1
|
Mei LN, Wang ZJ, Duan Y, Shen JS, Ye HB, Zhu YY, Luo XD. 4-Hydroxyboesenbergin B of Alpinia japonica protected gastrointestinal tract by inhibiting vancomycin-resistant enterococcus and balancing intestinal microbiota. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119737. [PMID: 40179999 DOI: 10.1016/j.jep.2025.119737] [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: 12/29/2024] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/05/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alpinia japonica, a traditional herb utilized in Miao medicine in southwestern China, has been employed to alleviate symptoms such as stomachache, diarrhea, and abdominal pain, some of these symptoms may be associated with bacterial infections of the gastrointestinal tract. AIM OF THE STUDY To explore antimicrobial compounds related to traditional uses of A. japonica and its potential pathway in vitro and in vivo. MATERIALS AND METHODS Bioactive components of A. japonica were isolated by bioguide separation method. The antibacterial bioactivity of 4-hydroxyboesenbergin B (4-HB) was evaluated by time-kill curve and drug resistance induction. The pathway of 4-HB against VRE was investigated through network pharmacological analysis and validated by in vitro experiments and RT-qPCR assays. Moreover, a mouse gastrointestinal tract model was established to validate the antibacterial bioactivity of 4-HB in vivo. RESULTS 4-HB from A. japonica inhibited VRE (MIC = 16 μg/mL), rapidly killed the bacteria within 4 h at the 4 MIC concentration and exhibited low susceptibility to drug resistance. 4-HB specifically targeted VRE biofilms by down-regulating the expression of AtlA, SgrA, GelE, and Ace. As a result, 4-HB diminished the adhesion and aggregation ability of VRE, reduced the extracellular matrix content, disrupted biofilm structure and morphology, thereby reducing VRE resistance and virulence. Additionally, 4-HB significantly reduced VRE colonization, enhanced intestinal microbiota diversity, and promoted the restoration of intestinal microbiota balance in vivo. Notably, 4-HB enhanced the abundance of beneficial bacteria genera, such as Lactobacillus and Limosilactobacillus. CONCLUSIONS 4-HB has a significant ability to destroy VRE biofilms and balance intestinal microbiota, which might be responsible for the traditional use of A. japonica partly.
Collapse
Affiliation(s)
- Li-Na Mei
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China
| | - Zhao-Jie Wang
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China
| | - Yu Duan
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China
| | - Jia-Shan Shen
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China
| | - Hong-Bo Ye
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China
| | - Yan-Yan Zhu
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China
| | - Xiao-Dong Luo
- Yunnan Characteristic Plant Extraction Laboratory Co., Ltd., Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences Kunming, 650201, China.
| |
Collapse
|
2
|
Wang F, Wang X, Li S, Yang Q, Mu H, Li J, Yang Y. Chitosan and gelatin based sprayable hydrogels incorporating photothermal and long-acting antibiotic sterilization for infected wound management with shape adaptability. Carbohydr Polym 2025; 350:123046. [PMID: 39647949 DOI: 10.1016/j.carbpol.2024.123046] [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: 09/21/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 12/10/2024]
Abstract
Severe skin damage resulting from acute trauma is often accompanied by uncontrolled bleeding, microbial infections, and delayed wound healing. Herein, multifunctional sprayable hydrogels (CT-CS-ZIF@CIP Gel) were developed for wound management by incorporating antibacterial nanoplatforms (CT-CS-ZIF@CIP) into photocurable gels consisting of chitosan methacrylate and gallic acid grafted gelatin. The nanoplatform was initially constructed by sequentially loading Cu2Se (CS) and ciprofloxacin-decorated zeolitic imidazolate framework-8 (ZIF@CIP) onto Cu-doped Ti MOF (CT), in which CS served as a photothermal agent, ZIF enabled pH-responsive release of CIP, and CT acted as carriers for CS and ZIF@CIP. The hydrogel precursor can be sprayed onto wound surface and photocured quickly, allowing hydrogel to fit the wound shape and form a protective barrier onsite. The resultant hydrogel exhibited excellent hemostatic ability, adhesion properties, cytocompatibility and toxin adsorption capacity. By integrating CS for short-term photothermal therapy with CIP for long-acting chemotherapy, the CT-CS-ZIF@CIP Gel demonstrated 100 % sterilization of three bacterial strains. Furthermore, moderate release of zinc and copper ions promoted wound healing. The therapeutic efficacy of hydrogel was validated in an infected cutaneous mouse model. Overall, this work presents a versatile sprayable hydrogel that can be flexibly applied to irregular dynamic wounds for safe and effective wound management.
Collapse
Affiliation(s)
- Fei Wang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, Xinjiang, China; College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xing Wang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Siwei Li
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Qisen Yang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Haibo Mu
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, Xinjiang, China.
| | - Yu Yang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, Xinjiang, China.
| |
Collapse
|
3
|
Crispino R, Lagreca E, Procopio A, D'Auria R, Corrado B, La Manna S, Onesto V, Di Natale C. Advanced polymeric systems for colon drug delivery: from experimental models to market applications. SOFT MATTER 2025; 21:792-818. [PMID: 39801430 DOI: 10.1039/d4sm01222d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
In recent years, nano and micro drug delivery systems targeting the colon have gained more attention due to increasing interest in treating colon diseases such as colorectal cancer and inflammatory bowel disease, i.e., Crohn's disease and ulcerative colitis. Usually, nanocarriers are exploited for their enhanced permeability properties, allowing higher penetration effects and bioavailability, while microcarriers are primarily used for localized and sustained release. In bowel diseases, carriers must go into a delicate environment with a strict balance of gut bacteria (e.g., colon), and natural or biodegradable polymers capable of ensuring lower toxicity are preferred. However, these systems are primarily delivered orally, so the carrier must go through the whole gastrointestinal tract, where it encounters significant pH fluctuations, different mucus layers, several enzymes, and a long transit time. For this reason, various approaches have been explored and evaluated, especially using pH-responsive and time-dependent systems. This review provides an overview of the contemporary methodologies employed in orally administered nano- and microparticles for colon delivery, encompassing both in vivo and in vitro investigations. It evaluates their strengths, weaknesses, constraints, and potential enhancements, leveraging mathematical and microfluidic models. Furthermore, it focuses explicitly on systems that have already reached the market and are presently employed in treating severe colon diseases.
Collapse
Affiliation(s)
- R Crispino
- Center for Advanced Biomaterials for Health Care (CABHC), Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, Napoli, Italy
- Department of Chemical Materials and Industrial Production (DICMaPI), University of Naples Federico II, P.le Tecchio 80, Naples 80125, Italy.
| | - E Lagreca
- Center for Advanced Biomaterials for Health Care (CABHC), Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, Napoli, Italy
| | - A Procopio
- Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| | - R D'Auria
- Center for Advanced Biomaterials for Health Care (CABHC), Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, Napoli, Italy
| | - B Corrado
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, P.le Tecchio 80, Naples 80125, Italy
| | - S La Manna
- Department of Pharmacy, University of Naples Federico II, Naples 80131, Italy.
| | - V Onesto
- Center for Advanced Biomaterials for Health Care (CABHC), Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, Napoli, Italy
| | - C Di Natale
- Department of Chemical Materials and Industrial Production (DICMaPI), University of Naples Federico II, P.le Tecchio 80, Naples 80125, Italy.
| |
Collapse
|
4
|
Hou W, Cao Y, Wang J, Yin F, Wang J, Guo N, Wang Z, Lv X, Ma C, Chen Q, Yang R, Wei H, Li J, Wang R, Qin H. Single-cell nanocapsules of gut microbiota facilitate fecal microbiota transplantation. Theranostics 2025; 15:2069-2084. [PMID: 39897545 PMCID: PMC11780513 DOI: 10.7150/thno.104852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/19/2024] [Indexed: 02/04/2025] Open
Abstract
Rationale: Fecal microbiota transplantation (FMT) is advantageous for treating intractable diseases via the microbiota-gut-organ axis. However, invasive administration of gut microbiota via nasal feeding tubes limits the widespread application of FMT. Here, we attempted to develop a novel strategy to deliver gut microbiota using nanocapsules. Methods: Single-cell nanocapsules were fabricated within 1 h by layer-by-layer assembly of silk fibroin and phosphatidylcholine to generate a protective nanoshell on the cell surface of complicated microbiota. The physical properties of the microbiota nanocapsules were analyzed. The protective effects of nanocapsules on the gastrointestinal tract were analyzed both in vitro and in vivo. The efficacy of FMT assisted by single-cell nanocapsules (NanoFMT) was evaluated using the inflammatory response, gut microbiota balance, and histopathological analysis in animal model. Results: The nanocapsules achieved a good coating ratio for a single type of microbe and complex microbiota, resulting in a remarkable increase in the survival rate of microbes in the gastrointestinal tract. NanoFMT improved the diversity and abundance of the gut microbiota better than common FMT in germ-free mice. Moreover, NanoFMT alleviated intestinal inflammation and positively reversed the microbiota balance in a mouse model of colitis compared with common FMT, assisted by the inherent anti-inflammatory effects of silk fibroin and phosphatidylcholine. Conclusions: Considering its rapid preparation, convenient delivery, and perfect therapeutic effect, we anticipate that NanoFMT may be a promising clinical candidate for next-generation FMT treatment.
Collapse
Affiliation(s)
- Weiliang Hou
- Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072 Shanghai, China
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital; National Key Laboratory of Immunity and Inflammation, Naval Medical University, 200433 Shanghai, China
- Shanghai Cancer Institute, Renji Hospital School of Medicine, Shanghai Jiao Tong University, 200030 Shanghai, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, SAR 999078 Taipa Macau, China
| | - Yuan Cao
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, 200032 Shanghai, China
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Jifeng Wang
- Department of Pathology, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Fang Yin
- Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Jiahui Wang
- Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Ning Guo
- Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Ziyi Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, SAR 999078 Taipa Macau, China
| | - Xiaoqiong Lv
- Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Chunlian Ma
- Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Qiyi Chen
- Intestinal Microenvironment Treatment Center, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Rong Yang
- Department of Pediatrics, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072 Shanghai, China
| | - Hong Wei
- Central Laboratory, Clinical Medicine Scientific and Technical Innovation Park, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200435 Shanghai, China
| | - Juanjuan Li
- School of Life Sciences, Hainan University, 570228 Haikou, China
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, SAR 999078 Taipa Macau, China
| | - Huanlong Qin
- Research Institute of Intestinal Diseases, Shanghai Tenth People's Hospital Affiliated to Tongji University, 200072 Shanghai, China
| |
Collapse
|
5
|
Liu P, Luo Y, Zhang M. Intestinal microbiota and tuberculosis: Insights from Mendelian randomization. Medicine (Baltimore) 2024; 103:e38762. [PMID: 38968531 PMCID: PMC11250452 DOI: 10.1097/md.0000000000038762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/07/2024] [Indexed: 07/07/2024] Open
Abstract
Respiratory tuberculosis (RTB), a global health concern affecting millions of people, has been observationally linked to the gut microbiota, but the depth and nature of this association remain elusive. Despite these findings, the underlying causal relationship is still uncertain. Consequently, we used the Mendelian randomization (MR) method to further investigate this potential causal connection. We sourced data on the gut microbiota from a comprehensive genome-wide association study (GWAS) conducted by the MiBioGen Consortium (7686 cases, and 115,893 controls). For RTB, we procured 2 distinct datasets, labeled the Fingen R9 TBC RESP and Fingen R9 AB1 RESP, from the Finnish Genetic Consortium. To decipher the potential relationship between the gut microbiota and RTB, we employed MR on both datasets. Our primary mode of analysis was the inverse variance weighting (IVW) method. To ensure robustness and mitigate potential confounders, we meticulously evaluated the heterogeneity and potential pleiotropy of the outcomes. In the TBC RESP (RTB1) dataset related to the gut microbiota, the IVW methodology revealed 7 microbial taxa that were significantly associated with RTB. In a parallel vein, the AB1 RESP (RTB2) dataset highlighted 4 microbial taxa with notable links. Notably, Lachnospiraceae UCG010 was consistently identified across both datasets. This correlation was especially evident in the data segments designated Fingen R9 TBC RESP (OR = 1.799, 95% CI = 1.243-2.604) and Finngen R9 AB1 RESP (OR = 2.131, 95% CI = 1.088-4.172). Our study identified a causal relationship between particular gut microbiota and RTB at the level of prediction based on genetics. This discovery sheds new light on the mechanisms of RTB development, which are mediated by the gut microbiota.
Collapse
Affiliation(s)
- Peijun Liu
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Yaomei Luo
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Minghua Zhang
- Department of Respiratory and Critical Care Medicine, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| |
Collapse
|
6
|
Zou Q, Han S, Liang J, Yan G, Wang Q, Wang Y, Zhang Z, Hu J, Li J, Yuan T, Liu Z. Alleviating effect of vagus nerve cutting in Salmonella-induced gut infections and anxiety-like behavior via enhancing microbiota-derived GABA. Brain Behav Immun 2024; 119:607-620. [PMID: 38663772 DOI: 10.1016/j.bbi.2024.04.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 04/03/2024] [Accepted: 04/22/2024] [Indexed: 04/30/2024] Open
Abstract
The vagus nerve, a pivotal link within the gut-brain axis, plays a critical role in maintaining homeostasis and mediating communication between the gastrointestinal tract and the brain. It has been reported that gastrointestinal infection by Salmonella typhimurium (S. typhimurium) triggers gut inflammation and manifests as anxiety-like behaviors, yet the mechanistic involvement of the vagus nerve remains to be elucidated. In this study, we demonstrated that unilateral cervical vagotomy markedly attenuated anxiety-like behaviors induced by S. typhimurium SL1344 infection in C57BL/6 mice, as evidenced by the open field test and marble burying experiment. Furthermore, vagotomy significantly diminished neuronal activation within the nucleus of the solitary tract and amygdala, alongside mitigating aberrant glial cell activation in the hippocampus and amygdala. Additionally, vagotomy notably decreases serum endotoxin levels, counters the increase in splenic Salmonella concentration, and modulates the expression of inflammatory cytokines-including IL-6, IL-1β, and TNF-α-in both the gastrointestinal tract and brain, with a concurrent reduction in IL-22 and CXCL1 expression. This intervention also fostered the enrichment of beneficial gut microbiota, including Alistipes and Lactobacillus species, and augmented the production of gamma-aminobutyric acid (GABA) in the gut. Administration of GABA replicated the vagotomy's beneficial effects on reducing gut inflammation and anxiety-like behavior in infected mice. However, blockade of GABA receptors with picrotoxin abrogated the vagotomy's protective effects against gut inflammation, without influencing its impact on anxiety-like behaviors. Collectively, these findings suggest that vagotomy exerts a protective effect against infection by promoting GABA synthesis in the colon and alleviating anxiety-like behavior. This study underscores the critical role of the vagus nerve in relaying signals of gut infection to the brain and posits that targeting the gut-brain axis may offer a novel and efficacious approach to preventing gastrointestinal infections and associated behavioral abnormalities.
Collapse
Affiliation(s)
- Qianhui Zou
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Shiyao Han
- Molecular Biology Laboratory of Stem Cells and Anti-infection Medicine, College of Veterinary medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jiarui Liang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Guiming Yan
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Qianxu Wang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Yajie Wang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Zilong Zhang
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao, China
| | - Jun Hu
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Jufang Li
- Heilongjiang Feihe Dairy Co., Ltd., Beijing, China
| | - Tian Yuan
- Northwest A&F University Shenzhen Research Institute, Shenzhen, Guangdong, China; Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhigang Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China; Northwest A&F University Shenzhen Research Institute, Shenzhen, Guangdong, China; Shaanxi Precision Nutrition and Health Research Institute, Xi'an, Shaanxi, China.
| |
Collapse
|
7
|
Sun F, Xiao Y, Kong L, Mu H, Wang X, Duan J. An "in control" hyaluronic acid nanogel with light-cleavable for rational use of antibiotics. Heliyon 2024; 10:e33287. [PMID: 39027455 PMCID: PMC11254603 DOI: 10.1016/j.heliyon.2024.e33287] [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: 03/01/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 07/20/2024] Open
Abstract
The consequences caused by bacterial resistance are becoming more and more serious. The rate of antibiotic development is far behind the rate of bacterial resistance, so it is urgent to develop a new drug system. In this study, photoresponsive nanogels based on hyaluronic acid were prepared and loaded with ciprofloxacin as a model molecule. The results showed that the nanogels had the advantages of high stability and good cytocompatibility. The inhibition effect of drug-loaded nanogels after light irradiation on the growth of Staphylococcus aureus and Salmonella typhimurium was significantly better than that before light irradiation, and ciprofloxacin could be released on demand and in control. This strategy is of great significance to reduce the unnecessary use of antibiotics and weaken bacterial resistance.
Collapse
Affiliation(s)
- Feifei Sun
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- University of Chinese Academy of Sciences, China
- College of Chemistry & Pharmacy, Shaanxi Key Laboratory of Natural Products & Chemical Biology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yao Xiao
- College of Chemistry & Pharmacy, Shaanxi Key Laboratory of Natural Products & Chemical Biology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Lili Kong
- College of Chemistry & Pharmacy, Shaanxi Key Laboratory of Natural Products & Chemical Biology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Haibo Mu
- College of Chemistry & Pharmacy, Shaanxi Key Laboratory of Natural Products & Chemical Biology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xing Wang
- College of Chemistry & Pharmacy, Shaanxi Key Laboratory of Natural Products & Chemical Biology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jinyou Duan
- College of Chemistry & Pharmacy, Shaanxi Key Laboratory of Natural Products & Chemical Biology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| |
Collapse
|
8
|
Xu Y, Niu C, Liang S, Guo J, Li K, Zhang J, Li J, Jin Y, Bai J, Dai J, Lu C. An inulin-based glycovesicle for pathogen-targeted drug delivery to ameliorate salmonellosis. Int J Biol Macromol 2024; 267:131656. [PMID: 38636749 DOI: 10.1016/j.ijbiomac.2024.131656] [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: 09/20/2023] [Revised: 04/07/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024]
Abstract
The gut microbiota plays a significant role in the pathogenesis and remission of inflammatory bowel disease. However, conventional antibiotic therapies may alter microbial ecology and lead to dysbiosis of the gut microbiome, which greatly limits therapeutic efficacy. To address this challenge, novel nanomicelles that couple inulin with levofloxacin via disulfide bonds for the treatment of salmonellosis were developed in this study. Owing to their H2S-responsiveness, the nanomicelles can target the inflamed colon and rapidly release levofloxacin to selectively fight against enteric pathogens. Moreover, the embedded inulin can serve as prebiotic fiber to increase the amount of Bifidobacteria and Lactobacilli in mice with salmonellosis, thus maintaining the intestinal mechanical barrier and regulating the balance of the intestinal flora. Therefore, multifunctional nanomicelles had a better curative effect than pure levofloxacin on ameliorating inflammation in vivo. The pathogen-targeted glycovesicle represents a promising drug delivery platform to maximize the efficacy of antibacterial drugs for the treatment of inflammatory bowel disease.
Collapse
Affiliation(s)
- Yujie Xu
- College of Chemistry & Pharmacy, Shaanxi Key Laboratory of Natural Products & Chemical Biology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Congmin Niu
- Shandong Second Med Univ, Sch Biosci & Technol, Key Lab Biol Med Univ Shandong Prov, Baotong Rd, Weifang 261053, Shangdong, China
| | - Shuyi Liang
- Shandong Second Med Univ, Sch Biosci & Technol, Key Lab Biol Med Univ Shandong Prov, Baotong Rd, Weifang 261053, Shangdong, China
| | - Jiayi Guo
- Shandong Second Med Univ, Sch Biosci & Technol, Key Lab Biol Med Univ Shandong Prov, Baotong Rd, Weifang 261053, Shangdong, China
| | - Kaiming Li
- Shandong Second Med Univ, Sch Biosci & Technol, Key Lab Biol Med Univ Shandong Prov, Baotong Rd, Weifang 261053, Shangdong, China
| | - Jiarui Zhang
- Shandong Second Med Univ, Sch Biosci & Technol, Key Lab Biol Med Univ Shandong Prov, Baotong Rd, Weifang 261053, Shangdong, China
| | - Jingyuan Li
- Shandong Second Med Univ, Sch Biosci & Technol, Key Lab Biol Med Univ Shandong Prov, Baotong Rd, Weifang 261053, Shangdong, China
| | - Yaju Jin
- Shandong Second Med Univ, Sch Biosci & Technol, Key Lab Biol Med Univ Shandong Prov, Baotong Rd, Weifang 261053, Shangdong, China
| | - Jingkun Bai
- Shandong Second Med Univ, Sch Biosci & Technol, Key Lab Biol Med Univ Shandong Prov, Baotong Rd, Weifang 261053, Shangdong, China.
| | - Jiangkun Dai
- Shandong Second Med Univ, Sch Biosci & Technol, Key Lab Biol Med Univ Shandong Prov, Baotong Rd, Weifang 261053, Shangdong, China.
| | - Chunbo Lu
- Shandong Second Med Univ, Sch Biosci & Technol, Key Lab Biol Med Univ Shandong Prov, Baotong Rd, Weifang 261053, Shangdong, China.
| |
Collapse
|
9
|
Wei G, Liu W, Zhang Y, Zhou Z, Wang Y, Wang X, Zhu S, Li T, Wei H. Nanozyme-Enhanced Probiotic Spores Regulate the Intestinal Microenvironment for Targeted Acute Gastroenteritis Therapy. NANO LETTERS 2024; 24:2289-2298. [PMID: 38341876 DOI: 10.1021/acs.nanolett.3c04548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2024]
Abstract
Antibiotic therapeutics to combat intestinal pathogen infections often exacerbate microbiota dysbiosis and impair mucosal barrier functions. Probiotics are promising strategies, because they inhibit pathogen colonization and improve intestinal microbiota imbalance. Nevertheless, their limited targeting ability and susceptibility to oxidative stress have hindered their therapeutic potential. To tackle these challenges, Ces3 is synthesized by in situ growth of CeO2 nanozymes with positive charges on probiotic spores, facilitating electrostatic interactions with negatively charged pathogens and possessing a high reactive oxygen species (ROS) scavenging activity. Importantly, Ces3 can resist the harsh environment of the gastrointestinal tract. In mice with S. Typhimurium-infected acute gastroenteritis, Ces3 shows potent anti-S. Typhimurium activity, thereby alleviating the dissemination of S. Typhimurium into other organs. Additionally, owing to its O2 deprivation capacity, Ces3 promotes the proliferation of anaerobic probiotics, reshaping a healthy intestinal microbiota. This work demonstrates the promise of combining antibacterial, anti-inflammatory, and O2 content regulation properties for acute gastroenteritis therapy.
Collapse
Affiliation(s)
- Gen Wei
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Wanling Liu
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Yihong Zhang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Zijun Zhou
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Yuting Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Xiaoyu Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
- Department of Chemistry and Material Science, College of Science, Nanjing Forestry University, Nanjing, Jiangsu 210037, China
| | - Shuaishuai Zhu
- School of Materials Science and Engineering, Nanjing Institute of Technology, Nanjing, Jiangsu 211167, China
| | - Tong Li
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Hui Wei
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu 210023, China
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, Jiangsu 210023, China
| |
Collapse
|
10
|
Han K, Xu J, Xie F, Crowther J, Moon JJ. Engineering Strategies to Modulate the Gut Microbiome and Immune System. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:208-215. [PMID: 38166246 PMCID: PMC10766079 DOI: 10.4049/jimmunol.2300480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 08/28/2023] [Indexed: 01/04/2024]
Abstract
The gut microbiota, predominantly residing in the colon, is a complex ecosystem with a pivotal role in the host immune system. Dysbiosis of the gut microbiota has been associated with various diseases, and there is an urgent need to develop new therapeutics that target the microbiome and restore immune functions. This Brief Review discusses emerging therapeutic strategies that focus on oral delivery systems for modulating the gut microbiome. These strategies include genetic engineering of probiotics, probiotic-biomaterial hybrids, dietary fibers, and oral delivery systems for microbial metabolites, antimicrobial peptides, RNA, and antibiotics. Engineered oral formulations have demonstrated promising outcomes in reshaping the gut microbiome and influencing immune responses in preclinical studies. By leveraging these approaches, the interplay between the gut microbiota and the immune system can be harnessed for the development of novel therapeutics against cancer, autoimmune disorders, and allergies.
Collapse
Affiliation(s)
- Kai Han
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jin Xu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Fang Xie
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Julia Crowther
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - James J. Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
11
|
Han L, Zang T, Tan L, Liang D, Long T, Liu X, Shen X, Ren H, Li Z, Lu Z, Tang S, Liao X, Liu Y, Zhang C, Sun J. Self-assembly of H 2S-responsive nanoprodrugs based on natural rhein and geraniol for targeted therapy against Salmonella Typhimurium. J Nanobiotechnology 2023; 21:483. [PMID: 38104180 PMCID: PMC10725032 DOI: 10.1186/s12951-023-02256-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023] Open
Abstract
Salmonellosis is a globally extensive food-borne disease, which threatens public health and results in huge economic losses in the world annually. The rising prevalence of antibiotic resistance in Salmonella poses a significant global concern, emphasizing an imperative to identify novel therapeutic agents or methodologies to effectively combat this predicament. In this study, self-assembly hydrogen sulfide (H2S)-responsive nanoprodrugs were fabricated with poly(α-lipoic acid)-polyethylene glycol grafted rhein and geraniol (PPRG), self-assembled into core-shell nanoparticles via electrostatic, hydrophilic and hydrophobic interactions, with hydrophilic exterior and hydrophobic interior. The rhein and geraniol are released from self-assembly nanoprodrugs PPRG in response to Salmonella infection, which is known to produce hydrogen sulfide (H2S). PPRG demonstrated stronger antibacterial activity against Salmonella compared with rhein or geraniol alone in vitro and in vivo. Additionally, PPRG was also able to suppress the inflammation and modulate gut microbiota homeostasis. In conclusion, the as-prepared self-assembly nanoprodrug sheds new light on the design of natural product active ingredients and provides new ideas for exploring targeted therapies for specific Enteropathogens. Graphical illustration for construction of self-assembly nanoprodrugs PPRG and its antibacterial and anti-inflammatory activities on experimental Salmonella infection in mice.
Collapse
Affiliation(s)
- Lu Han
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, People's Republic of China
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Henry Fok School of Biology and Agriculture, Shaoguan University, Shaoguan, 512005, People's Republic of China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, People's Republic of China
| | - Tao Zang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, People's Republic of China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, People's Republic of China
| | - Lulu Tan
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, People's Republic of China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, People's Republic of China
| | - Dunsheng Liang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, People's Republic of China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, People's Republic of China
| | - Tengfei Long
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, People's Republic of China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, People's Republic of China
| | - Xuwei Liu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, People's Republic of China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, People's Republic of China
| | - Xiaofan Shen
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, People's Republic of China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, People's Republic of China
| | - Hao Ren
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, People's Republic of China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, People's Republic of China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - ZhiPeng Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, People's Republic of China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, People's Republic of China
| | - Zhaoxiang Lu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, People's Republic of China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, People's Republic of China
| | - Shengqiu Tang
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Henry Fok School of Biology and Agriculture, Shaoguan University, Shaoguan, 512005, People's Republic of China
| | - Xiaoping Liao
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, People's Republic of China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, People's Republic of China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Yahong Liu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, People's Republic of China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, People's Republic of China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Chaoqun Zhang
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, 510642, People's Republic of China.
| | - Jian Sun
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, People's Republic of China.
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, People's Republic of China.
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, People's Republic of China.
| |
Collapse
|
12
|
de Nies L, Kobras CM, Stracy M. Antibiotic-induced collateral damage to the microbiota and associated infections. Nat Rev Microbiol 2023; 21:789-804. [PMID: 37542123 DOI: 10.1038/s41579-023-00936-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2023] [Indexed: 08/06/2023]
Abstract
Antibiotics have transformed medicine, saving millions of lives since they were first used to treat a bacterial infection. However, antibiotics administered to target a specific pathogen can also cause collateral damage to the patient's resident microbial population. These drugs can suppress the growth of commensal species which provide protection against colonization by foreign pathogens, leading to an increased risk of subsequent infection. At the same time, a patient's microbiota can harbour potential pathogens and, hence, be a source of infection. Antibiotic-induced selection pressure can cause overgrowth of resistant pathogens pre-existing in the patient's microbiota, leading to hard-to-treat superinfections. In this Review, we explore our current understanding of how antibiotic therapy can facilitate subsequent infections due to both loss of colonization resistance and overgrowth of resistant microorganisms, and how these processes are often interlinked. We discuss both well-known and currently overlooked examples of antibiotic-associated infections at various body sites from various pathogens. Finally, we describe ongoing and new strategies to overcome the collateral damage caused by antibiotics and to limit the risk of antibiotic-associated infections.
Collapse
Affiliation(s)
- Laura de Nies
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Carolin M Kobras
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Mathew Stracy
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| |
Collapse
|
13
|
Yang X, Wang C, Wang Q, Zhang Z, Nie W, Shang L. Armored probiotics for oral delivery. SMART MEDICINE 2023; 2:e20230019. [PMID: 39188298 PMCID: PMC11235677 DOI: 10.1002/smmd.20230019] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 06/26/2023] [Indexed: 08/28/2024]
Abstract
As a kind of intestinal flora regulator, probiotics show great potential in the treatment of many diseases. However, orally delivered probiotics are often vulnerable to unfriendly gastrointestinal environments, resulting in a low survival rate and decreased therapeutic efficacy. Decorating or encapsulating probiotics with functional biomaterials has become a facile yet useful strategy, and probiotics can be given different functions by wearing different armors. This review systematically discusses the challenges faced by oral probiotics and the research progress of armored probiotics delivery systems. We focus on how various functional armors help probiotics overcome different obstacles and achieve efficient delivery. We also introduce the applications of armor probiotics in disease treatment and analyze the future trends of developing advanced probiotics-based therapies.
Collapse
Affiliation(s)
- Xinyuan Yang
- Zhongshan‐Xuhui Hospital and the Shanghai Key Laboratory of Medical Epigenetics, the International Co‐laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology)Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Chong Wang
- Zhongshan‐Xuhui Hospital and the Shanghai Key Laboratory of Medical Epigenetics, the International Co‐laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology)Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Qiao Wang
- Zhongshan‐Xuhui Hospital and the Shanghai Key Laboratory of Medical Epigenetics, the International Co‐laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology)Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Zhuohao Zhang
- Zhongshan‐Xuhui Hospital and the Shanghai Key Laboratory of Medical Epigenetics, the International Co‐laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology)Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Weimin Nie
- Key Laboratory of Smart Drug DeliverySchool of PharmacyFudan UniversityShanghaiChina
| | - Luoran Shang
- Zhongshan‐Xuhui Hospital and the Shanghai Key Laboratory of Medical Epigenetics, the International Co‐laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology)Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| |
Collapse
|
14
|
Qian X, Jiang H, Wu Y, Shao H, He W, He Y, Bao X, He L, Jia Y, Xu Z. Fecal microbiota transplantation combined with prebiotics ameliorates ulcerative colitis in mice. Future Microbiol 2023; 18:1251-1263. [PMID: 37830929 DOI: 10.2217/fmb-2023-0001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 07/19/2023] [Indexed: 10/14/2023] Open
Abstract
Aim: To investigate the effect of treatment with fecal microbiota transplantation (FMT) and galacto- and fructo-oligosaccharides on ulcerative colitis (UC) in mice. Materials & methods: A total of 90 mice, divided into nine groups, were administered FMT or prebiotics or combined treatment. The disease activity index scores, gut microbiota and inflammation factors were evaluated. Results: The treatment using FMT combined with galacto- and fructo-oligosaccharides in a 9:1 ratio significantly reduced intestinal barrier damage and alleviated symptoms of UC. Lactobacillus and Bifidobacterium and short-chain fatty acids were significantly increased after the combined treatment. Conclusion: The results demonstrate that FMT with prebiotics is a new method for UC treatment.
Collapse
Affiliation(s)
- Xueyi Qian
- Precision Medicine Center, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, 241001, China
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, 241002, China
| | - Hua Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, 241001, China
| | - Yao Wu
- Precision Medicine Center, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, 241001, China
| | - Huimin Shao
- Precision Medicine Center, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, 241001, China
| | - Weijie He
- Precision Medicine Center, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, 241001, China
| | - Yinmei He
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, 241002, China
| | - Xin Bao
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, 241002, China
| | - Lianjun He
- Precision Medicine Center, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, 241001, China
| | - Yuliang Jia
- Department of Gastroenterology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, 241001, China
| | - Zhenyu Xu
- Precision Medicine Center, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, 241001, China
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui Province, 241002, China
| |
Collapse
|
15
|
Yan D, Sun M, Xiao L, Chen H, Zhang Q, Zhi Y, Fu Q. Interspecies interactions of non-O1/O139 Vibrio cholerae and Salmonella typhimurium: a rare coinfection case report. Diagn Microbiol Infect Dis 2023; 107:116049. [PMID: 37595338 DOI: 10.1016/j.diagmicrobio.2023.116049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/19/2023] [Accepted: 07/28/2023] [Indexed: 08/20/2023]
Abstract
Infectious diseases are commonly demonstrated to be caused by polymicrobial infections, which correlate with increased infection severity and poorer clinical outcomes. In this study, we report a rare intestinal coinfection case of non-O1/O139 Vibrio cholerae and Salmonella typhimurium, along with V. cholerae septicemia. The data of quantitative real-time PCR and competition assay showed that V. cholerae may present enhanced virulence in the presence of S. typhimurium, and exerted an inhibitory growth effect over S. typhimurium in vitro.
Collapse
Affiliation(s)
- Dongmei Yan
- Department of Clinical Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, Jiangsu, China
| | - Mingzhong Sun
- Department of Clinical Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, Jiangsu, China
| | - Lihua Xiao
- Department of Clinical Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, Jiangsu, China
| | - Hongmei Chen
- Department of Clinical Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, Jiangsu, China
| | - Qisi Zhang
- Department of Clinical Laboratory, Henan Provincial People's Hospital, Department of Clinical Laboratory of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yaru Zhi
- Department of Clinical Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, Jiangsu, China
| | - Qingping Fu
- Department of Clinical Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People's Hospital, Yancheng, Jiangsu, China.
| |
Collapse
|
16
|
Luo Y, De Souza C, Ramachandran M, Wang S, Yi H, Ma Z, Zhang L, Lin K. Precise oral delivery systems for probiotics: A review. J Control Release 2022; 352:371-384. [PMID: 36309096 DOI: 10.1016/j.jconrel.2022.10.030] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/16/2022] [Accepted: 10/17/2022] [Indexed: 11/07/2022]
Abstract
Probiotics have several health benefits to the host. However, low pH in the stomach, various digestive enzymes and bile salts in the intestine threaten their viability and function. Thus, probiotics need to be protected during gastric transit to address challenges associated with low viability and impaired function. At present, probiotic delivery systems with different trigger mechanisms have been constructed to successfully introduce numerous high-viability probiotics to the intestine. On this basis, the application of non-targeted/targeted probiotic delivery systems in different gut microenvironment and the adjuvant therapeutic effect of probiotic delivery systems on other disease were discussed in detail. It is important to also note that most of the current studies in this area focused on non-targeted probiotic delivery systems. Moreover, changes in intestinal microenvironment under disease state and discontinuous distribution of disease site limit their development. Thus, emphasis were made on the optimization of non-targeted probiotic delivery systems and the necessity of designing more precisely targeted ones.
Collapse
Affiliation(s)
- Ya Luo
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, China
| | - Cristabelle De Souza
- Department of Stem Cell Research and Regenerative Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mythili Ramachandran
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Shaolei Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, China
| | - Huaxi Yi
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, China
| | - Zhao Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmacy, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Lanwei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, China.
| | - Kai Lin
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong 266003, China.
| |
Collapse
|
17
|
Huang L, Wang J, Kong L, Wang X, Li Q, Zhang L, Shi J, Duan J, Mu H. ROS-responsive hyaluronic acid hydrogel for targeted delivery of probiotics to relieve colitis. Int J Biol Macromol 2022; 222:1476-1486. [PMID: 36195227 DOI: 10.1016/j.ijbiomac.2022.09.247] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/23/2022] [Accepted: 09/27/2022] [Indexed: 11/05/2022]
Abstract
Probiotics are generally used as therapeutic intervention in inflammatory bowel disease. However, the low survival rate in harsh gastrointestinal environment and limited retention in intestine greatly restrict their health benefits. To address this problem, a ROS-responsive hydrogel based on hyaluronic acid (HA) was developed for encapsulation and targeted delivery of probiotics. The hydrogel was prepared facilely by physiological crosslink with methacrylated HA and thiolated thioketal. As a model probiotic, Lactobacillu reuteri showed a significantly increased survival rate in simulated digestive conditions after encapsulated in hydrogel. The negative properties conferred the hydrogel preferential adhesions to inflammation sites. Meanwhile, the excess reactive oxygen species (ROS) produced by inflamed colon tissues selectively cleaved thioketal linkages resulted in hydrogel degradation and local probiotics release. Furthermore, the hydrogel exerted an appropriate ROS-scavenge capacity and protected HT-29 cells from oxidative damage. Animal experiments indicated that hydrogel-encapsulated L. reuteri could remarkably alleviate the symptoms and improve the survival rate of mice with dextran sulfate sodium (DSS)-induced colitis. These results suggested that the biocompatible hydrogel may be a delivery platform to target inflamed intestines and expand the application of probiotics as pharmaceuticals.
Collapse
Affiliation(s)
- Lijie Huang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Junjie Wang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Lili Kong
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xing Wang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Qiulei Li
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Lingjiao Zhang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jingru Shi
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jinyou Duan
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Haibo Mu
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|
18
|
Yuan Y, Liu Y, He Y, Zhang B, Zhao L, Tian S, Wang Q, Chen S, Li Z, Liang S, Hou G, Liu B, Li Y. Intestinal-targeted nanotubes-in-microgels composite carriers for capsaicin delivery and their effect for alleviation of Salmonella induced enteritis. Biomaterials 2022; 287:121613. [PMID: 35700621 DOI: 10.1016/j.biomaterials.2022.121613] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 04/05/2022] [Accepted: 05/30/2022] [Indexed: 02/09/2023]
Abstract
Salmonella is a word-wide food-borne pathogen, which can cause severe enteritis and intestinal microbiota imbalance. Capsaicin (Cap), a food-based bioactive ingredient, has antibacterial and anti-inflammatory properties. However, its low solubility, low bioavailability and the irritation to digestive tract greatly limit its applications. Here, an intestinal responsively "nanotubes-in-microgel" composite carrier was constructed by capturing α-lactalbumin (α-lac) nanotubes in low-methoxy pectin microgels (LMP-NT) (52 μm). Cap was loaded in such system via hydrophobic interaction with a loading capacity of 38.02 mg/g. The LMP microgels remained stable and protected NT/Cap from early releasing in the gastric condition. It showed an excellent mucoadhesive capacity, which can prolong the intestinal retention up to 12 h and control release NT/Cap in intestine. Afterward, NT/Cap could penetrate across the mucus layer deeply and enter the intestinal villi epithelial cells efficiently. LMP-NT microgels achieved a mucoadhesive-to-penetrating transition in response to intestinal pH, improving the epithelium absorption and the in vivo bioavailability of Cap. Oral administration of LMP-NT/Cap could effectively alleviate enteritis caused by Salmonella infection and maintain the homeostasis of gut microbiota. Overall, this work suggested that LMP-NT composite microgels were promising for intestine-targeted and oral delivery of hydrophobic bioactive food compounds.
Collapse
Affiliation(s)
- Yu Yuan
- Key Laboratory of Precision Nutrition and Food Quality, Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Ying Liu
- Key Laboratory of Precision Nutrition and Food Quality, Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Yang He
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Bingkun Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Liang Zhao
- Key Laboratory of Precision Nutrition and Food Quality, Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Simin Tian
- Key Laboratory of Precision Nutrition and Food Quality, Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Qimeng Wang
- Key Laboratory of Precision Nutrition and Food Quality, Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Shanan Chen
- Key Laboratory of Precision Nutrition and Food Quality, Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Zekun Li
- Key Laboratory of Precision Nutrition and Food Quality, Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Shuang Liang
- Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Guohua Hou
- Key Laboratory of Precision Nutrition and Food Quality, Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Bin Liu
- Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Yuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|
19
|
Tu H, Li S, Luo Z, Xu L, Zhang H, Xiang Y, Deng W, Zou G, Hou H, Ji X. Bi-doped carbon dots for a stable lithium metal anode. Chem Commun (Camb) 2022; 58:6449-6452. [PMID: 35552567 DOI: 10.1039/d2cc01334g] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Bi-doped carbon dots (Bi-CDs) with rich polar groups and good compatibility were employed as co-deposition electrolyte additives to homogenize Li+ flux for dendrite-free Li deposition. High coulombic efficiency (99.0%) and long-term stability (800 h) with reduced overpotential (∼15 mV) were achieved after introducing Bi-CDs in conventional electrolyte for high-performance Li-S batteries.
Collapse
Affiliation(s)
- Hanyu Tu
- College of Chemistry and Chemical Engineering, Central South University, Changsha, China.
| | - Shuo Li
- College of Chemistry and Chemical Engineering, Central South University, Changsha, China.
| | - Zheng Luo
- College of Chemistry and Chemical Engineering, Central South University, Changsha, China.
| | - Laiqiang Xu
- College of Chemistry and Chemical Engineering, Central South University, Changsha, China.
| | - Hao Zhang
- College of Chemistry and Chemical Engineering, Central South University, Changsha, China.
| | - Yinger Xiang
- College of Chemistry and Chemical Engineering, Central South University, Changsha, China.
| | - Wentao Deng
- College of Chemistry and Chemical Engineering, Central South University, Changsha, China.
| | - Guoqiang Zou
- College of Chemistry and Chemical Engineering, Central South University, Changsha, China.
| | - Hongshuai Hou
- College of Chemistry and Chemical Engineering, Central South University, Changsha, China.
| | - Xiaobo Ji
- College of Chemistry and Chemical Engineering, Central South University, Changsha, China. .,School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
20
|
Zhou Z, Li J, Tan L, Liu X, Zheng Y, Cui Z, Li C, Yeung KWK, Li Z, Liang Y, Zhu S, Wu S. Using tea nanoclusters as β-lactamase inhibitors to cure multidrug-resistant bacterial pneumonia: A promising therapeutic strategy by Chinese materioherbology. FUNDAMENTAL RESEARCH 2022; 2:496-504. [PMID: 38933406 PMCID: PMC11197604 DOI: 10.1016/j.fmre.2021.11.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/26/2021] [Accepted: 11/02/2021] [Indexed: 10/19/2022] Open
Abstract
β-lactamase, a kind of hydrolase in multi-drug resistant pathogens, can hydrolyze β-lactam antibiotics and make these kinds of antibiotics invalid. Small-molecular inhibitors about the enzyme and their mechanism are widely investigated but they may result in unavoidable adverse reactions and drug-resistance. Herein, we propose a new therapeutic strategy of Chinese materioherbology, in which herbal medicine or traditional Chinese medicinal herbs can be employed as biological functional materials or refreshed/excited by means of materialogy. Natural tea nanoclusters (TNCs) were extracted from tea to inhibit β-lactamase. Different from the mechanism of small-molecular inhibitors inhibiting enzymes by binding to the corresponding active sites, the TNCs as a cap cover the protein pocket and create a spatial barrier between the active sites and antibiotics, which was named "capping-pocket" effect. TNCs were combined with amoxicillin sodium (Amo) to treat the methicillin-resistant Staphylococcus aureus (MRSA) pneumonia in mice. This combinatorial therapy remarkably outperforms antibiotic monotherapy in reducing MRSA infections and the associated inflammation in mice. The therapeutic strategy exhibited excellent biosafety, without any side effects, even in piglets. Hence, TNCs have great clinical value in potentiating β-lactam antibiotic activity for combatting multi-drug resistant pathogen infections and the "pocket capping" effect can guide the design of new enzyme inhibitors in near future.
Collapse
Affiliation(s)
- Ziao Zhou
- Biomedical Materials Engineering Research Center, Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, Hubei University, Wuhan 430062, China
| | - Jun Li
- School of Materials Science & Engineering, Peking University, Beijing 100871, China
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China
| | - Lei Tan
- Biomedical Materials Engineering Research Center, Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, Hubei University, Wuhan 430062, China
| | - Xiangmei Liu
- Biomedical Materials Engineering Research Center, Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, Hubei University, Wuhan 430062, China
| | - Yufeng Zheng
- School of Materials Science & Engineering, Peking University, Beijing 100871, China
| | - Zhenduo Cui
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China
| | - Changyi Li
- Stomatological Hospital, Tianjin Medical University, No. 12, Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Kelvin Wai Kwok Yeung
- Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong 999077, China
| | - Zhaoyang Li
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China
| | - Yanqin Liang
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China
| | - Shengli Zhu
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China
| | - Shuilin Wu
- School of Materials Science & Engineering, Peking University, Beijing 100871, China
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin 300072, China
| |
Collapse
|
21
|
Chen QW, Qiao JY, Liu XH, Zhang C, Zhang XZ. Customized materials-assisted microorganisms in tumor therapeutics. Chem Soc Rev 2021; 50:12576-12615. [PMID: 34605834 DOI: 10.1039/d0cs01571g] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Microorganisms have been extensively applied as active biotherapeutic agents or drug delivery vehicles for antitumor treatment because of their unparalleled bio-functionalities. Taking advantage of the living attributes of microorganisms, a new avenue has been opened in anticancer research. The integration of customized functional materials with living microorganisms has demonstrated unprecedented potential in solving existing questions and even conferring microorganisms with updated antitumor abilities and has also provided an innovative train of thought for enhancing the efficacy of microorganism-based tumor therapy. In this review, we have summarized the emerging development of customized materials-assisted microorganisms (MAMO) (including bacteria, viruses, fungi, microalgae, as well as their components) in tumor therapeutics with an emphasis on the rational utilization of chosen microorganisms and tailored materials, the ingenious design of biohybrid systems, and the efficacious antitumor mechanisms. The future perspectives and challenges in this field are also discussed.
Collapse
Affiliation(s)
- Qi-Wen Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Ji-Yan Qiao
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Xin-Hua Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Cheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China.
| |
Collapse
|
22
|
Zhou Q, Gu R, Xue B, Li P, Gu Q. Phenyl lactic acid alleviates Samonella Typhimurium-induced colitis via regulating microbiota composition, SCFA production and inflammatory responses. Food Funct 2021; 12:5591-5606. [PMID: 34017972 DOI: 10.1039/d1fo00166c] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Colitis caused by non-typhoidal Salmonella (NST) infection is increasingly serious and widespread, so new effective treatment strategies with little or no side-effects are urgently needed. Our previous research found that phenyl lactic acid (PLA) derived from Lactobacillus plantarum ZJ316 can effectively inhibit Salmonella enterica Typhimurium (S. Typhimurium). In this study, we further investigated the protective effects of this PLA against S. Typhimurium-induced colitis in mice. An infection model was established using female C57BL/6J mice by oral administration of 109 CFU mL-1 of S. Typhimurium, and PLA was supplied for 10 days after infection. In colitic mice, PLA administration reduced the disease activity index, prevented the colon shortening and spleen enlargement, decreased liver enzyme (AST and ALT) activities, and alleviated the colonic tissue damage. RT-qPCR analysis showed that PLA significantly down-regulated the levels of NF-κB, TLR4 and pro-inflammatory cytokines (IFN-γ, IL-1β and TNF-α), but stimulated the mRNA expression of the anti-inflammatory cytokine IL-10. Changes in intestinal microecology were analyzed by 16S rRNA sequencing. PLA modulated colonic microbiota dysbiosis by increasing the abundance of Lactobacillus, Butyricicoccus and Roseburia, and reducing Salmonella and Alloprevotella at the genus level. In addition, PLA significantly increased the concentrations of short-chain fatty acids (SCFAs) in the colon, especially propionic acid and butyric acid. These findings revealed that PLA has potential benefits on alleviating S. Typhimurium-induced colitis mainly through intestinal microbiota regulation and inflammation elimination, providing a new perspective for the NTS infection treatment strategy.
Collapse
Affiliation(s)
- Qingqing Zhou
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Rongcheng Gu
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, USA
| | - Bingyao Xue
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Ping Li
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| | - Qing Gu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang 310018, China.
| |
Collapse
|
23
|
Wang H, Banerjee N, Liang Y, Wang G, Hoffman KL, Khan MF. Gut microbiome-host interactions in driving environmental pollutant trichloroethene-mediated autoimmunity. Toxicol Appl Pharmacol 2021; 424:115597. [PMID: 34051218 DOI: 10.1016/j.taap.2021.115597] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/18/2021] [Accepted: 05/24/2021] [Indexed: 12/15/2022]
Abstract
Trichloroethene (TCE), a widely used industrial solvent, is associated with the development of autoimmune diseases (ADs), including systemic lupus erythematosus and autoimmune hepatitis. Increasing evidence support a linkage between altered gut microbiome composition and the onset of ADs. However, it is not clear how gut microbiome contributes to TCE-mediated autoimmunity, and initial triggers for microbiome-host interactions leading to systemic autoimmune responses remain unknown. To achieve this, female MRL+/+ mice were treated with 0.5 mg/ml TCE for 52 weeks and fecal samples were subjected to 16S rRNA sequencing to determine the microbiome composition. TCE exposure resulted in distinct bacterial community revealed by β-diversity analysis. Notably, we observed reduction in Lactobacillaceae, Rikenellaceae and Bifidobacteriaceae families, and enrichment of Akkermansiaceae and Lachnospiraceae families after TCE exposure. We also observed significantly increased colonic oxidative stress and inflammatory markers (CD14 and IL-1β), and decreased tight junction proteins (ZO-2, occludin and claudin-3). These changes were associated with increases in serum antinuclear and anti-smooth muscle antibodies and cytokines (IL-6 and IL-12), together with increased PD1 + CD4+ T cells in TCE-exposed spleen and liver tissues. Importantly, fecal microbiota transplantation (FMT) using feces from TCE-treated mice to antibiotics-treated mice induced increased anti-dsDNA antibodies and hepatic CD4+ T cell infiltration in the recipient mice. Our studies thus delineate how imbalance in gut microbiome and mucosal redox status together with gut inflammatory response and permeability changes could be the key factors in contributing to TCE-mediated ADs. Furthermore, FMT studies provide a solid support to a causal role of microbiome in TCE-mediated autoimmunity.
Collapse
Affiliation(s)
- Hui Wang
- Department of Pathology, University of Texas Medical Branch, TX, United States of America
| | - Nivedita Banerjee
- Department of Pathology, University of Texas Medical Branch, TX, United States of America
| | - Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, TX, United States of America
| | - Gangduo Wang
- Department of Pathology, University of Texas Medical Branch, TX, United States of America
| | - Kristi L Hoffman
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - M Firoze Khan
- Department of Pathology, University of Texas Medical Branch, TX, United States of America.
| |
Collapse
|
24
|
Rational collaborative ablation of bacterial biofilms ignited by physical cavitation and concurrent deep antibiotic release. Biomaterials 2020; 262:120341. [PMID: 32911255 DOI: 10.1016/j.biomaterials.2020.120341] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 07/10/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022]
Abstract
Bacteria biofilm has extracellular polymeric substances to protect bacteria from external threats, which is a stubborn problem for human health. Herein, a kind of gasifiable nanodroplet is fabricated to ablate Staphylococcus aureus (S. aureus) biofilm. Upon NIR pulsed laser irradiation, the nanodroplets can gasify to generate destructive gas shockwave, which further potentiates initial acoustic cavitation effect, thus synergistically disrupting the protective biofilm and killing resident bacteria. More importantly, the gasification can further promote antibiotic release in deep biofilm for residual bacteria eradication. The nanodroplets not only exhibit deep biofilm penetration capacity and high potency to ablate biofilms, but also good biocompatibility without detectable side effects. In vivo mouse implant model indicates that the nanodroplets can accumulate at the S. aureus infected implant sites. Upon pulsed laser treatment, the nanodroplets efficiently eradicate bacteria biofilm in implanted catheter by synergistic contribution of gas shockwave-enhanced cavitation and deep antibiotic release. Current phase changeable nanodroplets with synergistic physical and chemical therapeutic modalities are promising to combat complex bacterial biofilms with drug resistance, which provides an alternative visual angle for biofilm inhibition in biomedicine.
Collapse
|
25
|
Xiao Y, Lu C, Liu Y, Kong L, Bai H, Mu H, Li Z, Geng H, Duan J. Encapsulation of Lactobacillus rhamnosus in Hyaluronic Acid-Based Hydrogel for Pathogen-Targeted Delivery to Ameliorate Enteritis. ACS APPLIED MATERIALS & INTERFACES 2020; 12:36967-36977. [PMID: 32702229 DOI: 10.1021/acsami.0c11959] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Probiotics were found to be effective in ameliorating the microbial dysbiosis and inflammation caused by intestinal pathogens. However, biological challenges encountered during oral delivery have greatly limited their potential health benefits. Here, a model probiotic (Lactobacillus rhamnosus) was encapsulated in an intestinal-targeted hydrogel to alleviate bacterial enteritis in a novel mode. The hydrogel was prepared simply by the self-cross-linking of thiolated hyaluronic acid. Upon exposure to H2S which were excreted by surrounding intestinal pathogens, the hydrogel can locally degrade and rapidly release cargos to compete with source pathogens in turn for binding to the host. The mechanical properties of hydrogel were studied by rheological analysis, and the ideal stability was achieved at a polymer concentration of 4% (w/v). The morphology of the optimal encapsulation system was further measured by a scanning electron microscope, exhibiting uniform payload of probiotics. Endurance experiments indicated that the encapsulation of L. rhamnosus significantly enhanced their viability under gastrointestinal tract insults. Compared with free cells, encapsulated L. rhamnosus exerted better therapeutic effect against Salmonella-induced enteritis with negligible toxicity in vivo. These results demonstrate that this redox-responsive hydrogel may be a promising encapsulation and delivery system for improving the efficacy of orally administered probiotics.
Collapse
Affiliation(s)
- Yao Xiao
- College of Chemistry & Pharmacy, Shaanxi Key Laboratory of Natural Products & Chemical Biology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Chunbo Lu
- College of Chemistry & Pharmacy, Shaanxi Key Laboratory of Natural Products & Chemical Biology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yinyin Liu
- College of Chemistry & Pharmacy, Shaanxi Key Laboratory of Natural Products & Chemical Biology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - LiLi Kong
- College of Chemistry & Pharmacy, Shaanxi Key Laboratory of Natural Products & Chemical Biology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Hu Bai
- College of Chemistry & Pharmacy, Shaanxi Key Laboratory of Natural Products & Chemical Biology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Haibo Mu
- College of Chemistry & Pharmacy, Shaanxi Key Laboratory of Natural Products & Chemical Biology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Zehao Li
- College of Chemistry & Pharmacy, Shaanxi Key Laboratory of Natural Products & Chemical Biology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Huiling Geng
- College of Chemistry & Pharmacy, Shaanxi Key Laboratory of Natural Products & Chemical Biology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jinyou Duan
- College of Chemistry & Pharmacy, Shaanxi Key Laboratory of Natural Products & Chemical Biology, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
26
|
Feng P, Cao Z, Wang X, Li J, Liu J. On-Demand Bacterial Reactivation by Restraining within a Triggerable Nanocoating. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002406. [PMID: 32686247 DOI: 10.1002/adma.202002406] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/17/2020] [Indexed: 06/11/2023]
Abstract
Bacteria have been widely exploited as bioagents for applications in diagnosis and treatment, benefitting from their living characteristics including colonization, rapid proliferation, and facile genetic manipulation. As such, bacteria being tailored to perform precisely in the right place at the right time to avoid potential side effects would be of great importance but has proven to be difficult. Here, a strategy of on-demand bacterial reactivation is described by individually restraining within a triggerable nanocoating. Upon reaching at a location of interest, nanocoatings can be triggered to dissolution in situ and subsequently decoat the bacteria which are able to recover their bioactivities as needed. It is demonstrated that gut microbiota coated with an enteric nanocoating can respond to gastrointestinal environments and reactivate in the intestine by a pH-triggered decoating. In virtue of this unique, coated bacteria remain inactive following oral administration to exempt acidic insults, while revive to restore therapeutic effects after gastric emptying. Consequently, improved oral availability and treatment efficacy are achieved in two mouse models of intestinal infection. Bacteria restrained by a triggerable nanocoating represent a smart therapeutic that can take effect when necessary. On-demand bacterial reactivation suggests a robust platform for the development of precision bacterial-mediated bioagents.
Collapse
Affiliation(s)
- Pingping Feng
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhenping Cao
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xinyue Wang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Juanjuan Li
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| |
Collapse
|
27
|
Varma DM, Zahid MSH, Bachelder EM, Ainslie KM. Formulation of host-targeted therapeutics against bacterial infections. Transl Res 2020; 220:98-113. [PMID: 32268128 PMCID: PMC10132281 DOI: 10.1016/j.trsl.2020.03.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/03/2020] [Accepted: 03/09/2020] [Indexed: 12/23/2022]
Abstract
The global burden of bacterial infections is rising due to increasing resistance to the majority of first-line antibiotics, rendering these drugs ineffective against several clinically important pathogens. Limited transport of antibiotics into cells compounds this problem for gram-negative bacteria that exhibit prominent intracellular lifecycles. Furthermore, poor bioavailability of antibiotics in infected tissues necessitates higher doses and longer treatment regimens to treat resistant infections. Although emerging antibiotics can combat these problems, resistance still may develop over time. Expanding knowledge of host-pathogen interactions has inspired research and development of host-directed therapies (HDTs). HDTs target host-cell machinery critical for bacterial pathogenesis to treat bacterial infections alone or as adjunctive treatment with traditional antibiotics. Unlike traditional antibiotics that directly affect bacteria, a majority of HDTs function by boosting the endogenous antimicrobial activity of cells and are consequently less prone to bacterial tolerance induced by selection pressure. Therefore, HDTs can be quite effective against intracellular cytosolic or vacuolar bacteria, which a majority of traditional antibiotics are unable to eradicate. However, in vivo therapeutic efficacy of HDTs is reliant on adequate bioavailability. Particle-based formulations demonstrate the potential to enable targeted drug delivery, enhance cellular uptake, and increase drug concentration in the host cell of HDTs. This review selected HDTs for clinically important pathogens, identifies formulation strategies that can improve their therapeutic efficacy and offers insights toward further development of HDTs for bacterial infections.
Collapse
Affiliation(s)
- Devika M Varma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina
| | - M Shamim Hasan Zahid
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina; Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina.
| |
Collapse
|