology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Liting Lin
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, China
| | - Qingrong Li
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, China
| | - Min Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, China
| | - Yiqun Zhang
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, China
| | - Yue Yu
- Department of Gastroenterology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Duanmin Hu
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Xianwen Wang
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
6
|
Cao B, Da X, Wu W, Xie J, Li X, Wang X, Xu H, Gao J, Yang H, Su J. Multifunctional human serum albumin-crosslinked and self-assembling nanoparticles for therapy of periodontitis by anti-oxidation, anti-inflammation and osteogenesis. Mater Today Bio 2024; 28:101163. [PMID: 39183771 PMCID: PMC11341939 DOI: 10.1016/j.mtbio.2024.101163] [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: 03/07/2024] [Revised: 07/02/2024] [Accepted: 07/18/2024] [Indexed: 08/27/2024] Open
Abstract
Periodontitis is a chronic inflammatory disease that can result in the irreversible loss of tooth-supporting tissues and elevate the likelihood and intensity of systemic diseases. The presence of reactive oxygen species (ROS) and associated related oxidative stress is intricately linked to the progression and severity of periodontal inflammation. Targeted removal of local ROS may serve to attenuate inflammation, improve the unfavorable periodontal microenvironment and potentially reverse ensuing pathological cascades. These ROS scavenging nanoparticles, which possess additional characteristics such as anti-inflammation and osteogenic differentiation, are highly sought after for the treatment of periodontitis. In this study, negative charged human serum albumin-crosslinked manganese-doped self-assembling Prussian blue nanoparticles (HSA-MDSPB NPs) were fabricated. These nanoparticles demonstrate the ability to scavenge multiple ROS including superoxide anion, free hydroxyl radicals, singlet oxygen and hydrogen peroxide. Additionally, HSA-MDSPB NPs exhibit the capacity to alleviate inflammation in gingiva and alveolar bone both in vitro and in vivo. Furthermore, HSA-MDSPB NPs have been shown to play a role in promoting the polarization of macrophages from the M1 to M2 phenotype, resulting in reduced production of pro-inflammatory cytokines. More attractively, HSA-MDSPB NPs have been demonstrated to enhance cellular osteogenic differentiation. These properties of HSA-MDSPB NPs contribute to decreased inflammation, extracellular matrix degradation and bone loss in periodontal tissue. In conclusion, the multifunctional nature of HSA-MDSPB NPs provides a promising therapeutic approach for the treatment of periodontitis.
Collapse
Affiliation(s)
- Bangping Cao
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Xuanbo Da
- Department of General Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710000, China
| | - Wenjing Wu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Jian Xie
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Xuejing Li
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Xin Wang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Hui Xu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Jianfang Gao
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Hui Yang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| | - Jiansheng Su
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Prosthodontics, Stomatological Hospital and Dental School, Tongji University, Shanghai, China
| |
Collapse
|
7
|
Hu S, Zhao R, Chen T, Chi X, Li Y, Wu D, Zhu B, Hu J. Construction of chlorogenic acid nanoparticles for effective alleviation of ulcerative colitis. Food Funct 2024; 15:9085-9099. [PMID: 39157985 DOI: 10.1039/d4fo02122c] [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: 08/20/2024]
Abstract
The onset and progression of ulcerative colitis (UC) are intricately linked to the worsening of intestinal inflammation, an imbalance in oxidative stress, and impairment of the intestinal mucosal barrier. Although chlorogenic acid (CA) shows potential in effectively alleviating the symptoms of UC, its clinical application is hindered by its poor bioavailability, stability, rapid metabolism, and quick excretion. This study utilized a one-step enzyme-catalyzed polymerization technique to create chlorogenic acid nanoparticles (CA NPs), aiming to improve the bioavailability and stability of CA. The CA NPs exhibited an optimal nanosize (106.65 ± 4.12 nm) and showed increased cellular uptake over time. Importantly, CA NPs significantly prolonged retention time in inflamed colonic tissues, enhancing accumulation and providing a targeted therapy for UC. Animal studies confirmed the substantial benefits of CA NPs, including reduced weight loss, lessened reduction in colon length, and a lowered disease activity index (DAI) score in DSS-induced UC mice. Moreover, CA NPs effectively reduced oxidative stress and levels of inflammatory factors in the colonic tissues of UC mice, thus mitigating tissue damage and restoring the integrity of the intestinal mucosal barrier. In conclusion, our research proposes a novel approach to increase the bioavailability and stability of CA, offering a promising avenue for its effective application in preventing UC.
Collapse
Affiliation(s)
- Shumeng Hu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, PR China.
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, PR China
| | - Runan Zhao
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, PR China.
- College of Biosystems Engineering and Food Science, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, PR China
| | - Tao Chen
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, PR China.
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, PR China
| | - Xuesong Chi
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, PR China.
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, PR China
| | - Yangjing Li
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, PR China.
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, PR China
| | - Di Wu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, PR China.
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, PR China
| | - Beiwei Zhu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, PR China.
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, PR China
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, PR China
| | - Jiangning Hu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, PR China.
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, PR China
| |
Collapse
|
8
|
Cui X, Guo J, Yuan P, Dai Y, Du P, Yu F, Sun Z, Zhang J, Cheng K, Tang J. Bioderived Nanoparticles for Cardiac Repair. ACS NANO 2024; 18:24622-24649. [PMID: 39185722 DOI: 10.1021/acsnano.3c07878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Biobased therapy represents a promising strategy for myocardial repair. However, the limitations of using live cells, including the risk of immunogenicity of allogeneic cells and inconsistent therapeutic efficacy of autologous cells together with low stability, result in an unsatisfactory clinical outcomes. Therefore, cell-free strategies for cardiac tissue repair have been proposed as alternative strategies. Cell-free strategies, primarily based on the paracrine effects of cellular therapy, have demonstrated their potential to inhibit apoptosis, reduce inflammation, and promote on-site cell migration and proliferation, as well as angiogenesis, after an infarction and have been explored preclinically and clinically. Among various cell-free modalities, bioderived nanoparticles, including adeno-associated virus (AAV), extracellular vesicles, cell membrane-coated nanoparticles, and exosome-mimetic nanovesicles, have emerged as promising strategies due to their improved biological function and therapeutic effect. The main focus of this review is the development of existing cellular nanoparticles and their fundamental working mechanisms, as well as the challenges and opportunities. The key processes and requirements for cardiac tissue repair are summarized first. Various cellular nanoparticle modalities are further highlighted, together with their advantages and limitations. Finally, we discuss various delivery approaches that offer potential pathways for researchers and clinicians to translate cell-free strategies for cardiac tissue repair into clinical practice.
Collapse
Affiliation(s)
- Xiaolin Cui
- Cardiac and Osteochondral Tissue Engineering (COTE) Group, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Jiacheng Guo
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan 450052, China
| | - Peiyu Yuan
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan 450052, China
| | - Yichen Dai
- Cardiac and Osteochondral Tissue Engineering (COTE) Group, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Pengchong Du
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan 450052, China
| | - Fengyi Yu
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan 450052, China
| | - Zhaowei Sun
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan 450052, China
| | - Jinying Zhang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan 450052, China
| | - Ke Cheng
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Junnan Tang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan 450052, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan 450052, China
| |
Collapse
|
9
|
Dong F, Hao L, Wang L, Huang Y. Clickable nanozyme enhances precise colonization of probiotics for ameliorating inflammatory bowel disease. J Control Release 2024; 373:749-765. [PMID: 39084465 DOI: 10.1016/j.jconrel.2024.07.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Convincing evidence suggests that aberrant gut microbiota changes play a critical role in the progression and pathogenesis of inflammatory bowel disease (IBD). Probiotic therapeutic interventions targeting the microbiota may provide alternative avenues to treat IBD, but currently available probiotics often suffer from low intestinal colonization and limited targeting capability. Here, we developed azido (N3)-modified Prussian blue nanozyme (PB@N3) spatio-temporal guidance enhances the targeted colonization of probiotics to alleviate intestinal inflammation. First, clickable PB@N3 targets intestinal inflammation, simultaneously, it scavenges reactive oxygen species (ROS). Subsequently, utilizing "click" chemistry to spatio-temporally guide targeted colonization of dibenzocyclooctyne (DBCO)-modified Lactobacillus reuteri DSM 17938 (LR@DBCO). The "click" reaction between PB@N3 and LR@DBCO has excellent specificity and efficacy both in vivo and in vitro. Despite the complex physiological environment of IBD, "click" reaction can prolong the retention time of probiotics in the intestine. Dextran sulfate sodium (DSS)-induced colitis mice model, demonstrates that the combination of PB@N3 and LR@DBCO effectively mitigates levels of ROS, enhances the colonization of probiotics, modulates intestinal flora composition and function, regulates immune profiles, restores intestinal barrier function, and alleviates intestinal inflammation. Hence, PB@N3 spatio-temporal guidance enhances targeted colonization of LR@DBCO provides a promising medical treatment strategy for IBD.
Collapse
Affiliation(s)
- Fang Dong
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, National Children's Medical Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Liangwen Hao
- The Institute for Biomedical Engineering and Nano Science School of Medicine, Tongji University, Shanghai 200072, China
| | - Lin Wang
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, National Children's Medical Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Ying Huang
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, National Children's Medical Center, Children's Hospital of Fudan University, Shanghai 201102, China.
| |
Collapse
|
10
|
Liu Y, Li C, Yang X, Yang B, Fu Q. Stimuli-responsive polymer-based nanosystems for cardiovascular disease theranostics. Biomater Sci 2024; 12:3805-3825. [PMID: 38967109 DOI: 10.1039/d4bm00415a] [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: 07/06/2024]
Abstract
Stimulus-responsive polymers have found widespread use in biomedicine due to their ability to alter their own structure in response to various stimuli, including internal factors such as pH, reactive oxygen species (ROS), and enzymes, as well as external factors like light. In the context of atherosclerotic cardiovascular diseases (CVDs), stimulus-response polymers have been extensively employed for the preparation of smart nanocarriers that can deliver therapeutic and diagnostic drugs specifically to inflammatory lesions. Compared with traditional drug delivery systems, stimulus-responsive nanosystems offer higher sensitivity, greater versatility, wider applicability, and enhanced biosafety. Recent research has made significant contributions towards designing stimulus-responsive polymer nanosystems for CVDs diagnosis and treatment. This review summarizes recent advances in this field by classifying stimulus-responsive polymer nanocarriers according to different responsiveness types and describing numerous stimuli relevant to these materials. Additionally, we discuss various applications of stimulus-responsive polymer nanomaterials in CVDs theranostics. We hope that this review will provide valuable insights into optimizing the design of stimulus-response polymers for accelerating their clinical application in diagnosing and treating CVDs.
Collapse
Affiliation(s)
- Yuying Liu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Congcong Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| | - Bin Yang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
11
|
Wang J, Wei X, Chen J, Zhang J, Guo Y, Xin Y. Versatile Ce(III)‐Terephthalic Acid@Au Metal Organic Frameworks for ROS Elimination and Photothermal Sterilization. CHEMNANOMAT 2024; 10. [DOI: 10.1002/cnma.202400073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Indexed: 10/01/2024]
Abstract
AbstractNanozymes have been widely used for treating reactive oxygen species (ROS) caused diseases. However, the ROS‐dependent antibacterial property is inevitably damaged during the process of scavenging ROS, which is unfavorable for the treatment of diseases related to both ROS accumulation and bacterial infections. To address the issues, biomedical materials with both ROS‐elimination ability and ROS‐independent antibacterial capacity are fabricated via in situ depositing spherical Au nanoparticles (Au NPs) on rough surface of metal organic frameworks composed of Ce(III) and terephthalic acid (Ce‐BDC@Au MOFs). The synthesized Ce‐BDC@Au MOFs show multi‐enzymatic activities owing to the reversible conversion between Ce3+ and Ce4+, and can significantly scavenge ROS in cells. The deposition of spherical Au NPs on surface of Ce‐BDC MOFs causes Au NPs to come close proximity for forming plasmon resonance coupling, inducing the resonance wavelength of Au NPs red shifted to NIR region. Based on this, Ce‐BDC@Au MOFs show good photothermal conversion efficiency under NIR laser (808 nm) irradiation. Benefitting from rough surface and photothermal conversion ability, Ce‐BDC@Au MOFs have high antibacterial efficiency against staphylococcus aureus through both mechanically damaging and photothermal destruction. This strategy is biosafety and effectiveness for treating diseases related to both ROS accumulation and bacterial infections.
Collapse
Affiliation(s)
- Jing Wang
- Department of Child and Adolescent Health School of Public Health Zhengzhou University Zhengzhou Henan 450001 P.R. China
| | - Xue Wei
- Henan Key Laboratory of Nanocomposite and Applications Institute of Nanostructured Functional Materials Huanghe Science and Technology College Zhengzhou Henan 450006 P.R. China
| | - Jian Chen
- Henan Key Laboratory of Nanocomposite and Applications Institute of Nanostructured Functional Materials Huanghe Science and Technology College Zhengzhou Henan 450006 P.R. China
| | - Jing Zhang
- Henan Key Laboratory of Nanocomposite and Applications Institute of Nanostructured Functional Materials Huanghe Science and Technology College Zhengzhou Henan 450006 P.R. China
| | - Yanzhen Guo
- Henan Key Laboratory of Nanocomposite and Applications Institute of Nanostructured Functional Materials Huanghe Science and Technology College Zhengzhou Henan 450006 P.R. China
| | - Yongjuan Xin
- Department of Child and Adolescent Health School of Public Health Zhengzhou University Zhengzhou Henan 450001 P.R. China
| |
Collapse
|
12
|
Cui Y, Yang D, Li Q, Peng Z, Zhong Z, Song Y, Han Q, Yang Y. Cu,Zn,I-Doped Carbon Dots with Boosted Triple Antioxidant Nanozyme Activity for Treatment of DSS-Induced Colitis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:32619-32632. [PMID: 38860867 DOI: 10.1021/acsami.4c03627] [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: 06/12/2024]
Abstract
Nanozyme-mediated antioxidative therapy is a promising star for treating a myriad of important diseases through eliminating excessive reactive oxygen species (ROS) such as O2·- and H2O2, a critical mechanism for inflammatory bowel disease (IBD). This work provides a high biocompatibility iodine-copper-zinc covalent doped carbon dots (Cu,Zn,I-CDs) with the catalase (CAT)-, superoxide dismutase (SOD)- and glutathione peroxidase (GPx)-like catalytic activities for treating ulcerative colitis (UC) by scavenging overproduced ROS. We found that I dopant aids in counteracting the positive charge at Cu,Zn dopants brought on by low pH, enabling Cu,Zn,I-CDs to process strong triple antioxidant nanozyme activities rather than Cu,Zn-CDs. Vitro experiments displayed that the Cu,Zn,I-CDs could scavenge the excessive ROS to protect cellular against oxidative stress and reduce the expression of proinflammatory cytokines, such as TNF-α, IL-1β, and IL-6. In sodium dextran sulfate (DSS)-induced colitis mice models, Cu,Zn,I-CDs with excellent biocompatibility could effectively relieve the inflammation of the colon, containing the reduction of the colon length, the damaged epithelium, the infiltration of inflammatory cells, and upregulation of antioxidant genes. Therefore, the therapy of Cu,Zn,I-CD antioxidant nanozymes is an effective approach and provides a novel strategy for UC treatment.
Collapse
Affiliation(s)
- Yifan Cui
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Dezhi Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Qiulan Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Zhongmei Peng
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Zitao Zhong
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Yuzhu Song
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Qinqin Han
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Yaling Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| |
Collapse
|
13
|
Wang Q, Jin L, Yang H, Yu L, Cao X, Mao Z. Bacteria/Nanozyme Composites: New Therapeutics for Disease Treatment. SMALL METHODS 2024:e2400610. [PMID: 38923867 DOI: 10.1002/smtd.202400610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/11/2024] [Indexed: 06/28/2024]
Abstract
Bacterial therapy is recognized as a cost-effective treatment for several diseases. However, its development is hindered by limited functionality, weak inherent therapeutic effects, and vulnerability to harsh microenvironmental conditions, leading to suboptimal treatment activity. Enhancing bacterial activity and therapeutic outcomes emerges as a pivotal challenge. Nanozymes have garnered significant attention due to their enzyme-mimic activities and high stability. They enable bacteria to mimic the functions of gene-edited bacteria expressing the same functional enzymes, thereby improving bacterial activity and therapeutic efficacy. This review delineates the therapeutic mechanisms of bacteria and nanozymes, followed by a summary of strategies for preparing bacteria/nanozyme composites. Additionally, the synergistic effects of such composites in biomedical applications such as gastrointestinal diseases and tumors are highlighted. Finally, the challenges of bacteria/nanozyme composites are discussed and propose potential solutions. This study aims to provide valuable insights to offer theoretical guidance for the advancement of nanomaterial-assisted bacterial therapy.
Collapse
Affiliation(s)
- Qirui Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Lulu Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Huang Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Lisha Yu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Xinran Cao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transvascular Implantation Devices, Zhejiang, Hangzhou, 310009, China
| |
Collapse
|
14
|
Hu C, Yuan X, Zhao R, Hong B, Chen C, Zhu Q, Zheng Y, Hu J, Yuan Y, Wu Z, Zhang J, Tang C. Scale-Up Preparation of Manganese-Iron Prussian Blue Nanozymes as Potent Oral Nanomedicines for Acute Ulcerative Colitis. Adv Healthc Mater 2024; 13:e2400083. [PMID: 38447228 DOI: 10.1002/adhm.202400083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/04/2024] [Indexed: 03/08/2024]
Abstract
Prussian blue (PB) nanozymes are demonstrated as effective therapeutics for ulcerative colitis (UC), yet an unmet practical challenge remains in the scalable production of these nanozymes and uncertainty over their efficacy. With a novel approach, a series of porous manganese-iron PB (MnPB) colloids, which are shown to be efficient scavengers for reactive oxygen species (ROS) including hydroxyl radical, superoxide anion, and hydrogen peroxide, are prepared. In vitro cellular experiments confirm the capability of the nanozyme to protect cells from ROS attack. In vivo, the administration of MnPB nanozyme through gavage at a dosage of 10 mg kg-1 per day for three doses in total potently ameliorates the pathological symptoms of acute UC in a murine model, resulting in mitigated inflammatory responses and improved viability rate. Significantly, the nanozyme produced at a large scale can be achieved at an unprecedented yield weighting ≈11 g per batch of reaction, demonstrating comparable anti-ROS activities and treatment efficacy to its small-scale counterpart. This work represents the first demonstration of the scale-up preparation of PB analog nanozymes for UC without compromising treatment efficacy, laying the foundation for further testing of these nanozymes on larger animals and promising clinical translation.
Collapse
Affiliation(s)
- Chengyun Hu
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Xue Yuan
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Ronghua Zhao
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Biao Hong
- College & Hospital of Stomatology, Anhui Provincial Key Laboratory of Oral Diseases Research, Anhui Medical University, Hefei, 230032, China
| | - Chuang Chen
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Qingjun Zhu
- Anhui Provincial Key Laboratory of High Magnetic Resonance Image, High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Yanmin Zheng
- Anhui Provincial Key Laboratory of High Magnetic Resonance Image, High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Jinming Hu
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Yue Yuan
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Zhengyan Wu
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Jia Zhang
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Department of Chemistry, University of Science and Technology of China, Hefei, 230026, China
| | - Chaoliang Tang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| |
Collapse
|
15
|
Sui Y, Jiang R, Niimi M, Wang X, Xu Y, Zhang Y, Shi Z, Suda M, Mao Z, Fan J, Yao J. Gut bacteria exacerbates TNBS-induced colitis and kidney injury through oxidative stress. Redox Biol 2024; 72:103140. [PMID: 38593629 PMCID: PMC11016804 DOI: 10.1016/j.redox.2024.103140] [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: 02/05/2024] [Revised: 03/18/2024] [Accepted: 03/27/2024] [Indexed: 04/11/2024] Open
Abstract
Gut microbiota has been implicated in the initiation and progression of various diseases; however, the underlying mechanisms remain elusive and effective therapeutic strategies are scarce. In this study, we investigated the role and mechanisms of gut microbiota in TNBS-induced colitis and its associated kidney injury while evaluating the potential of dietary protein as a therapeutic intervention. The intrarectal administration of TNBS induced colitis in mice, concurrently with kidney damage. Interestingly, this effect was absent when TNBS was administered intraperitoneally, indicating a potential role of gut microbiota. Depletion of gut bacteria with antibiotics significantly attenuated the severity of TNBS-induced inflammation, oxidative damage, and tissue injury in the colon and kidneys. Mechanistic investigations using cultured colon epithelial cells and bone-marrow macrophages unveiled that TNBS induced cell oxidation, inflammation and injury, which was amplified by the bacterial component LPS and mitigated by thiol antioxidants. Importantly, in vivo administration of thiol-rich whey protein entirely prevented TNBS-induced colonic and kidney injury. Our findings suggest that gut bacteria significantly contribute to the initiation and progression of colitis and associated kidney injury, potentially through mechanisms involving LPS-induced exaggeration of oxidative cellular damage. Furthermore, our research highlights the potential of dietary thiol antioxidants as preventive and therapeutic interventions.
Collapse
Affiliation(s)
- Yang Sui
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Rui Jiang
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Manabu Niimi
- Division of Molecular Pathology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Xin Wang
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Yijun Xu
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Yingyu Zhang
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Zhuheng Shi
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Mika Suda
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Zhimin Mao
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan; Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, Jiangsu, China.
| | - Jianglin Fan
- Division of Molecular Pathology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan.
| | - Jian Yao
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan.
| |
Collapse
|
16
|
Shi Z, Li X, Chen J, Dai Z, Zhu Y, Wu T, Liu Q, Qin H, Zhang Y, Chen H. Enzyme-like biomimetic oral-agent enabling modulating gut microbiota and restoring redox homeostasis to treat inflammatory bowel disease. Bioact Mater 2024; 35:167-180. [PMID: 38318229 PMCID: PMC10839225 DOI: 10.1016/j.bioactmat.2024.01.016] [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: 11/23/2023] [Revised: 01/16/2024] [Accepted: 01/16/2024] [Indexed: 02/07/2024] Open
Abstract
Reactive oxygen species (ROS), immune dysregulation-induced inflammatory outbreaks and microbial imbalance play critical roles in the development of inflammatory bowel disease (IBD). Herein, a novel enzyme-like biomimetic oral-agent ZnPBA@YCW has been developed, using yeast cell wall (YCW) as the outer shell and zinc-doped Prussian blue analogue (ZnPBA) nanozyme inside. When orally administered, the ZnPBA@YCW is able to adhere to Escherichia coli occupying the ecological niche in IBD and subsequently release the ZnPBA nanozyme for removal of E. coli, meanwhile exhibiting improved intestinal epithelial barrier repair. Moreover, it is found that the ZnPBA nanozyme exhibits remarkable capability in restoring redox homeostasis by scavenging ROS and inhibiting NF-κB signaling pathway. More importantly, the 16S ribosomal RNA gene sequencing results indicate that post-oral of ZnPBA@YCW can effectively regulate gut microbiota by enhancing the bacterial richness and diversity, significantly increasing the abundance of probiotics with anti-inflammatory phenotype while downgrading pathogenic E. coli to the same level as normal mice. Such a novel nanomedicine provides a new idea for efficient treating those ROS-mediated diseases accompanying with flora disorders.
Collapse
Affiliation(s)
- Zhangpeng Shi
- Nanotechnology and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Xiaohong Li
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Jufeng Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Zideng Dai
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Yefei Zhu
- Nanotechnology and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
| | - Tan Wu
- Nanotechnology and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China
| | - Qing Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Huanlong Qin
- Nanotechnology and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
| | - Yang Zhang
- Nanotechnology and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
- School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China
| | - Hangrong Chen
- Nanotechnology and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, PR China
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| |
Collapse
|
17
|
Zhao C, Yang X, Fan M, Tian L, Sun T, Sun C, Jiang T. The investigation on sialic acid-modified pectin nanoparticles loaded with oxymatrine for orally targeting and inhibiting the of ulcerative colitis. Colloids Surf B Biointerfaces 2024; 236:113809. [PMID: 38447446 DOI: 10.1016/j.colsurfb.2024.113809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024]
Abstract
The aim of the study was to develop an oral targeting drug delivery system (OTDDS) of oxymatrine (OMT) to effectively treat ulcerative colitis (UC). The OTDDS of OMT (OMT/SA-NPs) was constructed with OMT, pectin, Ca2+, chitosan (CS) and sialic acid (SA). The obtained particles were characterized in terms of particle size, zeta potential, morphology, drug loading, encapsulation efficiency, drug release and stability. The average size of OMT/SA-NPs was 255.0 nm with a zeta potential of -12.4 mV. The loading content and encapsulation efficiency of OMT/SA-NPs were 14.65% and 84.83%, respectively. The particle size of OMT/SA-NPs changed slightly in the gastrointestinal tract. The nanoparticles can delivery most of the drug to the colon region. In vitro cell experiments showed that the SA-NPs had excellent biocompatibility and anti-inflammation, and the uptake of SA-NPs by RAW 264.7 cells was time and concentration-dependent. The conjugated SA can help the internalization of NPs into target cells. In vivo experiments showed that OMT/SA-NPs had a superior anti-inflammation effect and the effect of reducing UC, which was attributed to the delivery most of OMT to the colonic lumen, the specific targeting and retention in colitis site and the combined anti-inflammation of OMT and NPs.
Collapse
Affiliation(s)
- Chunying Zhao
- Shenyang Pharmaceutical University, Benxi, Liaoning 110016, PR China
| | - Xin Yang
- Shenyang Pharmaceutical University, Benxi, Liaoning 110016, PR China
| | - Mengyao Fan
- Shenyang Pharmaceutical University, Benxi, Liaoning 110016, PR China
| | - Linan Tian
- Shenyang Pharmaceutical University, Benxi, Liaoning 110016, PR China
| | - Tongtong Sun
- Shenyang Pharmaceutical University, Benxi, Liaoning 110016, PR China
| | - Changshan Sun
- Shenyang Pharmaceutical University, Benxi, Liaoning 110016, PR China.
| | - Tongying Jiang
- Shenyang Pharmaceutical University, Benxi, Liaoning 110016, PR China.
| |
Collapse
|
18
|
Singh S. Antioxidant nanozymes as next-generation therapeutics to free radical-mediated inflammatory diseases: A comprehensive review. Int J Biol Macromol 2024; 260:129374. [PMID: 38242389 DOI: 10.1016/j.ijbiomac.2024.129374] [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: 09/12/2023] [Revised: 12/30/2023] [Accepted: 01/08/2024] [Indexed: 01/21/2024]
Abstract
Recent developments in exploring the biological enzyme mimicking properties in nanozymes have opened a separate avenue, which provides a suitable alternative to the natural antioxidants and enzymes. Due to high and tunable catalytic activity, low cost of synthesis, easy surface modification, and good biocompatibility, nanozymes have garnered significant research interest globally. Several inorganic nanomaterials have been investigated to exhibit catalytic activities of some of the key natural enzymes, including superoxide dismutase (SOD), catalase, glutathione peroxidase, peroxidase, and oxidase, etc. These nanozymes are used for diverse biomedical applications including therapeutics, imaging, and biosensing in various cells/tissues and animal models. In particular, inflammation-related diseases are closely associated with reactive oxygen and reactive nitrogen species, and therefore effective antioxidants could be excellent therapeutics due to their free radical scavenging ability. Although biological enzymes and other artificial antioxidants could perform well in scavenging the reactive oxygen and nitrogen species, however, suffer from several drawbacks such as the requirement of strict physiological conditions for enzymatic activity, limited stability in the environment beyond their optimum pH and temperature, and high cost of synthesis, purification, and storage make then unattractive for broad-spectrum applications. Therefore, this review systematically and comprehensively presents the free radical-mediated evolution of various inflammatory diseases (inflammatory bowel disease, mammary gland fibrosis, and inflammation, acute injury of the liver and kidney, mammary fibrosis, and cerebral ischemic stroke reperfusion) and their mitigation by various antioxidant nanozymes in the biological system. The mechanism of free radical scavenging by antioxidant nanozymes under in vitro and in vivo experimental models and catalytic efficiency comparison with corresponding natural enzymes has also been presented.
Collapse
Affiliation(s)
- Sanjay Singh
- National Institute of Animal Biotechnology (NIAB), Opposite Journalist Colony, Near Gowlidoddy, Extended Q-City Road, Gachibowli, Hyderabad 500032, Telangana, India.
| |
Collapse
|
19
|
Zhang DG, Pan YJ, Chen BQ, Lu XC, Xu QX, Wang P, Kankala RK, Jiang NN, Wang SB, Chen AZ. Protein-guided biomimetic nanomaterials: a versatile theranostic nanoplatform for biomedical applications. NANOSCALE 2024; 16:1633-1649. [PMID: 38168813 DOI: 10.1039/d3nr05495k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Over the years, bioinspired mineralization-based approaches have been applied to synthesize multifunctional organic-inorganic nanocomposites. These nanocomposites can address the growing demands of modern biomedical applications. Proteins, serving as vital biological templates, play a pivotal role in the nucleation and growth processes of various organic-inorganic nanocomposites. Protein-mineralized nanomaterials (PMNMs) have attracted significant interest from researchers due to their facile and convenient preparation, strong physiological activity, stability, impressive biocompatibility, and biodegradability. Nevertheless, few comprehensive reviews have expounded on the progress of these nanomaterials in biomedicine. This article systematically reviews the principles and strategies for constructing nanomaterials using protein-directed biomineralization and biomimetic mineralization techniques. Subsequently, we focus on their recent applications in the biomedical field, encompassing areas such as bioimaging, as well as anti-tumor, anti-bacterial, and anti-inflammatory therapies. Furthermore, we discuss the challenges encountered in practical applications of these materials and explore their potential in future applications. This review aspired to catalyze the continued development of these bioinspired nanomaterials in drug development and clinical diagnosis, ultimately contributing to the fields of precision medicine and translational medicine.
Collapse
Affiliation(s)
- Da-Gui Zhang
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Yu-Jing Pan
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Biao-Qi Chen
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Xiao-Chang Lu
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Qin-Xi Xu
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Pei Wang
- Jiangxi Provincial Key Laboratory of Oral Biomedicine, Jiangxi Province Clinical Research Center for Oral Diseases, School of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Ranjith Kumar Kankala
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Ni-Na Jiang
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Shi-Bin Wang
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Ai-Zheng Chen
- Fujian Provincial Key Laboratory of Biochemical Technology & Institute of Biomaterials and Tissue Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| |
Collapse
|
20
|
Wojcik-Grzybek D, Sliwowski Z, Kwiecien S, Ginter G, Surmiak M, Hubalewska-Mazgaj M, Chmura A, Wojcik A, Kosciolek T, Danielak A, Targosz A, Strzalka M, Szczyrk U, Ptak-Belowska A, Magierowski M, Bilski J, Brzozowski T. Alkaline Phosphatase Relieves Colitis in Obese Mice Subjected to Forced Exercise via Its Anti-Inflammatory and Intestinal Microbiota-Shaping Properties. Int J Mol Sci 2024; 25:703. [PMID: 38255781 PMCID: PMC10815191 DOI: 10.3390/ijms25020703] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 12/28/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Intestinal alkaline phosphatase (IAP) is an enzyme that plays a protective role in the gut. This study investigated the effect of IAP treatment on experimental colitis in mice subjected to forced exercise on a high-fat diet. C57BL/6 mice with TNBS colitis were fed a high-fat diet and subjected to forced treadmill exercise with or without IAP treatment. Disease activity, oxidative stress, inflammatory cytokines, and gut microbiota were assessed. Forced exercise exacerbated colitis in obese mice, as evidenced by increased disease activity index (DAI), oxidative stress markers, and proinflammatory adipokines and cytokines. IAP treatment significantly reduced these effects and promoted the expression of barrier proteins in the colonic mucosa. Additionally, IAP treatment altered the gut microbiota composition, favoring beneficial Verrucomicrobiota and reducing pathogenic Clostridia and Odoribacter. IAP treatment ameliorates the worsening effect of forced exercise on murine colitis by attenuating oxidative stress, downregulating proinflammatory biomarkers, and modulating the gut microbiota. IAP warrants further investigation as a potential therapeutic strategy for ulcerative colitis.
Collapse
Affiliation(s)
- Dagmara Wojcik-Grzybek
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland; (D.W.-G.); (S.K.); (G.G.)
| | - Zbigniew Sliwowski
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland; (D.W.-G.); (S.K.); (G.G.)
| | - Slawomir Kwiecien
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland; (D.W.-G.); (S.K.); (G.G.)
| | - Grzegorz Ginter
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland; (D.W.-G.); (S.K.); (G.G.)
| | - Marcin Surmiak
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, 31-008 Cracow, Poland
| | - Magdalena Hubalewska-Mazgaj
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland; (D.W.-G.); (S.K.); (G.G.)
| | - Anna Chmura
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland; (D.W.-G.); (S.K.); (G.G.)
| | - Adrianna Wojcik
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland; (D.W.-G.); (S.K.); (G.G.)
| | - Tomasz Kosciolek
- Malopolska Centre of Biotechnology, Jagiellonian University, 30-387 Cracow, Poland
| | - Aleksandra Danielak
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland; (D.W.-G.); (S.K.); (G.G.)
| | - Aneta Targosz
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland; (D.W.-G.); (S.K.); (G.G.)
| | - Malgorzata Strzalka
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland; (D.W.-G.); (S.K.); (G.G.)
| | - Urszula Szczyrk
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland; (D.W.-G.); (S.K.); (G.G.)
| | - Agata Ptak-Belowska
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland; (D.W.-G.); (S.K.); (G.G.)
| | - Marcin Magierowski
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland; (D.W.-G.); (S.K.); (G.G.)
| | - Jan Bilski
- Department of Biomechanics and Kinesiology, Chair of Biomedical Sciences, Faculty of Health Sciences, Jagiellonian University Medical College, 31-008 Cracow, Poland;
| | - Tomasz Brzozowski
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland; (D.W.-G.); (S.K.); (G.G.)
| |
Collapse
|
21
|
Li Q, Lin L, Zhang C, Zhang H, Ma Y, Qian H, Chen XL, Wang X. The progression of inorganic nanoparticles and natural products for inflammatory bowel disease. J Nanobiotechnology 2024; 22:17. [PMID: 38172992 PMCID: PMC10763270 DOI: 10.1186/s12951-023-02246-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/03/2023] [Indexed: 01/05/2024] Open
Abstract
There is a growing body of evidence indicating a close association between inflammatory bowel disease (IBD) and disrupted intestinal homeostasis. Excessive production of reactive oxygen species (ROS) and reactive nitrogen species (RNS), along with an increase in M1 proinflammatory macrophage infiltration during the activation of intestinal inflammation, plays a pivotal role in disrupting intestinal homeostasis in IBD. The overabundance of ROS/RNS can cause intestinal tissue damage and the disruption of crucial gut proteins, which ultimately compromises the integrity of the intestinal barrier. The proliferation of M1 macrophages contributes to an exaggerated immune response, further compromising the intestinal immune barrier. Currently, intestinal nanomaterials have gained widespread attention in the context of IBD due to their notable characteristics, including the ability to specifically target regions of interest, clear excess ROS/RNS, and mimic biological enzymes. In this review, we initially elucidated the gut microenvironment in IBD. Subsequently, we delineate therapeutic strategies involving two distinct types of nanomedicine, namely inorganic nanoparticles and natural product nanomaterials. Finally, we present a comprehensive overview of the promising prospects associated with the application of nanomedicine in future clinical settings for the treatment of IBD (graphic abstract). Different classes of nanomedicine are used to treat IBD. This review primarily elucidates the current etiology of inflammatory bowel disease and explores two prominent nanomaterial-based therapeutic approaches. First, it aims to eliminate excessive reactive oxygen species and reactive nitrogen species. Second, they focus on modulating the polarization of inflammatory macrophages and reducing the proportion of pro-inflammatory macrophages. Additionally, this article delves into the treatment of inflammatory bowel disease using inorganic metal nanomaterials and natural product nanomaterials.
Collapse
Affiliation(s)
- Qingrong Li
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Liting Lin
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Cong Zhang
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230026, People's Republic of China
| | - Hengguo Zhang
- Key Laboratory of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Yan Ma
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230026, People's Republic of China
| | - Haisheng Qian
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230026, People's Republic of China.
| | - Xu-Lin Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China.
| | - Xianwen Wang
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230026, People's Republic of China.
| |
Collapse
|
22
|
Zhang Z, Zhang Y, Zhang M, Yu C, Yang P, Xu M, Ling J, Wu Y, Zhu Z, Chen Y, Shi A, Liu X, Zhang J, Yu P, Zhang D. Food-derived peptides as novel therapeutic strategies for NLRP3 inflammasome-related diseases: a systematic review. Crit Rev Food Sci Nutr 2023:1-32. [PMID: 38153262 DOI: 10.1080/10408398.2023.2294164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3), a member of the nucleotide-binding domain (NOD) and leucine-rich repeat sequence (LRR) protein (NLR) family, plays an essential role in the inflammation initiation and inflammatory mediator secretion, and thus is also associated with many disease progressions. Food-derived bioactive peptides (FDBP) exhibit excellent anti-inflammatory activity in both in vivo and in vitro models. They are encrypted in plant, meat, and milk proteins and can be released under enzymatic hydrolysis or fermentation conditions, thereby hindering the progression of hyperuricemia, inflammatory bowel disease, chronic liver disease, neurological disorders, lung injury and periodontitis by inactivating the NLRP3. However, there is a lack of systematic review around FDBP, NLRP3, and NLRP3-related diseases. Therefore, this review summarized FDBP that exert inhibiting effects on NLRP3 inflammasome from different protein sources and detailed their preparation and purification methods. Additionally, this paper also compiled the possible inhibitory mechanisms of FDBP on NLRP3 inflammasomes and its regulatory role in NLRP3 inflammasome-related diseases. Finally, the progress of cutting-edge technologies, including nanoparticle, computer-aided screening strategy and recombinant DNA technology, in the acquisition or encapsulation of NLRP3 inhibitory FDBP was discussed. This review provides a scientific basis for understanding the anti-inflammatory mechanism of FDBP through the regulation of the NLRP3 inflammasome and also provides guidance for the development of therapeutic adjuvants or functional foods enriched with these FDBP.
Collapse
Affiliation(s)
- Ziqi Zhang
- The Second Clinical Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Jiangxi, China
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuan Zhang
- School of Public Health, Nanchang University, Jiangxi, China
| | - Meiying Zhang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Chenfeng Yu
- Huankui College, Nanchang University, Jiangxi, China
| | - Pingping Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Minxuan Xu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Jitao Ling
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Yuting Wu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Zicheng Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yixuan Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ao Shi
- School of Medicine, St. George University of London, London, UK
| | - Xiao Liu
- Cardiology Department, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Deju Zhang
- The Second Clinical Medical College, The Second Affiliated Hospital of Nanchang University, Nanchang University, Jiangxi, China
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Hong Kong
| |
Collapse
|
23
|
Yan J, Tang Z, Li Y, Wang H, Hsu JC, Shi M, Fu Z, Ji X, Cai W, Ni D, Qu J. Molybdenum Nanodots for Acute Lung Injury Therapy. ACS NANO 2023; 17:23872-23888. [PMID: 38084420 PMCID: PMC10760930 DOI: 10.1021/acsnano.3c08147] [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] [Indexed: 12/18/2023]
Abstract
Acute respiratory disease syndrome (ARDS) is a common critical disease with high morbidity and mortality rates, yet specific and effective treatments for it are currently lacking. ARDS was especially apparent and rampant during the COVID-19 pandemic. Excess reactive oxygen species (ROS) production and an uncontrolled inflammatory response play a critical role in the disease progression of ARDS. Herein, we developed molybdenum nanodots (MNDs) as a functional nanomaterial with ultrasmall size, good biocompatibility, and excellent ROS scavenging ability for the treatment of acute lung injury (ALI). MNDs, which were administered intratracheally, significantly ameliorated lung oxidative stress, inflammatory response, protein permeability, and histological severity in ALI mice without inducing any safety issues. Importantly, transcriptomics analysis indicated that MNDs protected lung tissues by inhibiting the activation of the Nod-like receptor protein 3 (NLRP3)-dependent pyroptotic pathway. This work presents a promising therapeutic agent for patients suffering from ARDS.
Collapse
Affiliation(s)
- Jiayang Yan
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Zhongmin Tang
- Departments of Radiology and Medical Physics, University of Wisconsin, Madison, Wisconsin 53705, United States
| | - Yanan Li
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Han Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jessica C Hsu
- Departments of Radiology and Medical Physics, University of Wisconsin, Madison, Wisconsin 53705, United States
| | - Mengmeng Shi
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| | - Zi Fu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiuru Ji
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin, Madison, Wisconsin 53705, United States
| | - Dalong Ni
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jieming Qu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai 200025, China
| |
Collapse
|
24
|
Min K, Sahu A, Jeon SH, Tae G. Emerging drug delivery systems with traditional routes - A roadmap to chronic inflammatory diseases. Adv Drug Deliv Rev 2023; 203:115119. [PMID: 37898338 DOI: 10.1016/j.addr.2023.115119] [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: 09/15/2022] [Revised: 07/17/2023] [Accepted: 10/23/2023] [Indexed: 10/30/2023]
Abstract
Inflammation is prevalent and inevitable in daily life but can generally be accommodated by the immune systems. However, incapable self-healing and persistent inflammation can progress to chronic inflammation, leading to prevalent or fatal chronic diseases. This review comprehensively covers the topic of emerging drug delivery systems (DDSs) for the treatment of chronic inflammatory diseases (CIDs). First, we introduce the basic biology of the chronic inflammatory process and provide an overview of the main CIDs of the major organs. Next, up-to-date information on various DDSs and the associated strategies for ensuring targeted delivery and stimuli-responsiveness applied to CIDs are discussed extensively. The implementation of traditional routes of drug administration to maximize their therapeutic effects against CIDs is then summarized. Finally, perspectives on future DDSs against CIDs are presented.
Collapse
Affiliation(s)
- Kiyoon Min
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Abhishek Sahu
- Department of Biotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Hajipur, 844102, India
| | - Sae Hyun Jeon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea.
| |
Collapse
|
25
|
Das T, Das S, Kumar P, C A B, Mandal D. Coal waste-derived synthesis of yellow oxidized graphene quantum dots with highly specific superoxide dismutase activity: characterization, kinetics, and biological studies. NANOSCALE 2023; 15:17861-17878. [PMID: 37885430 DOI: 10.1039/d3nr04259f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
The disintegration of coal-based precursors for the scalable production of nanozymes relies on the fate of solvothermal pyrolysis. Herein, we report a novel economic and scalable strategy to fabricate yellow luminescent graphene quantum dots (YGQDs) by remediating unburnt coal waste (CW). The YGQDs (size: 7-8 nm; M.W: 3157.9 Da) were produced using in situ "anion-radical" assisted bond cleavage in water (within 8 h; at 121 °C) with yields of ∼87%. The presence of exposed surface and edge groups, such as COOH, C-O-C, and O-H, as structural defects accounted for its high fluorescence with εmax ∼530 nm at pH 7. Besides, these defects also acted as radical stabilizers, demonstrating prominent anti-oxidative activity of ∼4.5-fold higher than standard ascorbic acid (AA). In addition, the YGQDs showed high biocompatibility towards mammalian cells, with 500 μM of treatment dose showing <15% cell death. The YGQDs demonstrated specific superoxide dismutase (SOD) activity wherein 15 μM YGQDs equalled the activity of 1-unit biological SOD (bSOD), measured using the pyrogallol assay. The Km for YGQDs was ∼10-fold higher than that for bSOD. However, the YGQDs retained their SOD activity in harsh conditions like high temperatures or denaturing reactions, where the activity of bSOD is completely lost. The binding affinity of YGQDs for superoxide ions, measured from isothermal calorimetry (ITC) studies, was only 10-fold lower than that of bSOD (Kd of 586 nM vs. 57.3 nM). Further, the pre-treatment of YGQDs (∼10-25 μM) increased the cell survivability to >75-90% in three cell lines during ROS-mediated cell death, with the highest survivability being shown for C6-cells. Next, the ROS-induced apoptosis in C6-cells (model for neurodegenerative diseases study), wherein YGQDs uptake was confirmed by confocal microscopy, showed ∼5-fold apoptosis alleviation with only 5 μM pretreatment. The YGQDs also restored the expression of pro-inflammatory Th1 cytokines (TNF-α, IFN-γ, IL-6) and anti-inflammatory Th2 cytokines (IL-10) to their basal levels, with a net >3-fold change observed. This further explains the molecular mechanism for the antioxidant property of YGQDs. The high specific SOD activity associated with YGQDs may provide the cheapest alternative source for producing large-scale SOD-based nanozymes that can treat various oxidative stress-linked disorders/diseases.
Collapse
Affiliation(s)
- Tushar Das
- Department of Chemistry, National Institute of Technology Patna, Bihar 800005, India.
| | - Subrata Das
- Department of Chemistry, National Institute of Technology Patna, Bihar 800005, India.
| | - Prakash Kumar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research Hajipur, Vaishali 844102, India.
| | - Betty C A
- Chemistry Division, Bhabha Atomic Research Centre, Mumbai 400085, India
- Homi Bhabha National Institute, Mumbai 400085, India
| | - Debabrata Mandal
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research Hajipur, Vaishali 844102, India.
| |
Collapse
|
26
|
Liu H, Zhang Y, Zhang M, Yu Z, Zhang M. Oral Administration of Platinum Nanoparticles with SOD/CAT Cascade Catalytic Activity to Alleviate Ulcerative Colitis. J Funct Biomater 2023; 14:548. [PMID: 37998117 PMCID: PMC10672654 DOI: 10.3390/jfb14110548] [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: 10/01/2023] [Revised: 11/04/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023] Open
Abstract
Ulcerative colitis (UC) is a refractory chronic inflammatory disease involving the colon and rectum, falling under the category of inflammatory bowel disease (IBD). The accumulation of reactive oxygen species (ROS) in local tissues has been identified as a crucial contributor to the escalation of inflammatory responses. Therefore, eliminating ROS in the inflamed colon is a promising approach to treating UC. Nanomaterials with intrinsic enzyme-like activities (nanozymes) have shown significant therapeutic potential in UC. In this study, we found that platinum nanoparticles (Pt NPs) exhibited remarkable superoxide dismutase (SOD) and catalase (CAT) cascade catalytic activities, as well as effective hydroxyl radical (•OH) scavenging ability. The in vitro experiments showed that Pt NPs could eliminate excessive ROS to protect cells against oxidative stress. In the colitis model, oral administration of Pt NPs (loaded in chitosan/alginate hydrogel) could significantly alleviate UC, including reducing the colon length, the damaged epithelium, and the infiltration of inflammatory cells. Without appreciable systemic toxicity, Pt NPs represent a novel therapeutic approach to UC and are expected to achieve long-term inflammatory remission.
Collapse
Affiliation(s)
- Hao Liu
- Second Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang 712046, China;
| | - Yujie Zhang
- School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (Y.Z.); (M.Z.)
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (Y.Z.); (M.Z.)
| | - Zhaoxiang Yu
- Department of General Surgery, The First Affiliated Hospital of Xi’an Medical University, Xi’an 710077, China
| | - Mingxin Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi’an Medical University, Xi’an 710077, China
| |
Collapse
|
27
|
Li F, Huang H, Zhao P, Jiang J, Ding X, Lu D, Ji L. Curculigoside mitigates dextran sulfate sodium‑induced colitis by activation of KEAP1‑NRF2 interaction to inhibit oxidative damage and autophagy of intestinal epithelium barrier. Int J Mol Med 2023; 52:107. [PMID: 37772380 PMCID: PMC10558217 DOI: 10.3892/ijmm.2023.5310] [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: 06/08/2023] [Accepted: 09/15/2023] [Indexed: 09/30/2023] Open
Abstract
Curculigoside (CUR), a primary active ingredient of Curculigo orchioides Gaertn, serves an important role in the intervention of numerous diseases, including ulcerative colitis, rheumatoid arthritis, myocardial ischemia, etc. However its specific mechanisms of therapy have not been fully elucidated. The aim of the present study was to elucidate the mechanisms underlying the anti‑oxidative stress and anti‑ulcerative colitis (UC) effects of CUR. Mouse model of dextran sulfate sodium (DSS)‑induced colitis, along with Caco2 and mouse intestine organoid in vitro models were used. The effect of CUR on mitigating the symptoms of chronic colitis was investigated. Through ELISA experiments, it was observed that CUR alleviated the inflammation status in mice with chronic colitis. This was evidenced by the downregulation of inflammatory cytokines such as TNF‑α and IL‑6 and ‑1β and decreased neutrophil infiltration along with downregulated myeloperoxidase activity. CUR helped in maintaining the barrier functions of intestinal epithelium. In vitro TNF‑α stimulation of organoids and H2O2 stimulation of Caco2 cells demonstrated the capabilities of CUR to rescue cells from oxidative stress. There was activation of Nrf2 both in vivo and in vitro, accompanied by enhanced autophagy. Mechanistic studies of cells and Nrf2 knockout mice demonstrated that Nrf2 served a pivotal role in inhibition of UC by curculigoside via interaction with Kelch‑like ECH‑associated protein 1 (Keap1). In vitro and in vivo experiments confirmed that CUR activated Nrf2 via Keap1/Nrf2 interaction, resulting in decreased oxidative stress and promoted autophagy. These findings demonstrated that CUR could effectively mitigate colitis and may have clinical application in UC therapy.
Collapse
Affiliation(s)
- Fang Li
- Department of Digestive System, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, Suzhou 215500, P.R. China
| | - Hua Huang
- Department of Digestive System, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, Suzhou 215500, P.R. China
| | - Ping Zhao
- Department of Anorectal Surgery, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, Suzhou 215500, P.R. China
| | - Jie Jiang
- Department of Anorectal Surgery, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, Suzhou 215500, P.R. China
| | - Xufeng Ding
- Department of Anorectal Surgery, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, Suzhou 215500, P.R. China
| | - Donxgue Lu
- Department of Clinical Nutrition, Academy of Health and Rehabilitation, Academy of Acupuncture and Tuina, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Lijiang Ji
- Department of Digestive System, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, Suzhou 215500, P.R. China
| |
Collapse
|
28
|
Fu W, Xu L, Chen Z, Kan L, Ma Y, Qian H, Wang W. Recent advances on emerging nanomaterials for diagnosis and treatment of inflammatory bowel disease. J Control Release 2023; 363:149-179. [PMID: 37741461 DOI: 10.1016/j.jconrel.2023.09.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic idiopathic inflammatory disorder that affects the entire gastrointestinal tract and is associated with an increased risk of colorectal cancer. Mainstream clinical testing methods are time-consuming, painful for patients, and insufficiently sensitive to detect early symptoms. Currently, there is no definitive cure for IBD, and frequent doses of medications with potentially severe side effects may affect patient response. In recent years, nanomaterials have demonstrated considerable potential for IBD management due to their diverse structures, composition, and physical and chemical properties. In this review, we provide an overview of the advances in nanomaterial-based diagnosis and treatment of IBD in recent five years. Multi-functional bio-nano platforms, including contrast agents, near-infrared (NIR) fluorescent probes, and bioactive substance detection agents have been developed for IBD diagnosis. Based on a series of pathogenic characteristics of IBD, the therapeutic strategies of antioxidant, anti-inflammatory, and intestinal microbiome regulation of IBD based on nanomaterials are systematically introduced. Finally, the future challenges and prospects in this field are presented to facilitate the development of diagnosis and treatment of IBD.
Collapse
Affiliation(s)
- Wanyue Fu
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, PR China; Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, Anhui 230012, China
| | - Lingling Xu
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, PR China; Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, Anhui 230012, China
| | - Zetong Chen
- School of Stomatology, Anhui Medical University, Hefei, Anhui 230032, PR China
| | - Lingling Kan
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, PR China; Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, Anhui 230012, China
| | - Yan Ma
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, PR China; Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, Anhui 230012, China.
| | - Haisheng Qian
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, PR China; Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, Anhui 230012, China.
| | - Wanni Wang
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, PR China; Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, Anhui 230012, China.
| |
Collapse
|
29
|
Arrè V, Scialpi R, Centonze M, Giannelli G, Scavo MP, Negro R. The 'speck'-tacular oversight of the NLRP3-pyroptosis pathway on gastrointestinal inflammatory diseases and tumorigenesis. J Biomed Sci 2023; 30:90. [PMID: 37891577 PMCID: PMC10612184 DOI: 10.1186/s12929-023-00983-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/23/2023] [Indexed: 10/29/2023] Open
Abstract
The NLRP3 inflammasome is an intracellular sensor and an essential component of the innate immune system involved in danger recognition. An important hallmark of inflammasome activation is the formation of a single supramolecular punctum, known as a speck, per cell, which is the site where the pro-inflammatory cytokines IL-1β and IL-18 are converted into their bioactive form. Speck also provides the platform for gasdermin D protein activation, whose N-terminus domain perforates the plasma membrane, allowing the release of mature cytokines alongside with a highly inflammatory form of cell death, namely pyroptosis. Although controlled NLRP3 inflammasome-pyroptosis pathway activation preserves mucosal immunity homeostasis and contributes to host defense, a prolonged trigger is deleterious and could lead, in genetically predisposed subjects, to the onset of inflammatory bowel disease, including Crohn's disease and ulcerative colitis, as well as to gastrointestinal cancer. Experimental evidence shows that the NLRP3 inflammasome has both protective and pathogenic abilities. In this review we highlight the impact of the NLRP3-pyroptosis axis on the pathophysiology of the gastrointestinal tract at molecular level, focusing on newly discovered features bearing pro- and anti-inflammatory and neoplastic activity, and on targeted therapies tested in preclinical and clinical trials.
Collapse
Affiliation(s)
- Valentina Arrè
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, 70013, Castellana Grotte, BA, Italy
| | - Rosanna Scialpi
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, 70013, Castellana Grotte, BA, Italy
| | - Matteo Centonze
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, 70013, Castellana Grotte, BA, Italy
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, 70013, Castellana Grotte, BA, Italy
| | - Maria Principia Scavo
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, 70013, Castellana Grotte, BA, Italy
| | - Roberto Negro
- Personalized Medicine Laboratory, National Institute of Gastroenterology "S. de Bellis", IRCCS Research Hospital, Via Turi 27, 70013, Castellana Grotte, BA, Italy.
| |
Collapse
|
30
|
Song X, Ding Q, Wei W, Zhang J, Sun R, Yin L, Liu S, Pu Y. Peptide-Functionalized Prussian Blue Nanomaterial for Antioxidant Stress and NIR Photothermal Therapy against Alzheimer's Disease. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206959. [PMID: 37322406 DOI: 10.1002/smll.202206959] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 05/18/2023] [Indexed: 06/17/2023]
Abstract
Excessive accumulations of reactive oxygen species (ROS) and amyloid-β (Aβ) protein are closely associated with the complex pathogenesis of Alzheimer's disease (AD). Therefore, approaches that synergistically exert elimination of ROS and dissociation of Aβ fibrils are effective therapeutic strategies for correcting the AD microenvironment. Herein, a novel near infrared (NIR) responsive Prussian blue-based nanomaterial (PBK NPs) is established with excellent antioxidant activity and photothermal effect. PBK NPs possess similar activities to multiple antioxidant enzymes, including superoxide dismutase, peroxidase, and catalase, which can eliminate massive ROS and relieve oxidative stress. Under the NIR irradiation, PBK NPs can generate local heat to disaggregate Aβ fibrils efficiently. By modifying CKLVFFAED peptide, PBK NPs display obvious targeting ability for blood-brain barrier penetration and Aβ binding. Furthermore, in vivo studies demonstrate that PBK NPs have outstanding ability to decompose Aβ plaques and alleviate neuroinflammation in AD mouse model. Overall, PBK NPs provide evident neuroprotection by reducing ROS levels and regulating Aβ deposition, and may accelerate the development of multifunctional nanomaterials for delaying the progression of AD.
Collapse
Affiliation(s)
- Xiaolei Song
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, P. R. China
| | - Qin Ding
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, P. R. China
| | - Wei Wei
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, P. R. China
- State Key Laboratory of Bioelectronics, Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, P. R. China
| | - Juan Zhang
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, P. R. China
| | - Rongli Sun
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, P. R. China
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, P. R. China
| | - Songqin Liu
- State Key Laboratory of Bioelectronics, Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, P. R. China
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, 210009, P. R. China
| |
Collapse
|
31
|
Liu C, Wang Q, Wu YL. Recent Advances in Nanozyme-Based Materials for Inflammatory Bowel Disease. Macromol Biosci 2023; 23:e2300157. [PMID: 37262405 DOI: 10.1002/mabi.202300157] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/07/2023] [Indexed: 06/03/2023]
Abstract
Inflammatory bowel disease (IBD) is a type of chronic inflammatory disorder that interferes with the patient's lifestyle and, in extreme situations, can be deadly. Fortunately, with the ever-deepening understanding of the pathological cause of IBD, recent studies using nanozyme-based materials have indicated the potential toward effective IBD treatment. In this review, the recent advancement of nanozymes for the treatment of enteritis is summarized from the perspectives of the structural design of nanozyme-based materials and therapeutic strategies, intending to serve as a reference to produce effective nanozymes for moderating inflammation in the future. Last but not least, the potential and current restrictions for using nanozymes in IBD will also be discussed. In short, this review may provide a guidance for the development of innovative enzyme-mimetic nanomaterials that offer a novel and efficient approach toward the effective treatment of IBD.
Collapse
Affiliation(s)
- Chuyi Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Qi Wang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
32
|
He H, Long M, Duan Y, Gu N. Prussian blue nanozymes: progress, challenges, and opportunities. NANOSCALE 2023; 15:12818-12839. [PMID: 37496423 DOI: 10.1039/d3nr01741a] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Prussian Blue Nanozymes (PBNZs) have emerged as highly efficient agents for reactive oxygen species (ROS) elimination, owing to their multiple enzyme-like properties encompassing catalase (CAT), peroxidase (POD), and superoxide dismutase (SOD) activities. As a functional nanomaterial mimicking enzyme, PBNZs not only surmount the limitations of natural enzymes, such as instability and high manufacturing costs, but also exhibit superior stability, tunable activity, low storage expenses, and remarkable reusability. Consequently, PBNZs have gained significant attention in diverse biomedical applications, including disease diagnosis and therapy. Over the past decade, propelled by advancements in catalysis science, biotechnology, computational science, and nanotechnology, PBNZs have witnessed remarkable progress in the exploration of their enzymatic activities, elucidation of catalytic mechanisms, and wide-ranging applications. This comprehensive review aims to provide a systematic overview of the discovery and catalytic mechanisms of PBNZ, along with the strategies employed to modulate their multiple enzyme-like activities. Furthermore, we extensively survey the recent advancements in utilizing PBNZs for scavenging ROS in various biomedical applications. Lastly, we analyze the existing challenges of translating PBNZs into therapeutic agents for clinical use and outline future research directions in this field. By presenting a comprehensive synopsis of the current state of knowledge, this review seeks to contribute to a deeper understanding of the immense potential of PBNZs as an innovative therapeutic agent in biomedicine.
Collapse
Affiliation(s)
- Hongliang He
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, 210009, People's Republic of China
| | - Mengmeng Long
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, 210009, People's Republic of China
| | - Yifan Duan
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, 210009, People's Republic of China
| | - Ning Gu
- School of Medicine, Nanjing University, Nanjing, 210093, People's Republic of China.
| |
Collapse
|
33
|
Aldrich JL, Panicker A, Ovalle R, Sharma B. Drug Delivery Strategies and Nanozyme Technologies to Overcome Limitations for Targeting Oxidative Stress in Osteoarthritis. Pharmaceuticals (Basel) 2023; 16:1044. [PMID: 37513955 PMCID: PMC10383173 DOI: 10.3390/ph16071044] [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: 04/21/2023] [Revised: 06/26/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Oxidative stress is an important, but elusive, therapeutic target for osteoarthritis (OA). Antioxidant strategies that target oxidative stress through the elimination of reactive oxygen species (ROS) have been widely evaluated for OA but are limited by the physiological characteristics of the joint. Current hallmarks in antioxidant treatment strategies include poor bioavailability, poor stability, and poor retention in the joint. For example, oral intake of exogenous antioxidants has limited access to the joint space, and intra-articular injections require frequent dosing to provide therapeutic effects. Advancements in ROS-scavenging nanomaterials, also known as nanozymes, leverage bioactive material properties to improve delivery and retention. Material properties of nanozymes can be tuned to overcome physiological barriers in the knee. However, the clinical application of these nanozymes is still limited, and studies to understand their utility in treating OA are still in their infancy. The objective of this review is to evaluate current antioxidant treatment strategies and the development of nanozymes as a potential alternative to conventional small molecules and enzymes.
Collapse
Affiliation(s)
| | | | | | - Blanka Sharma
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA; (J.L.A.)
| |
Collapse
|
34
|
Luo S, Gao J, Yuan H, Yang J, Fan Y, Wang L, Ouyang H, Fu Z. Mn Single-Atom Nanozymes with Superior Loading Capability and Superb Superoxide Dismutase-like Activity for Bioassay. Anal Chem 2023. [PMID: 37276189 DOI: 10.1021/acs.analchem.3c01623] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Single-atom nanozymes (SANs) with highly exposed active sites and remarkable catalytic activity have shown noteworthy practicability in heterogeneous catalysis-based bioassay. Nevertheless, most of them were reported with peroxidase-like activity and ordinary loading capability. It is still a challenge to prepare high-loading SANs with desirable superoxide dismutase (SOD)-like activity. In this work, Mn SAN was successfully confined in the frameworks of Prussian blue analogues formed on Ti3C2 MXene sheets with the assistance of massive surfactants, which show a superior loading efficiency of 13.5 wt % (typically <2.0 wt %). The prepared Mn SAN exhibits desirable superoxide radical anion elimination capability because of its SOD-like activity. Moreover, due to the wide-spectrum absorption behavior of the carriers, Mn SAN shows a synergistically quenching efficiency up to 98.89% on the emission of the reactive oxygen species-mediated chemiluminescent (CL) system. Inspired by these features, a CL quenching method was developed on a lateral flow test strip platform by utilizing Mn SAN as a signal quencher and acetamiprid as a model analyte. The method for detecting acetamiprid shows a detection range of 1.0-10,000 pg mL-1 and a limit of detection of 0.3 pg mL-1. Its accuracy has been validated by detecting acetamiprid in medicinal herbs with acceptable recoveries. This work opens an avenue for preparing SANs with a surfactant-assisted protocol and pioneers the study of SANs with SOD-like activity in bioassay.
Collapse
Affiliation(s)
- Shuai Luo
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Jiaqi Gao
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Hongwei Yuan
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Jin Yang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Yehan Fan
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Lin Wang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Hui Ouyang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zhifeng Fu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
35
|
Zhou C, Zhang L, Xu Z, Sun T, Gong M, Liu Y, Zhang D. Self-Propelled Ultrasmall AuNPs-Tannic Acid Hybrid Nanozyme with ROS-Scavenging and Anti-Inflammatory Activity for Drug-Induced Liver Injury Alleviation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206408. [PMID: 36759965 DOI: 10.1002/smll.202206408] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/05/2023] [Indexed: 05/11/2023]
Abstract
Developing nanomedicines with superior reactive oxygen species (ROS) scavenging capability has emerged as a promising strategy in treating ROS-related diseases, for example, drug-induced liver injury. However, designing nanoscavengers with the self-propelling ability to scavenge ROS actively remains challenging. Here, a self-propelled silica-supported ultrasmall gold nanoparticles-tannic acid hybrid nanozyme (SAuPTB) is designed that can effectively alleviate acetaminophen (APAP)-induced liver injury by scavenging excessive ROS and regulating inflammation. SAuPTB exhibits multienzyme activity and displays significantly enhanced diffusion under hydrogen peroxide (H2 O2 ). This in vitro research shows that SAuPTB can effectively eliminate ROS, increasing the viability of H2 O2 -stimulated cells and reducing the cytotoxicity of APAP/H2 O2 -treated AML12 cells. The in vivo studies show that SAuPTB can accumulate at inflammatory sites in mouse liver, resulting in the decrease of alanine aminotransferase, aspartate aminotransferase, and ROS, reduction in pro-inflammatory cytokines and chemokines, hence reduced hepatocyte necrosis, liver injury, and mortality. Furthermore, SAuPTB activates the nuclear erythroid 2-related factor 2 pathway to upregulate antioxidative genes and reduce oxidative stress. Finally, the liver shows decreased high mobility group box 1 and F4/80+ macrophages, suggesting an anti-inflammatory response. This work provides a novel design strategy of nanozymes for ROS-related disease treatment.
Collapse
Affiliation(s)
- Chunyu Zhou
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Liang Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Zhongsheng Xu
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Tao Sun
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Mingfu Gong
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Yun Liu
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| | - Dong Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
| |
Collapse
|
36
|
Zhang C, Hu Y, Yuan Y, Guo J, Li H, Li Q, Liu S. Liposome-embedded SOD attenuated DSS-induced ulcerative colitis in mice by ameliorating oxidative stress and intestinal barrier dysfunction. Food Funct 2023; 14:4392-4405. [PMID: 37092895 DOI: 10.1039/d2fo03312g] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Oxidative stress is generally considered inseparable from the development and exacerbation of ulcerative colitis (UC). Therefore, reducing oxidative stress has become a possible way to alleviate UC. In this study, the therapeutic effects of different doses of liposome-embedded superoxide dismutase (L-SOD) on mice with DSS-induced UC were systematically investigated. The results showed that L-SOD significantly attenuated the signs of colitis in mice, including colonic shortening, diarrhoea, bloody stools, and histopathological changes. L-SOD ameliorated DSS-induced oxidative damage, increased SOD, catalase (CAT), and glutathione (GSH) activities, and decreased malondialdehyde (MDA) levels. In addition, L-SOD ameliorated the inflammatory response by inhibiting the expression of myeloperoxidase (MPO) and pro-inflammatory cytokines and protected barrier function by promoting the expression of the tight junction proteins occludin and ZO-1 in the colon. Importantly, the results demonstrated a bell-shaped distribution of therapeutic effects relative to the administered dose, with an optimal dose of 150 000 U kg-1. These results indicate that L-SOD has great potential as an ingredient in functional foods for the prevention and mitigation of UC.
Collapse
Affiliation(s)
- Chi Zhang
- Institute of Biotechnology, Fuzhou University, Fuzhou 350108, China.
| | - Yujia Hu
- Institute of Biotechnology, Fuzhou University, Fuzhou 350108, China.
| | - Yi Yuan
- Institute of Biotechnology, Fuzhou University, Fuzhou 350108, China.
| | - Jingke Guo
- Institute of Biotechnology, Fuzhou University, Fuzhou 350108, China.
- Department of Food and Biological Engineering, Zhicheng College, Fuzhou University, Fuzhou 350002, China
| | - Henian Li
- Institute of Biotechnology, Fuzhou University, Fuzhou 350108, China.
| | - Qiaoling Li
- Institute of Biotechnology, Fuzhou University, Fuzhou 350108, China.
| | - Shutao Liu
- Institute of Biotechnology, Fuzhou University, Fuzhou 350108, China.
- Department of Food and Biological Engineering, Zhicheng College, Fuzhou University, Fuzhou 350002, China
| |
Collapse
|
37
|
Sun W, Chen Y, Wang L, Wang Z, Liu S, Zhang M, Liu Y, Li Q, Zhang H. Gram-scale preparation of quercetin supramolecular nanoribbons for intestinal inflammatory diseases by oral administration. Biomaterials 2023; 295:122039. [PMID: 36791522 DOI: 10.1016/j.biomaterials.2023.122039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 01/20/2023] [Accepted: 02/08/2023] [Indexed: 02/11/2023]
Abstract
Gastrointestinal (GI) tract, which possesses the largest surface area of mucosa in the body, is easily suffered from inflammatory damages under the exposure of external stimulations. Excessive reactive oxygen species (ROS) production and continuous oxidative stress in intestines can elicit local mucosal injury, accelerate mucosal ulceration, and amplify the inflammatory response. Thereby, antioxidant therapy is a potential strategy against intestinal inflammatory diseases. Herein, we demonstrate the gram-scale preparation of quercetin supramolecular nanoribbons (SNRs) by using free quercetin molecules as the sole building block for preventing and treating intestinal inflammatory diseases. Unlike current clinical medicines, which mainly confront with poor response and severe adverse effects via bloodstream delivery, our quercetin SNRs possess an excellent antioxidant activity in the harsh environments of GI tract, a relative long retention time in GI tract, an admirable metabolism in GI tract without burdening other organs, and a specific adhesion to the inflamed intestinal epithelium via electrostatic interactions. These advantages strongly guarantee the applications of quercetin SNRs as oral medicines for intestinal inflammatory diseases. After establishing the models of intestinal inflammatory diseases caused by irradiation and drug stimulations, our quercetin SNRs exhibit the promising protective and therapeutic effects for radiation-induced acute enteritis and dextran sulfate sodium (DSS)-induced acute colitis. Because the super easy and fast preparation procedure and the nearly 100% loading capacity of quercetin SNRs, the current work provides a supramolecular nanomedicine with great clinical translation potential against intestinal inflammatory diseases.
Collapse
Affiliation(s)
- Wei Sun
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Yingxuan Chen
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130021, PR China
| | - Liang Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Ze Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Shuwei Liu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130021, PR China
| | - Mengzhu Zhang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130021, PR China
| | - Yi Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China.
| | - Quanshun Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130021, PR China.
| | - Hao Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China; Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130021, PR China; Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou, 450001, PR China
| |
Collapse
|
38
|
Zhao J, Guo F, Hou L, Zhao Y, Sun P. Electron transfer-based antioxidant nanozymes: Emerging therapeutics for inflammatory diseases. J Control Release 2023; 355:273-291. [PMID: 36731800 DOI: 10.1016/j.jconrel.2023.01.068] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/27/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023]
Abstract
Inflammatory diseases are usually featured with relatively high level of reactive oxygen species (ROS). The excess ROS facilitate the polarization of microphages into proinflammatory M1 phenotype, and cause DNA damage, protein carbonylation, and lipid peroxidation, resulting in further deterioration of inflammatory diseases. Therefore, alleviating oxidative stress by ROS scavenging has been an effective strategy for reversing inflammation. Inspired by the natural antioxidant enzymes, electron transfer-based artificial antioxidant nanozymes have been emerging therapeutics for the treatment of inflammatory diseases. The present review starts with the basic knowledge of ROS and diseases, followed by summarizing the possible active centers for the preparation of antioxidant nanozymes. The strategies for the design of antioxidant nanozymes on the purpose of higher catalytic activity are provided, and the applications of the developed antioxidant nanozymes on the therapy of inflammatory diseases are discussed. A perspective is included for the design and applications of artificial antioxidant nanozymes in biomedicine as well.
Collapse
Affiliation(s)
- Jingnan Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Fanfan Guo
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Lin Hou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, Zhengzhou University, Zhengzhou 450001, PR China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China
| | - Yongxing Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, Zhengzhou University, Zhengzhou 450001, PR China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou 450052, PR China
| | - Pengchao Sun
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, PR China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, Zhengzhou University, Zhengzhou 450001, PR China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, PR China.
| |
Collapse
|
39
|
Li D, Dai D, Xiong G, Lan S, Zhang C. Metal-Based Nanozymes with Multienzyme-Like Activities as Therapeutic Candidates: Applications, Mechanisms, and Optimization Strategy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205870. [PMID: 36513384 DOI: 10.1002/smll.202205870] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/21/2022] [Indexed: 06/17/2023]
Abstract
Most nanozymes in development for medical applications only exhibit single-enzyme-like activity, and are thus limited by insufficient catalytic activity and dysfunctionality in complex pathological microenvironments. To overcome the impediments of limited substrate availabilities and concentrations, some metal-based nanozymes may mimic two or more activities of natural enzymes to catalyze cascade reactions or to catalyze multiple substrates simultaneously, thereby amplifying catalysis. Metal-based nanozymes with multienzyme-like activities (MNMs) may adapt to dissimilar catalytic conditions to exert different enzyme-like effects. These multienzyme-like activities can synergize to realize "self-provision of the substrate," in which upstream catalysts produce substrates for downstream catalytic reactions to overcome the limitation of insufficient substrates in the microenvironment. Consequently, MNMs exert more potent antitumor, antibacterial, and anti-inflammatory effects in preclinical models. This review summarizes the cellular effects and underlying mechanisms of MNMs. Their potential medical utility and optimization strategy from the perspective of clinical requirements are also discussed, with the aim to provide a theoretical reference for the design, development, and therapeutic application of their catalytic effects.
Collapse
Affiliation(s)
- Dan Li
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Danni Dai
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Gege Xiong
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Shuquan Lan
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Chao Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| |
Collapse
|
40
|
Palaniyandi T, B K, Prabhakaran P, Viswanathan S, Rahaman Abdul Wahab M, Natarajan S, Kumar Kaliya Moorthy S, Kumarasamy S. Nanosensors for the diagnosis and therapy of neurodegenerative disorders and inflammatory bowel disease. Acta Histochem 2023; 125:151997. [PMID: 36682145 DOI: 10.1016/j.acthis.2023.151997] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/27/2022] [Accepted: 01/05/2023] [Indexed: 01/21/2023]
Abstract
One of the areas of science which has immensely advanced in the recent years is nanotechnology. This area broadly revolves around matter at scales between 1 and 100 nm, where peculiar phenomena make way for cutting-edge applications. Today, nanotechnology has a daily impact on human life with numerous and varied possible advantages. Nanosensors are one of the products of nanotechnology and any sensor that uses nanoscale phenomena qualifies to be known as a nanosensor. Nanosensors have proven very useful in a number of sectors including medical applications, food quality analysis and agricultural controlling process, etc. One of the major human healthcare applications of nanosensors is for disease diagnosis. With the aid of nanosensors, numerous neurodegenerative disorders and inflammatory diseases are commonly identified and treated of late. Alzheimer's disease (AD) and inflammatory bowel disease fall under the categories of neurodegenerative illnesses and inflammatory diseases. There are more than 20 million cases of (AD) making it the most prevalent neurological condition globally and "inflammatory bowel disease" (IBD) refers to a variety of conditions that cause persistent inflammation of the digestive tract. Here we present a comprehensive account on the utility of nanosensors for the diagnosis and treatment of (AD) and (IBD).
Collapse
Affiliation(s)
- Thirunavukkarsu Palaniyandi
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India; Department of Anatomy, Biomedical Reseach Unit and Laboratory Animal Centre, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| | - Kanagavalli B
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India
| | - Pranav Prabhakaran
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India
| | - Sandhiya Viswanathan
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India
| | - Mugip Rahaman Abdul Wahab
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India
| | - Sudhakar Natarajan
- ICMR - National Institute for Research in Tuberculosis (NIRT), Chetpet, Chennai, Tamil Nadu, India
| | - Senthil Kumar Kaliya Moorthy
- Department of electronics and communication engineering, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India
| | - Saravanan Kumarasamy
- Department of electrical and electronics engineering, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India
| |
Collapse
|
41
|
NIR responsive nanoenzymes via photothermal ablation and hypoxia reversal to potentiate the STING-dependent innate antitumor immunity. Mater Today Bio 2023; 19:100566. [PMID: 36816600 PMCID: PMC9932208 DOI: 10.1016/j.mtbio.2023.100566] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 01/30/2023] Open
Abstract
Despite advances in combined photothermal/immunotherapy of tumor, the therapeutic effect has been impaired due to hypoxic microenvironment and inadequate immune activation. Manganese ions directly activated the stimulator of interferon genes (STING) pathway and induced innate antitumor immunity. Herein, a near infrared light (NIR)-responsive nanoenzyme (PB-Mn/OVA NE) was constructed by doping manganese into the ovalbumin (OVA)-templated Prussian blue (PB) nanoparticles. The resultant PB-Mn/OVA NEs exhibited favorable catalase activity to produce oxygen, which was conducive to alleviate the tumor hypoxic microenvironment. Under 808 nm NIR irradiation, the PB-Mn/OVA NEs with outstanding photothermal conversion efficiency of 30% significantly destroyed tumor cells by inducing immunogenic cell death (ICD). Impressively, the PB-Mn/OVA NEs could activate the cGAS-STING pathway to promote the maturation and the antigen cross-presentation ability of dendritic cells (DCs), which further activated cytotoxic T lymphocytes and memory T lymphocytes. Overall, this work presents a powerful nanoenzyme formula to integrate photothermal ablation and hypoxic reversal for triggering robust innate and adaptive antitumor immune response.
Collapse
|
42
|
Li M, Liu J, Shi L, Zhou C, Zou M, Fu D, Yuan Y, Yao C, Zhang L, Qin S, Liu M, Cheng Q, Wang Z, Wang L. Gold nanoparticles-embedded ceria with enhanced antioxidant activities for treating inflammatory bowel disease. Bioact Mater 2023; 25:95-106. [PMID: 36789001 PMCID: PMC9900456 DOI: 10.1016/j.bioactmat.2023.01.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/16/2023] [Accepted: 01/16/2023] [Indexed: 01/27/2023] Open
Abstract
The excessive reactive oxygen species (ROS) is a hallmark associated with the initiation and progression of inflammatory bowel disease (IBD), which execrably form a vicious cycle of ROS and inflammation to continually promote disease progression. Here, the gold nanoparticles-embedded ceria nanoparticles (Au/CeO2) with enhanced antioxidant activities are designed to block this cycle reaction for treating IBD by scavenging overproduced ROS. The Au/CeO2 with core-shell and porous structure exhibits significantly higher enzymatic catalytic activities compared with commercial ceria nanoparticles, likely due to the effective exposure of catalytic sites, higher content of Ce (III) and oxygen vacancy, and accelerated reduction from Ce (IV) to Ce (III). Being coated with negatively-charged hyaluronic acid, the Au/CeO2@HA facilitates accumulation in inflamed colon tissues via oral administration, reduces pro-inflammatory cytokines, and effectively alleviates colon injury in colitis mice. Overall, the Au/CeO2@HA with good biocompatibility is a promising nano-therapeutic for treating IBD.
Collapse
Affiliation(s)
- Mingyi Li
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Hubei Key Laboratory of Regenerative Medicine and Multi-disciplinary Translational Research, Wuhan, 430022, China
| | - Jia Liu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Hubei Key Laboratory of Regenerative Medicine and Multi-disciplinary Translational Research, Wuhan, 430022, China
| | - Lin Shi
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Hubei Key Laboratory of Regenerative Medicine and Multi-disciplinary Translational Research, Wuhan, 430022, China
| | - Cheng Zhou
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430022, China,Hubei Key Laboratory of Regenerative Medicine and Multi-disciplinary Translational Research, Wuhan, 430022, China
| | - Meizhen Zou
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Hubei Key Laboratory of Regenerative Medicine and Multi-disciplinary Translational Research, Wuhan, 430022, China
| | - Daan Fu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430022, China
| | - Ye Yuan
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430022, China
| | - Chundong Yao
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Hubei Key Laboratory of Regenerative Medicine and Multi-disciplinary Translational Research, Wuhan, 430022, China
| | - Lifang Zhang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Sumei Qin
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Hubei Key Laboratory of Regenerative Medicine and Multi-disciplinary Translational Research, Wuhan, 430022, China
| | - Miaodeng Liu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Hubei Key Laboratory of Regenerative Medicine and Multi-disciplinary Translational Research, Wuhan, 430022, China
| | - Qian Cheng
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Hubei Key Laboratory of Regenerative Medicine and Multi-disciplinary Translational Research, Wuhan, 430022, China
| | - Zheng Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430022, China,Hubei Key Laboratory of Regenerative Medicine and Multi-disciplinary Translational Research, Wuhan, 430022, China,Corresponding author. Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Lin Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China,Hubei Key Laboratory of Regenerative Medicine and Multi-disciplinary Translational Research, Wuhan, 430022, China,Corresponding author. Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
43
|
Liu Y, Yi Y, Zhong C, Ma Z, Wang H, Dong X, Yu F, Li J, Chen Q, Lin C, Li X. Advanced bioactive nanomaterials for diagnosis and treatment of major chronic diseases. Front Mol Biosci 2023; 10:1121429. [PMID: 36776741 PMCID: PMC9909026 DOI: 10.3389/fmolb.2023.1121429] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 01/17/2023] [Indexed: 01/27/2023] Open
Abstract
With the rapid innovation of nanoscience and technology, nanomaterials have also been deeply applied in the medical and health industry and become one of the innovative methods to treat many diseases. In recent years, bioactive nanomaterials have attracted extensive attention and have made some progress in the treatment of some major chronic diseases, such as nervous system diseases and various malignant tumors. Bioactive nanomaterials depend on their physical and chemical properties (crystal structure, surface charge, surface functional groups, morphology, and size, etc.) and direct produce biological activity and play to the role of the treatment of diseases, compared with the traditional nanometer pharmaceutical preparations, biological active nano materials don't exert effects through drug release, way more directly, also is expected to be more effective for the treatment of diseases. However, further studies are needed in the evaluation of biological effects, fate in vivo, structure-activity relationship and clinical transformation of bionanomaterials. Based on the latest research reports, this paper reviews the application of bioactive nanomaterials in the diagnosis and treatment of major chronic diseases and analyzes the technical challenges and key scientific issues faced by bioactive nanomaterials in the diagnosis and treatment of diseases, to provide suggestions for the future development of this field.
Collapse
Affiliation(s)
- Yongfei Liu
- Department of Urology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| | - Yi Yi
- Department of Urology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China,*Correspondence: Yi Yi,
| | - Chengqian Zhong
- Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| | - Zecong Ma
- Department of Urology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| | - Haifeng Wang
- Department of Urology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| | - Xingmo Dong
- Department of Urology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| | - Feng Yu
- Department of Urology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| | - Jing Li
- Department of Urology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| | - Qinqi Chen
- Department of Urology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| | - Chaolu Lin
- Department of Urology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| | - Xiaohong Li
- Longyan First Hospital Affiliated to Fujian Medical University, Longyan, China
| |
Collapse
|
44
|
Chen C, Wu H, Li Q, Liu M, Yin F, Wu M, Wei X, Wang H, Zha Z, Wang F. Manganese Prussian blue nanozymes with antioxidant capacity prevent acetaminophen-induced acute liver injury. Biomater Sci 2023; 11:2348-2358. [PMID: 36722889 DOI: 10.1039/d2bm01968j] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
As one of the leading cases of acute liver failure triggered by excessive Acetaminophen (APAP), breakdown of the antioxidant system, inflammatory response, and inescapable apoptosis following overaccumulation of reactive oxygen species (ROS) play crucial roles in the mechanisms of APAP-induced liver injury (AILI). Therefore, cutting off ROS overproduction at the source is considered promising. Here, manganese Prussian blue nanozymes (MPBZs) with superior antioxidant enzyme-like activity are prepared as an effective strategy for hepatocyte protection, in which MPBZs accumulated in the liver show anti-oxidation properties by scavenging superfluous ROS. Importantly, in addition to alleviating oxidative stress, bioactive MPBZs with abundant variable valence states as a natural antioxidant enzymes mediated the responses of multi-biological signaling pathways in vitro and in vivo, including Nrf2-Keap1, NF-κB, and mitochondrial-induced apoptosis signaling pathways, enhancing tolerance for imminent AILI. Taking nanomedicine, hepatology, and catalytic chemistry into consideration, the revealed superior performance of AILI prevention suggests that MPBZ-based nano-detoxification therapy may offer an effective alternative against AILI.
Collapse
Affiliation(s)
- Chongqing Chen
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Haitao Wu
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China. .,School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China.
| | - Qianhui Li
- China Guangdong Provincial Key Laboratory of Digestive Cancer Research and The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Menghua Liu
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Fan Yin
- School of Pharmacy, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Miaomiao Wu
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| | - Xiaoli Wei
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China.
| | - Zhengbao Zha
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China.
| | - Fei Wang
- China Guangdong Provincial Key Laboratory of Digestive Cancer Research and The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| |
Collapse
|
45
|
Mohapatra A, Park IK. Recent Advances in ROS-Scavenging Metallic Nanozymes for Anti-Inflammatory Diseases: A Review. Chonnam Med J 2023; 59:13-23. [PMID: 36794252 PMCID: PMC9900225 DOI: 10.4068/cmj.2023.59.1.13] [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: 12/03/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 02/01/2023] Open
Abstract
Oxidative stress and dysregulated inflammatory responses are the hallmarks of inflammatory disorders, which are key contributors to high mortality rates and impose a substantial economic burden on society. Reactive oxygen species (ROS) are vital signaling molecules that promote the development of inflammatory disorders. The existing mainstream therapeutic approaches, including steroid and non-steroidal anti-inflammatory drugs, and proinflammatory cytokine inhibitors with anti-leucocyte inhibitors, are not efficient at curing the adverse effects of severe inflammation. Moreover, they have serious side effects. Metallic nanozymes (MNZs) mimic the endogenous enzymatic process and are promising candidates for the treatment of ROS-associated inflammatory disorders. Owing to the existing level of development of these metallic nanozymes, they are efficient at scavenging excess ROS and can resolve the drawbacks of traditional therapies. This review summarizes the context of ROS during inflammation and provides an overview of recent advances in metallic nanozymes as therapeutic agents. Furthermore, the challenges associated with MNZs and an outline for future to promote the clinical translation of MNZs are discussed. Our review of this expanding multidisciplinary field will benefit the current research and clinical application of metallic-nanozyme-based ROS scavenging in inflammatory disease treatment.
Collapse
Affiliation(s)
- Adityanarayan Mohapatra
- Department of Biomedical Science, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju, Korea
| | - In-Kyu Park
- Department of Biomedical Science, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
46
|
Zhang X, Zhao X, Hua Z, Xing S, Li J, Fei S, Tan M. ROS-triggered self-disintegrating and pH-responsive astaxanthin nanoparticles for regulating the intestinal barrier and colitis. Biomaterials 2023; 292:121937. [PMID: 36495803 DOI: 10.1016/j.biomaterials.2022.121937] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 11/12/2022] [Accepted: 11/30/2022] [Indexed: 12/03/2022]
Abstract
Smart delivery systems with stimuli-responsive capability are able to improve the bioaccessibility through increasing the solubility, physicochemical stability and biocompatibility of bioactive compounds. In this study, the astaxanthin nanoparticles with reactive oxygen species (ROS) and pH dual-response function were design and constructed using poly (propylene sulfide) covalently modified sodium alginate as carriers based on ultrasonic assisted self-assembly strategy. Atomic force microscope and scanning electron microscope analysis showed that the nanoparticles were spherical in shape with a size of around 260 nm. Meanwhile, the astaxanthin nanoparticles showed both pH and ROS stimuli-responsive release characteristics. In vitro cell experiments showed that astaxanthin nanoparticles significantly inhibited the production of ROS and mitochondrial depolarization induced by oxidative stress. In vivo colitis experiment of mice revealed that astaxanthin nanoparticles could significantly relieve colitis, protect the integrity of colon tissue and restore the expression of tight junction proteins ZO-1 and occludin. The abundance of Lactobacillus and Lachnospiraceae, and the ratio of Firmicutes/Bacteroidota of gut microbiota were significantly improved after intervention of the stimuli-responsive astaxanthin nanoparticles. This work provided a simple strategy for constructing ROS/pH dual response delivery system, which provided an experimental basis for improving the oral bioavailability of hydrophobic active compounds.
Collapse
Affiliation(s)
- Xuedi Zhang
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, Liaoning, China
| | - Xue Zhao
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, Liaoning, China
| | - Zheng Hua
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, Liaoning, China
| | - Shanghua Xing
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, Liaoning, China
| | - Jiaxuan Li
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, Liaoning, China
| | - Siyuan Fei
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, Liaoning, China
| | - Mingqian Tan
- Academy of Food Interdisciplinary Science, Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, Liaoning, China.
| |
Collapse
|
47
|
Hederacoside C ameliorates colitis via restoring impaired intestinal barrier through moderating S100A9/MAPK and neutrophil recruitment inactivation. Acta Pharmacol Sin 2023; 44:105-119. [PMID: 35732707 PMCID: PMC9214479 DOI: 10.1038/s41401-022-00933-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 06/02/2022] [Indexed: 01/18/2023] Open
Abstract
Hederacoside C (HSC) has attracted much attention as a novel modulator of inflammation, but its anti-inflammatory mechanism remains elusive. In the present study, we investigated how HSC attenuated intestinal inflammation in vivo and in vitro. HSC injection significantly alleviated TNBS-induced colitis by inhibiting pro-inflammatory cytokine production and colonic epithelial cell apoptosis, and partially restored colonic epithelial cell proliferation. The therapeutic effect of HSC injection was comparable to that of oral administration of mesalazine (200 mg·kg-1·d-1, i.g.). In LPS-stimulated human intestinal epithelial Caco-2 cells, pretreatment with HSC (0.1, 1, 10 μM) significantly inhibited activation of MAPK/NF-κB and its downstream signaling pathways. Pretreatment with HSC prevented LPS-induced TLR4 dimerization and MyD88 recruitment in vitro. Quantitative proteomic analysis revealed that HSC injection regulated 18 proteins in the colon samples, mainly clustered in neutrophil degranulation. Among them, S100A9 involved in the degranulation of neutrophils was one of the most significantly down-regulated proteins. HSC suppressed the expression of S100A9 and its downstream genes including TLR4, MAPK, and NF-κB axes in colon. In Caco-2 cells, recombinant S100A9 protein activated the MAPK/NF-κB signaling pathway and induced inflammation, which were ameliorated by pretreatment with HSC. Notably, HSC attenuated neutrophil recruitment and degranulation as well as S100A9 release in vitro and in vivo. In addition, HSC promoted the expression of tight junction proteins and repaired the epithelial barrier via inhibiting S100A9. Our results verify that HSC ameliorates colitis via restoring impaired intestinal barrier through moderating S100A9/MAPK and neutrophil recruitment inactivation, suggesting that HSC is a promising therapeutic candidate for colitis.
Collapse
|
48
|
Recent advances in multi-configurable nanomaterials for improved chemodynamic therapy. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
49
|
Chao D, Dong Q, Yu Z, Qi D, Li M, Xu L, Liu L, Fang Y, Dong S. Specific Nanodrug for Diabetic Chronic Wounds Based on Antioxidase-Mimicking MOF-818 Nanozymes. J Am Chem Soc 2022; 144:23438-23447. [PMID: 36512736 DOI: 10.1021/jacs.2c09663] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Chronic wound is a common complication for diabetic patients, which entails substantial inconvenience, persistent pain, and significant economic burden to patients. However, current clinical treatments for diabetic chronic wounds remain unsatisfactory. A prolonged but ineffective inflammation phase in chronic wounds is the primary difference between diabetic chronic wounds and normal wounds. Herein, we present an effective antioxidative system (MOF/Gel) for chronic wound healing of diabetic rats through integrating a metal organic framework (MOF) nanozyme with antioxidant enzyme-like activity with a hydrogel (Gel). MOF/Gel can continuously scavenge reactive oxygen species to modulate the oxidative stress microenvironment in diabetic chronic wounds, which leads to a natural transition from the inflammation phase to the proliferation phase. Impressively, the efficacy of one-time-applied MOF/Gel was comparable to that of the human epidermal growth factor Gel, a widely used clinical drug for various wound treatments. Such an effective, safe, and convenient MOF/Gel system can meet complex clinical demands.
Collapse
Affiliation(s)
- Daiyong Chao
- College of Chemistry, Jilin University, Changchun, Jilin 130012, China.,State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Qing Dong
- College of Chemistry, Jilin University, Changchun, Jilin 130012, China.,State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Zhixuan Yu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.,University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Desheng Qi
- College of Chemistry, Jilin University, Changchun, Jilin 130012, China.,State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Minghua Li
- College of Chemistry, Jilin University, Changchun, Jilin 130012, China.,State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Lili Xu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.,University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Ling Liu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Youxing Fang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Shaojun Dong
- College of Chemistry, Jilin University, Changchun, Jilin 130012, China.,State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.,University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
50
|
Lu Y, Cao C, Pan X, Liu Y, Cui D. Structure design mechanisms and inflammatory disease applications of nanozymes. NANOSCALE 2022; 15:14-40. [PMID: 36472125 DOI: 10.1039/d2nr05276h] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Nanozymes are artificial enzymes with high catalytic activity, low cost, and good biocompatibility, and have received ever-increasing attention in recent years. Various inorganic and organic nanoparticles have been found to exhibit enzyme-like activities and are used as nanozymes for diverse biomedical applications ranging from tumor imaging and therapeutics to detection. However, their further clinical applications are hindered by the potential toxicity and long-term retention of nanomaterials in vivo. Clarifying the catalytic mechanism of nanozymes and identifying the key factors responsible for their behavior can guide the design of nanozyme structure, enlighten the ways to improve their enzyme-like activities, and minimize the dosage of nanozymes, leading to reduced toxicity to the human body for a real biomedical application prospect. In particular, inflammation occurring in numerous diseases is closely related to reactive oxygen species, and the active oxygen scavenging ability of nanozymes potentially exerts excellent therapeutic effects on inflammatory diseases. In this review, we systematically summarize the structure-activity relationship of nanozymes, including regulation strategies for size and morphology, surface structure, and composition. Based on the structure-activity mechanisms, a series of chemically designed nanozymes developed to target various inflammatory diseases are briefly summarized.
Collapse
Affiliation(s)
- Yi Lu
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China.
| | - Cheng Cao
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China.
| | - Xinni Pan
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanlei Liu
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China.
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Research Centre for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China.
- National Engineering Center for Nanotechnology, Shanghai 200240, People's Republic of China.
| |
Collapse
|