1
|
Sun R, Lv Z, Wang Y, Li M, Qi J, Wang K, Yang H, Yue T, Yuan Y. Different polysaccharide-enhanced probiotic and polyphenol dual-functional factor co-encapsulated microcapsules demonstrate acute colitis alleviation efficacy and food fortification. Carbohydr Polym 2024; 345:122572. [PMID: 39227107 DOI: 10.1016/j.carbpol.2024.122572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/19/2024] [Accepted: 08/01/2024] [Indexed: 09/05/2024]
Abstract
Probiotics and polyphenols have multiple bioactivities, and developing co-encapsulated microcapsules (CM) is a novel strategy to enhance their nutritional diversity. However, the development of CMs is challenged by complicated processing, single types, and unclear in vivo effects and applications. In this study, the co-microencapsulations of polyphenol and probiotic were constructed using pectin, alginate (WGCA@LK), and Fu brick tea polysaccharides (WGCF@LK), respectively, with chitosan-whey isolate proteins by layer-by-layer coacervation reaction, and their protective effects, in vivo effectiveness, and application potential were evaluated. WGCA@LK improved the encapsulation rate of polyphenols (42.41 %), and remained high viability of probiotics after passing through gastric acidic environment (8.79 ± 0.04 log CFU/g) and storage for 4 weeks (4.59 ± 0.06 log CFU/g). WGCF@LK exhibited the highest total antioxidant activity (19.40 ± 0.25 μmol/mL) and its prebiotic activity removed the restriction on probiotic growth. WGCA@LK showed strong in vitro colonic adhesion, but WGCF@LK promoted in vivo retention of probiotics at 48 h. WGCF@LK showed excellent anti-inflammatory effects and alleviated symptoms of acute colitis in mice. These findings provide unique insights into the fortification of probiotic-polyphenol CMs by different polysaccharides and the development of novel health foods with rich functional hierarchies and superior therapeutic effects.
Collapse
Affiliation(s)
- Rui Sun
- College of Food Science and Technology, Northwest University, Xi'an, 710069, China; College of Food Science and Engineering, Northwest A&F University, YangLing 712100, Shaanxi, China
| | - Zhongyi Lv
- College of Food Science and Technology, Northwest University, Xi'an, 710069, China; College of Food Science and Engineering, Northwest A&F University, YangLing 712100, Shaanxi, China
| | - Ying Wang
- College of Food Science and Engineering, Northwest A&F University, YangLing 712100, Shaanxi, China
| | - Menghui Li
- College of Food Science and Engineering, Northwest A&F University, YangLing 712100, Shaanxi, China
| | - Jianrui Qi
- College of Food Science and Engineering, Northwest A&F University, YangLing 712100, Shaanxi, China
| | - Kai Wang
- College of Food Science and Engineering, Northwest A&F University, YangLing 712100, Shaanxi, China
| | - Haihua Yang
- College of Food Science and Engineering, Northwest A&F University, YangLing 712100, Shaanxi, China
| | - Tianli Yue
- College of Food Science and Technology, Northwest University, Xi'an, 710069, China; College of Food Science and Engineering, Northwest A&F University, YangLing 712100, Shaanxi, China; Selenium-rich Tea Research and Development Center, Northwest University, Xi'an, 710069, China.
| | - Yahong Yuan
- College of Food Science and Technology, Northwest University, Xi'an, 710069, China; College of Food Science and Engineering, Northwest A&F University, YangLing 712100, Shaanxi, China; Selenium-rich Tea Research and Development Center, Northwest University, Xi'an, 710069, China.
| |
Collapse
|
2
|
Huang Y, Peng S, Zeng R, Yao H, Feng G, Fang J. From probiotic chassis to modification strategies, control and improvement of genetically engineered probiotics for inflammatory bowel disease. Microbiol Res 2024; 289:127928. [PMID: 39405668 DOI: 10.1016/j.micres.2024.127928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 10/05/2024] [Accepted: 10/05/2024] [Indexed: 11/02/2024]
Abstract
With the rising morbidity of inflammatory bowel disease (IBD) year by year, conventional therapeutic drugs with systemic side effects are no longer able to meet the requirements of patients. Probiotics can improve gut microbiota, enhance intestinal barrier function, and regulate mucosal immunity, making them a potential complementary or alternative therapy for IBD. To compensate for the low potency of probiotics, genetic engineering technology has been widely used to improve their therapeutic function. In this review, we systematically summarize the genetically engineered probiotics used for IBD treatment, including probiotic chassis, genetic modification strategies, methods for controlling probiotics, and means of improving efficacy. Finally, we provide prospects on how genetically engineered probiotics can be extended to clinical applications.
Collapse
Affiliation(s)
- Yuewen Huang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Shan Peng
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Rong Zeng
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Hao Yao
- Changsha IMADEK Intelligent Technology Co., LTD, Changsha 410081, China
| | - Guangfu Feng
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China.
| | - Jun Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
3
|
Gui Y, Sun Q, Li K, Lin L, Zhou H, Ma J, Li C. Bioinspired gelated cell sheet-supported lactobacillus biofilm for aerobic vaginitis diagnosis and treatment. SCIENCE ADVANCES 2024; 10:eadq2732. [PMID: 39485840 PMCID: PMC11529721 DOI: 10.1126/sciadv.adq2732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024]
Abstract
Aerobic vaginitis (AV) is a long-standing inflammatory disease that affects female patients. The use of antibiotics is a common means for AV treatment, but it will indiscriminately kill both pathogenic bacteria and beneficial strains, which easily causes vaginal dysbacteriosis and infection recurrence. Herein, we describe a bioinspired strategy for fabricating gelated cell sheet-supported lactobacillus biofilms (GCS-LBs) for AV treatment. Compared with common planktonic probiotic formulations, probiotic biofilms forming on a robust GCS exhibit enhanced stress tolerance and better colonization capacity in the mouse vagina. Moreover, DNA nanodevices are decorated on the GCS and dynamically report the microenvironment change of biofilms for timely evaluating bacterium activity, both in vitro and in vivo. Consequently, GCS-LBs are used for treating AV in an Escherichia coli-infected mouse model, which shows enhanced therapeutic efficacy compared with conventional antibiotic or lactobacillus monotherapy. Overall, the GCS-LB shows promise as a potent multifunctional tool to combat bacterial infection.
Collapse
Affiliation(s)
- Yueyue Gui
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P. R. China
| | - Qingfei Sun
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Kexin Li
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Longjia Lin
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Han Zhou
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Jiehua Ma
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P. R. China
| | - Chao Li
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| |
Collapse
|
4
|
Xu C, Guo J, Chang B, Zhang Y, Tan Z, Tian Z, Duan X, Ma J, Jiang Z, Hou J. Design of probiotic delivery systems and their therapeutic effects on targeted tissues. J Control Release 2024; 375:20-46. [PMID: 39214316 DOI: 10.1016/j.jconrel.2024.08.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 08/14/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
The microbiota at different sites in the body is closely related to disease. The intake of probiotics is an effective strategy to alleviate diseases and be adjuvant in their treatment. However, probiotics may suffer from harsh environments and colonization resistance, making it difficult to maintain a sufficient number of live probiotics to reach the target sites and exert their original probiotic effects. Encapsulation of probiotics is an effective strategy. Therefore, probiotic delivery systems, as effective methods, have been continuously developed and innovated to ensure that probiotics are effectively delivered to the targeted site. In this review, initially, the design of probiotic delivery systems is reviewed from four aspects: probiotic characteristics, processing technologies, cell-derived wall materials, and interactions between wall materials. Subsequently, the review focuses on the effects of probiotic delivery systems that target four main microbial colonization sites: the oral cavity, skin, intestine, and vagina, as well as disease sites such as tumors. Finally, this review also discusses the safety concerns of probiotic delivery systems in the treatment of disease and the challenges and limitations of implementing this method in clinical studies. It is necessary to conduct more clinical studies to evaluate the effectiveness of different probiotic delivery systems in the treatment of diseases.
Collapse
Affiliation(s)
- Cong Xu
- College of Food Science and Engineering, Guiyang University, Guiyang 550005, China; Key Laboratory of Dairy Science, Northeast Agricultural University, College of Food Science, Harbin 150030, China; Heilongjiang Green Food Science Research Institute, Harbin 150028, China
| | - Jiahui Guo
- Key Laboratory of Dairy Science, Northeast Agricultural University, College of Food Science, Harbin 150030, China
| | - Baoyue Chang
- Key Laboratory of Dairy Science, Northeast Agricultural University, College of Food Science, Harbin 150030, China
| | - Yiming Zhang
- Department of Psychiatry and Mental Health, Dalian Medical University, Dalian 116044, China
| | - Zhongmei Tan
- Key Laboratory of Dairy Science, Northeast Agricultural University, College of Food Science, Harbin 150030, China
| | - Zihao Tian
- Key Laboratory of Dairy Science, Northeast Agricultural University, College of Food Science, Harbin 150030, China
| | - Xiaolei Duan
- Key Laboratory of Dairy Science, Northeast Agricultural University, College of Food Science, Harbin 150030, China
| | - Jiage Ma
- Key Laboratory of Dairy Science, Northeast Agricultural University, College of Food Science, Harbin 150030, China; Heilongjiang Green Food Science Research Institute, Harbin 150028, China
| | - Zhanmei Jiang
- Key Laboratory of Dairy Science, Northeast Agricultural University, College of Food Science, Harbin 150030, China.
| | - Juncai Hou
- College of Food Science and Engineering, Guiyang University, Guiyang 550005, China; Key Laboratory of Dairy Science, Northeast Agricultural University, College of Food Science, Harbin 150030, China; Heilongjiang Green Food Science Research Institute, Harbin 150028, China.
| |
Collapse
|
5
|
Gu S, Zhao X, Wan F, Gu D, Xie W, Gao C. Intracellularly Gelated Macrophages Loaded with Probiotics for Therapy of Colitis. NANO LETTERS 2024; 24:13504-13512. [PMID: 39418594 DOI: 10.1021/acs.nanolett.4c02699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Probiotics therapy has garnered significant attention in the treatment of inflammatory bowel disease (IBD). However, a large number of oral administrated probiotics are inactivated after passing through the gastric acid environment, and their ability to colonize in the intestine is also weak. Herein, this study develops a novel probiotics formulation (GM-EcN) by incorporating Escherichia coli Nissle 1917 (EcN) into intracellularly gelated macrophages (GM). Intracellular hydrogel is designed to load and prevent EcN from digestion in gastric juice, and GM acts as a macrophage-like carrier to carry the attached probiotics to colonize in the inflammatory intestine. In addition, hydrogel serves as an ideal cytoskeletal structure to maintain the intact cell morphology and membrane structure of GM, comparable to source macrophages. Due to the receptor-ligand interaction, inflammation-related membrane proteins enable GM as a cell sponge to sequestrate and neutralize multiple inflammatory cytokines. In vivo treatment demonstrates that GM-EcN efficiently alleviates IBD symptoms and enhances gut microbiota recovery.
Collapse
Affiliation(s)
- Siyao Gu
- State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Open FIESTA Center, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Shenzhen Key Laboratory of Health Science and Technology, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Xiaona Zhao
- Department of Laboratory Medicine, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
- Guangxi University of Chinese Medicine, 530004 Nanning, China
| | - Fang Wan
- State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Shenzhen Key Laboratory of Health Science and Technology, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Dayong Gu
- Department of Laboratory Medicine, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Weidong Xie
- State Key Laboratory of Chemical Oncogenomics, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Open FIESTA Center, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Shenzhen Key Laboratory of Health Science and Technology, Institute of Biopharmaceutical and Health Engineering, Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Cheng Gao
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, P. R. China
| |
Collapse
|
6
|
Sun C, Wang S, Xu Y, Wang S, Zhou D, Liu H. Enhancing Lactobacillus plantarum delivery: Impact of gluconolactone concentration on high-internal-phase emulsion gels and gastrointestinal viability. Food Chem 2024; 455:139928. [PMID: 38850974 DOI: 10.1016/j.foodchem.2024.139928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/18/2024] [Accepted: 05/30/2024] [Indexed: 06/10/2024]
Abstract
In this study, the impact of Gluconolactone (GDL) concentration on the formation of high-internal-phase emulsion gels (HIPEGs) and the gastrointestinal digestive viability of Lactobacillus plantarum encapsulated within these HIPEGs were demonstrated. Increasing GDL concentrations led to cross-linking of particles at the oil-water interface, thereby stabilizing smaller oil droplets. The addition of GDL to HIPEs results in a significant increase in the secondary structure of SPI, specifically in β-sheet and β-turn formations, accompanied by a reduction in α-helix percentage. This alteration enhanced the binding effect of protein on water, leading to changes in intermolecular force. Notably, HIPEGs containing 3.0% GDL demonstrated superior encapsulation efficiency and delivery efficiency, reaching 99.0% and 84.5%, respectively. After 14 d of continuous zebrafishs feeding, the intestinal viable cells count of Lactobacillus plantarum reached 1.18 × 107 CFU/mL. This finding supports the potential use of HIPEGs as a probiotic delivery carrier, effectively enhancing the intestinal colonization rate.
Collapse
Affiliation(s)
- Chenyuan Sun
- College of Food Science and Technology, Bohai University, Jinzhou 121013, China..
| | - Shengnan Wang
- College of Food Science and Technology, Bohai University, Jinzhou 121013, China.; Grain and Cereal Food Bio-efficient Transformation Engineering Research Center of Liaoning Province, Jinzhou 121013, China..
| | - Yan Xu
- College of Food Science and Technology, Bohai University, Jinzhou 121013, China
| | - Shumin Wang
- College of Food Science and Technology, Bohai University, Jinzhou 121013, China
| | - Dayu Zhou
- College of Food Science and Technology, Bohai University, Jinzhou 121013, China
| | - He Liu
- College of Food Science and Technology, Bohai University, Jinzhou 121013, China.; Grain and Cereal Food Bio-efficient Transformation Engineering Research Center of Liaoning Province, Jinzhou 121013, China
| |
Collapse
|
7
|
Daisley BA, Allen-Vercoe E. Microbes as medicine. Ann N Y Acad Sci 2024. [PMID: 39392836 DOI: 10.1111/nyas.15237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Abstract
Over the last two decades, advancements in sequencing technologies have significantly deepened our understanding of the human microbiome's complexity, leading to increased concerns about the detrimental effects of antibiotics on these intricate microbial ecosystems. Concurrently, the rise in antimicrobial resistance has intensified the focus on how beneficial microbes can be harnessed to treat diseases and improve health and offer potentially promising alternatives to traditional antibiotic treatments. Here, we provide a comprehensive overview of both established and emerging microbe-centric therapies, from probiotics to advanced microbial ecosystem therapeutics, examine the sophisticated ways in which microbes are used medicinally, and consider their impacts on microbiome homeostasis and health outcomes through a microbial ecology lens. In addition, we explore the concept of rewilding the human microbiome by reintroducing "missing microbes" from nonindustrialized societies and personalizing microbiome modulation to fit individual microbial profiles-highlighting several promising directions for future research. Ultimately, the advancements in sequencing technologies combined with innovative microbial therapies and personalized approaches herald a new era in medicine poised to address antibiotic resistance and improve health outcomes through targeted microbiome management.
Collapse
Affiliation(s)
- Brendan A Daisley
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Emma Allen-Vercoe
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
8
|
Zhao R, Yu T, Li J, Niu R, Liu D, Wang W. Single-cell encapsulation systems for probiotic delivery: Armor probiotics. Adv Colloid Interface Sci 2024; 332:103270. [PMID: 39142064 DOI: 10.1016/j.cis.2024.103270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 05/28/2024] [Accepted: 08/03/2024] [Indexed: 08/16/2024]
Abstract
Functional foods or drugs based on probiotics have gained unprecedented attention and development due to the increasingly clear relationship between probiotics and human health. Probiotics can regulate intestinal microbiota, dynamically participating in various physiological activities to directly affect human health. Some probiotic-based functional preparations have shown great potential in treating multiple refractory diseases. Currently, the survival and activity of probiotic cells in complex environments in vitro and in vivo have taken priority, and various encapsulation systems based on food-derived materials have been designed and constructed to protect and deliver probiotics. However, traditional encapsulation technology cannot achieve precise protection for a single probiotic, which makes it unable to have a significant effect after release. In this case, single-cell encapsulation systems can be assembled based on biological interfaces to protect and functionalize individual probiotic cells, maximizing their physiological activity. This review discussed the arduous challenges of probiotics in food processing, storage, human digestion, and the commonly used probiotic encapsulation system. Besides, a novel technology of probiotic encapsulation was introduced based on single-cell coating, namely, "armor probiotics". We focused on the classification, structural design, and functional characteristics of armor coatings, and emphasized the essential functional characteristics of armor probiotics in human health regulation, including regulating intestinal health and targeted bioimaging and treatment of diseased tissues. Subsequently, the benefits, limitations, potential challenges, as well as future direction of armor probiotics were put forward. We hope this review may provide new insights and ideas for developing a single-cell probiotics encapsulating system.
Collapse
Affiliation(s)
- Runan Zhao
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China
| | - Ting Yu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China
| | - Jiaheng Li
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China
| | - Ruihao Niu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China
| | - Donghong Liu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China; Ningbo Research Institute, Zhejiang University, Ningbo 315100, China
| | - Wenjun Wang
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang Engineering Laboratory of Food Technology and Equipment, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China.
| |
Collapse
|
9
|
Hu F, Gao Q, Zheng C, Zhang W, Yang Z, Wang S, Zhang Y, Lu T. Encapsulated lactiplantibacillus plantarum improves Alzheimer's symptoms in APP/PS1 mice. J Nanobiotechnology 2024; 22:582. [PMID: 39304919 PMCID: PMC11414319 DOI: 10.1186/s12951-024-02862-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder that can result in neurotoxicity and an imbalance in gut microbiota. Probiotics have been shown to play an important role in regulating the gut microbiota, but their viability and bioactivity are often compromised as they traverse the gastrointestinal tract, thereby reducing their efficacy and limiting their clinical utility. RESULTS In this work, layer-by-layer (LbL) encapsulation technology was used to encapsulate Lactiplantibacillus plantarum (LP) to improve the above shortcomings. Studies in APPswe/PS1dE9 (APP/PS1) transgenic mice show that LbL-encapsulated LP ((CS/SP)2-LP) protects LP from gastrointestinal damage while (CS/SP)2-LP treatment It improves brain neuroinflammation and neuronal damage in AD mice, reduces Aβ deposition, improves tau protein phosphorylation levels, and restores intestinal barrier damage in AD mice. In addition, post-synaptic density protein 95 (PSD-95) expression increased in AD mice after treatment, indicating enhanced synaptic plasticity. Fecal metabolomic and microbiological analyzes showed that the disordered intestinal microbiota composition of AD mice was restored and short-chain fatty acids (SCFAs) levels were significantly increased after (CS/SP)2-LP treatment. CONCLUSION Overall, the above evidence suggests that (CS/SP)2-LP can improve AD symptoms by restoring the balance of intestinal microbiota, and (CS/SP)2-LP treatment will provide a new method to improve the symptoms of AD patients.
Collapse
Affiliation(s)
- Fangfang Hu
- Key Laboratory of Space Bioscience and Biotechnology, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Qian Gao
- Key Laboratory of Space Bioscience and Biotechnology, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Caiyun Zheng
- Key Laboratory of Space Bioscience and Biotechnology, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Wenhui Zhang
- Key Laboratory of Space Bioscience and Biotechnology, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Ziyi Yang
- Key Laboratory of Space Bioscience and Biotechnology, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Shihao Wang
- Key Laboratory of Space Bioscience and Biotechnology, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Yanni Zhang
- Key Laboratory of Space Bioscience and Biotechnology, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China.
| | - Tingli Lu
- Key Laboratory of Space Bioscience and Biotechnology, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, P. R. China.
| |
Collapse
|
10
|
Chu L, Xie L, Chen B, Jiang Y, Wang W. Carboxymethyl chitosan-dialdehyde glucan/polydopamine carrier targeted delivery Bacillus subtilis on enhancing oral utilization and intestinal colonization in mice. Int J Biol Macromol 2024; 280:135574. [PMID: 39270914 DOI: 10.1016/j.ijbiomac.2024.135574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/12/2024] [Accepted: 09/10/2024] [Indexed: 09/15/2024]
Abstract
Most probiotics are difficult to resist the invasion of gastrointestinal physiological and pathological environments, which limits their beneficial effects. The design of a pH-responsive and adhesive double-layer carrier (Carboxymethyl chitosan polyaldehyde polysaccharide, CMCS-DHG/PDA) aims to safeguard the activity of probiotics and enhance their intestinal colonization. The results obtained from UV-vis spectroscopy and XPS analysis revealed the formation of a polydopamine nanocoating surrounding Bacillus subtilis, and the outer carrier formed a Schiff base covalent bond, providing sufficient mechanical properties for the carrier. The carrier exhibited a significantly higher degree of swelling under pH 1.2 compared to pH 7.4, indicating its pronounced pH responsiveness. The CMCS-DHG/PDA carrier not only provided protection for B. subtilis against simulated digestive fluids, but also improved its tolerance to bile and antibiotics. In addition, carrier-protected probiotics showed extraordinary mucosal adhesion, which could significantly improve oral bioavailability and intestinal colonization. Finally, the impact of carrier-protected B. subtilis on gut microbiota was explored, revealing that the carrier protected B. subtilis could significantly improve the diversity, richness, and composition of gut microbiota. Concurrently, it promoted the formation of short chain fatty acids, creating a more beneficial environment for intestinal health.
Collapse
Affiliation(s)
- Lulu Chu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Luyu Xie
- Institute of Dataspace, Hefei Comprehensive National Science Center, Hefei 230000, China
| | - Bingzhi Chen
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Yuji Jiang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China.
| | - Wenjie Wang
- College of Life Science, Linyi University, Linyi 276000, China.
| |
Collapse
|
11
|
Zhai Z, Wang X, Qian Z, Wang A, Zhao W, Xiong J, Wang J, Wang Y, Cao H. Lactobacillus rhamnosus GG coating with nanocomposite ameliorates intestinal inflammation. Biomed Pharmacother 2024; 178:117197. [PMID: 39084077 DOI: 10.1016/j.biopha.2024.117197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/08/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
The steady increase in the prevalence of inflammatory bowel disease (IBD) is regarded as a worldwide health issue. Gut microorganisms could modulate host immune and metabolic status and are associated with health effects. Probiotics, Lactobacillus rhamnosus GG (LGG), are beneficial microorganisms that ameliorate disease and exert advantageous effects on intestinal homeostasis. However, the viability of probiotics will suffer from various risk factors in the digestive tract. In this view, we developed a probiotic coating with nanocomposite using tannic acid (TA) and casein phosphopeptide (CPP) through layer-by-layer technology to overcome the challenges after oral administration. LGG showed an improved survival rate in simulated gastrointestinal conditions after coated. The coating (LGG/TA-Mg2+/CPP) had potent reactive oxygen species (ROS) scavenging ability and improved the survival rate of colorectal epithelial cells after H2O2 stimulation. In DSS-induced colitis, administration of LGG/TA-Mg2+/CPP ameliorated intestinal inflammation and reduced the disruption of barrier function. Furthermore, LGG/TA-Mg2+/CPP increased the abundance and diversity of the gut microbiota. In the mouse model of DSS colitis, LGG/TA-Mg2+/CPP can better activate the EGFR/AKT signaling pathway, thereby protecting the epithelial barrier function of the colon epithelium. In conclusion, the probiotic coating with nanocomposite may become a delivery platform for probiotics applied to IBD.
Collapse
Affiliation(s)
- Zihan Zhai
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China; School of Health, Binzhou Polytechnic, Binzhou, China
| | - Xin Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China
| | - Zhanying Qian
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Aili Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China; Department of Gastroenterology, Binzhou Medical University Hospital (BMUH), No. 662 Huanghe 2nd Road, Binzhou City, Shandong Province, China
| | - Wenjing Zhao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China
| | - Jie Xiong
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China
| | - Jingyi Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China
| | - Yinsong Wang
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China.
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, National Key Clinical Specialty, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China.
| |
Collapse
|
12
|
Dipta SS, Christofferson AJ, Kumar PV, Kundi V, Hanif M, Tang J, Flores N, Kalantar‐Zadeh K, Uddin A, Rahim MA. Stable and Lead-Safe Polyphenol-Encapsulated Perovskite Solar Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403057. [PMID: 38889238 PMCID: PMC11336907 DOI: 10.1002/advs.202403057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 05/17/2024] [Indexed: 06/20/2024]
Abstract
Lead (Pb) halide perovskite solar cells (PSCs) exhibit impressive power conversion efficiencies close to those of their silicon counterparts. However, they suffer from moisture instability and Pb safety concerns. Previous studies have endeavoured to address these issues independently, yielding minimal advancements. Here, a general nanoencapsulation platform using natural polyphenols is reported for Pb-halide PSCs that simultaneously addresses both challenges. The polyphenol-based encapsulant is solution-processable, inexpensive (≈1.6 USD m-2), and requires only 5 min for the entire process, highlighting its potential scalability. The encapsulated devices with a power conversion efficiency of 20.7% retained up to 80% of their peak performance for 2000 h and up to 70% for 7000 h. Under simulated rainfall conditions, the encapsulant rich in catechol groups captures the Pb ions released from the degraded perovskites via coordination, keeping the Pb levels within the safe drinking water threshold of 15 ppb.
Collapse
Affiliation(s)
- Shahriyar Safat Dipta
- School of Photovoltaic and Renewable Energy EngineeringUniversity of New South WalesSydneyNew South Wales2052Australia
| | | | - Priyank V. Kumar
- School of Chemical EngineeringUniversity of New South Wales (UNSW)SydneyNew South Wales2052Australia
| | - Varun Kundi
- School of Chemical EngineeringUniversity of New South Wales (UNSW)SydneyNew South Wales2052Australia
| | - Muhammad Hanif
- School of Photovoltaic and Renewable Energy EngineeringUniversity of New South WalesSydneyNew South Wales2052Australia
| | - Jianbo Tang
- School of Chemical EngineeringUniversity of New South Wales (UNSW)SydneyNew South Wales2052Australia
| | - Nieves Flores
- School of Chemical and Biomolecular EngineeringUniversity of SydneySydneyNew South Wales2006Australia
| | - Kourosh Kalantar‐Zadeh
- School of Chemical EngineeringUniversity of New South Wales (UNSW)SydneyNew South Wales2052Australia
- School of Chemical and Biomolecular EngineeringUniversity of SydneySydneyNew South Wales2006Australia
| | - Ashraf Uddin
- School of Photovoltaic and Renewable Energy EngineeringUniversity of New South WalesSydneyNew South Wales2052Australia
| | - Md. Arifur Rahim
- School of Chemical EngineeringUniversity of New South Wales (UNSW)SydneyNew South Wales2052Australia
- School of Chemical and Biomolecular EngineeringUniversity of SydneySydneyNew South Wales2006Australia
- Department of Chemical and Biological EngineeringMonash UniversityClaytonVictoria3800Australia
| |
Collapse
|
13
|
Jung YH, Chae CW, Han HJ. The potential role of gut microbiota-derived metabolites as regulators of metabolic syndrome-associated mitochondrial and endolysosomal dysfunction in Alzheimer's disease. Exp Mol Med 2024; 56:1691-1702. [PMID: 39085351 PMCID: PMC11372123 DOI: 10.1038/s12276-024-01282-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/20/2024] [Accepted: 05/10/2024] [Indexed: 08/02/2024] Open
Abstract
Although the role of gut microbiota (GMB)-derived metabolites in mitochondrial and endolysosomal dysfunction in Alzheimer's disease (AD) under metabolic syndrome remains unclear, deciphering these host-metabolite interactions represents a major public health challenge. Dysfunction of mitochondria and endolysosomal networks (ELNs) plays a crucial role in metabolic syndrome and can exacerbate AD progression, highlighting the need to study their reciprocal regulation for a better understanding of how AD is linked to metabolic syndrome. Concurrently, metabolic disorders are associated with alterations in the composition of the GMB. Recent evidence suggests that changes in the composition of the GMB and its metabolites may be involved in AD pathology. This review highlights the mechanisms of metabolic syndrome-mediated AD development, focusing on the interconnected roles of mitochondrial dysfunction, ELN abnormalities, and changes in the GMB and its metabolites. We also discuss the pathophysiological role of GMB-derived metabolites, including amino acids, fatty acids, other metabolites, and extracellular vesicles, in mediating their effects on mitochondrial and ELN dysfunction. Finally, this review proposes therapeutic strategies for AD by directly modulating mitochondrial and ELN functions through targeting GMB metabolites under metabolic syndrome.
Collapse
Affiliation(s)
- Young Hyun Jung
- Department of Physiology, College of Medicine, Soonchunhyang University, Cheonan, 31151, Korea
| | - Chang Woo Chae
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, South Korea.
| |
Collapse
|
14
|
Li C, Wang ZX, Xiao H, Wu FG. Intestinal Delivery of Probiotics: Materials, Strategies, and Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310174. [PMID: 38245861 DOI: 10.1002/adma.202310174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/04/2024] [Indexed: 01/22/2024]
Abstract
Probiotics with diverse and crucial properties and functions have attracted broad interest from many researchers, who adopt intestinal delivery of probiotics to modulate the gut microbiota. However, the major problems faced for the therapeutic applications of probiotics are the viability and colonization of probiotics during their processing, oral intake, and subsequent delivery to the gut. The challenges of simple oral delivery (stability, controllability, targeting, etc.) have greatly limited the use of probiotics in clinical therapies. Nanotechnology can endow the probiotics to be delivered to the intestine with improved survival rate and increased resistance to the adverse environment. Additionally, the progress in synthetic biology has created new opportunities for efficiently and purposefully designing and manipulating the probiotics. In this article, a brief overview of the types of probiotics for intestinal delivery, the current progress of different probiotic encapsulation strategies, including the chemical, physical, and genetic strategies and their combinations, and the emerging single-cell encapsulation strategies using nanocoating methods, is presented. The action mechanisms of probiotics that are responsible for eliciting beneficial effects are also briefly discussed. Finally, the therapeutic applications of engineered probiotics are discussed, and the future trends toward developing engineered probiotics with advanced features and improved health benefits are proposed.
Collapse
Affiliation(s)
- Chengcheng Li
- International Innovation Center for Forest Chemicals and Materials and Jiangsu Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing, 210037, China
| | - Zi-Xi Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Huining Xiao
- Department of Chemical Engineering, University of New Brunswick, Fredericton, New Brunswick, E3B 5A3, Canada
| | - Fu-Gen Wu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| |
Collapse
|
15
|
Ramazanidoroh F, Hosseininezhad M, Shahrampour D, Wu X. Edible Packaging as a Functional Carrier of Prebiotics, Probiotics, and Postbiotics to Boost Food Safety, Quality, and Shelf Life. Probiotics Antimicrob Proteins 2024; 16:1327-1347. [PMID: 37389789 DOI: 10.1007/s12602-023-10110-5] [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] [Accepted: 06/08/2023] [Indexed: 07/01/2023]
Abstract
The safety limitations of chemical preservatives led to an increasing trend among industries and customers toward preservative-free foods; hence, the necessity has arisen for developing innovative, safe antimicrobial elements to prolong the shelf life. Beneficial microorganisms that are described as probiotics and also their metabolites are increasingly being considered as bioprotective agents. These microorganisms could be beneficial for extending food shelf-life and boosting human health. During distribution and storage (25 °C or 4 °C), they could contribute to suppressing unwanted microbes and then improving food safety and quality. Also, by tolerating the harsh conditions of gastrointestinal tract (low pH (~3), presence of bile salts, digestive enzymes, competition with other microbes, etc.), probiotics could exert several biological effects at the host. Besides inclusion in foods and supplements, probiotics and their functional metabolites could be delivered via edible packaging (EP). Recent studies have demonstrated the strong potential of pre/pro/post-biotic EP in food biopreservation. These packaging systems may show different potency of food biopreservation. Among others, postbiotics, as metabolic by-products of probiotics, have gained tremendous attention among researchers due to their unique properties like presenting a variety of antimicrobial activities, convenience in use in different industrial stages and commercialization, extended shelf life, and stability in a wide range of pH and temperature. In addition to antimicrobial activities, various bio-EP could differently influence physical or sensorial attributes of food commodities, impacting their acceptance by consumers. Hence, this study is aimed at presenting a comprehensive review of the application of bio-EP, not only by providing a protective barrier against physical damage but also by creating a controlled atmosphere to improve the health and shelf life of food.
Collapse
Affiliation(s)
- Fahimeh Ramazanidoroh
- Department of Food Biotechnology, Research Institute of Food Science and Technology (RIFST), Mashhad, Iran
| | - Marzieh Hosseininezhad
- Department of Food Biotechnology, Research Institute of Food Science and Technology (RIFST), Mashhad, Iran.
| | - Dina Shahrampour
- Department of Food Safety and Quality Control, Research Institute of Food Science and Technology (RIFST), Mashhad, Iran
| | - Xiyang Wu
- Department of Food Science and Engineering, Jinan University, Guangzhou, China
| |
Collapse
|
16
|
Yang X, Nie W, Wang C, Fang Z, Shang L. Microfluidic-based multifunctional microspheres for enhanced oral co-delivery of probiotics and postbiotics. Biomaterials 2024; 308:122564. [PMID: 38581763 DOI: 10.1016/j.biomaterials.2024.122564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/16/2024] [Accepted: 04/01/2024] [Indexed: 04/08/2024]
Abstract
Probiotic-based therapies have shown great potential in the prevention and treatment of many diseases by positively regulating intestinal flora homeostasis. However, the efficacy of oral probiotics is severely limited due to the loss of bioactivity, short intestinal retention time, and insufficient therapeutic effect. Here, based on droplet microfluidics, we developed a hydrogel microsphere with colonic targeting and mucoadhesive capabilities as a multifunctional delivery platform, which can be used for co-delivery of probiotics (Escherichia coli Nissle 1917, EcN) and auxiliary molecules (indole-3-propionic acid, IPA), achieving synergistic therapeutic effects. In vivo studies shown that the integrated multifunctional microspheres can significantly reduce intestinal inflammation, repair intestinal barrier function, enhance probiotic colonization in the intestine, and modulate disordered intestinal flora, demonstrating enhanced therapeutic effects in a mouse model of colitis. This work reveals that microfluidic-based smart droplet microspheres can provide a versatile platform for advanced microbial therapies.
Collapse
Affiliation(s)
- Xinyuan Yang
- Shanghai Xuhui Central Hospital, 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 Sciences, Fudan University, Shanghai, 200032, China
| | - Weimin Nie
- Shanghai Xuhui Central Hospital, 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 Sciences, Fudan University, Shanghai, 200032, China
| | - Chong Wang
- Shanghai Xuhui Central Hospital, 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 Sciences, Fudan University, Shanghai, 200032, China
| | - Zhonglin Fang
- Shanghai Xuhui Central Hospital, 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 Sciences, Fudan University, Shanghai, 200032, China
| | - Luoran Shang
- Shanghai Xuhui Central Hospital, 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 Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
17
|
Wang Q, Jin L, Yang H, Yu L, Cao X, Mao Z. Bacteria/Nanozyme Composites: New Therapeutics for Disease Treatment. SMALL METHODS 2024:e2400610. [PMID: 38923867 DOI: 10.1002/smtd.202400610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/11/2024] [Indexed: 06/28/2024]
Abstract
Bacterial therapy is recognized as a cost-effective treatment for several diseases. However, its development is hindered by limited functionality, weak inherent therapeutic effects, and vulnerability to harsh microenvironmental conditions, leading to suboptimal treatment activity. Enhancing bacterial activity and therapeutic outcomes emerges as a pivotal challenge. Nanozymes have garnered significant attention due to their enzyme-mimic activities and high stability. They enable bacteria to mimic the functions of gene-edited bacteria expressing the same functional enzymes, thereby improving bacterial activity and therapeutic efficacy. This review delineates the therapeutic mechanisms of bacteria and nanozymes, followed by a summary of strategies for preparing bacteria/nanozyme composites. Additionally, the synergistic effects of such composites in biomedical applications such as gastrointestinal diseases and tumors are highlighted. Finally, the challenges of bacteria/nanozyme composites are discussed and propose potential solutions. This study aims to provide valuable insights to offer theoretical guidance for the advancement of nanomaterial-assisted bacterial therapy.
Collapse
Affiliation(s)
- Qirui Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Lulu Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Huang Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Lisha Yu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Xinran Cao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transvascular Implantation Devices, Zhejiang, Hangzhou, 310009, China
| |
Collapse
|
18
|
D'Amico V, Lopalco A, Iacobazzi RM, Vacca M, Siragusa S, De Angelis M, Lopedota AA, Denora N. Multistimuli responsive microcapsules produced by the prilling/vibration technique for targeted colonic delivery of probiotics. Int J Pharm 2024; 658:124223. [PMID: 38744413 DOI: 10.1016/j.ijpharm.2024.124223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024]
Abstract
This study aimed to microencapsulate the probiotic strain Lactiplantibacillus plantarum 4S6R (basonym Lactobacillus plantarum) in both microcapsules and microspheres by prilling/vibration technique. A specific polymeric mixture, selected for its responsiveness to parallel colonic stimuli, was individuated as a carrier of microparticles. Although the microspheres were consistent with some critical quality parameters, they showed a low encapsulation efficiency and were discarded. The microcapsules produced demonstrated high yields (97.52%) and encapsulation efficiencies (90.06%), with dimensional analysis and SEM studies confirming the desired size morphology and structure. The results of thermal stress tests indicate the ability of the microcapsules to protect the probiotic. Stability studies showed a significant advantage of the microcapsules over non-encapsulated probiotics, with greater stability over time. The release study under simulated gastrointestinal conditions demonstrated the ability of the microcapsules to protect the probiotics from gastric acid and bile salts, ensuring their viability. Examination in a simulated faecal medium revealed the ability of the microcapsules to release the bacteria into the colon, enhancing their beneficial impact on gut health. This research suggests that the selected mixture of reactive polymers holds promise for improving the survival and efficacy of probiotics in the gastrointestinal tract, paving the way for the development of advanced probiotic products.
Collapse
Affiliation(s)
- Vita D'Amico
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari "Aldo Moro", 4, E. Orabona Street, 70125 Bari, Italy
| | - Antonio Lopalco
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari "Aldo Moro", 4, E. Orabona Street, 70125 Bari, Italy
| | - Rosa Maria Iacobazzi
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari "Aldo Moro", 4, E. Orabona Street, 70125 Bari, Italy
| | - Mirco Vacca
- Department of Soil, Plant and Food Sciences, University of Bari "Aldo Moro", 165/A, G. Amendola Street, 70126 Bari, Italy
| | - Sonya Siragusa
- Department of Soil, Plant and Food Sciences, University of Bari "Aldo Moro", 165/A, G. Amendola Street, 70126 Bari, Italy
| | - Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari "Aldo Moro", 165/A, G. Amendola Street, 70126 Bari, Italy
| | - Angela Assunta Lopedota
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari "Aldo Moro", 4, E. Orabona Street, 70125 Bari, Italy.
| | - Nunzio Denora
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari "Aldo Moro", 4, E. Orabona Street, 70125 Bari, Italy
| |
Collapse
|
19
|
Hou Y, Zhu L, Ye X, Ke Q, Zhang Q, Xie X, Piao JG, Wei Y. Integrated oral microgel system ameliorates renal fibrosis by hitchhiking co-delivery and targeted gut flora modulation. J Nanobiotechnology 2024; 22:305. [PMID: 38822364 PMCID: PMC11143587 DOI: 10.1186/s12951-024-02586-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/26/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND Renal fibrosis is a progressive process associated with chronic kidney disease (CKD), contributing to impaired kidney function. Active constituents in traditional Chinese herbs, such as emodin (EMO) and asiatic acid (AA), exhibit potent anti-fibrotic properties. However, the oral administration of EMO and AA results in low bioavailability and limited kidney accumulation. Additionally, while oral probiotics have been accepted for CKD treatment through gut microbiota modulation, a significant challenge lies in ensuring their viability upon administration. Therefore, our study aims to address both renal fibrosis and gut microbiota imbalance through innovative co-delivery strategies. RESULTS In this study, we developed yeast cell wall particles (YCWPs) encapsulating EMO and AA self-assembled nanoparticles (NPYs) and embedded them, along with Lactobacillus casei Zhang, in chitosan/sodium alginate (CS/SA) microgels. The developed microgels showed significant controlled release properties for the loaded NPYs and prolonged the retention time of Lactobacillus casei Zhang (L. casei Zhang) in the intestine. Furthermore, in vivo biodistribution showed that the microgel-carried NPYs significantly accumulated in the obstructed kidneys of rats, thereby substantially increasing the accumulation of EMO and AA in the impaired kidneys. More importantly, through hitchhiking delivery based on yeast cell wall and positive modulation of gut microbiota, our microgels with this synergistic strategy of therapeutic and modulatory interactions could regulate the TGF-β/Smad signaling pathway and thus effectively ameliorate renal fibrosis in unilateral ureteral obstruction (UUO) rats. CONCLUSION In conclusion, our work provides a new strategy for the treatment of renal fibrosis based on hitchhiking co-delivery of nanodrugs and probiotics to achieve synergistic effects of disease treatment and targeted gut flora modulation.
Collapse
Affiliation(s)
- Yu Hou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Lin Zhu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Xiaofeng Ye
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Qiaoying Ke
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Qibin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Xiaowei Xie
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Ji-Gang Piao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Yinghui Wei
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| |
Collapse
|
20
|
Agriopoulou S, Smaoui S, Chaari M, Varzakas T, Can Karaca A, Jafari SM. Encapsulation of Probiotics within Double/Multiple Layer Beads/Carriers: A Concise Review. Molecules 2024; 29:2431. [PMID: 38893306 PMCID: PMC11173482 DOI: 10.3390/molecules29112431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
An increased demand for natural products nowadays most specifically probiotics (PROs) is evident since it comes in conjunction with beneficial health effects for consumers. In this regard, it is well known that encapsulation could positively affect the PROs' viability throughout food manufacturing and long-term storage. This paper aims to analyze and review various double/multilayer strategies for encapsulation of PROs. Double-layer encapsulation of PROs by electrohydrodynamic atomization or electrospraying technology has been reported along with layer-by-layer assembly and water-in-oil-in-water (W1/O/W2) double emulsions to produce multilayer PROs-loaded carriers. Finally, their applications in food products are presented. The resistance and viability of loaded PROs to mechanical damage, during gastrointestinal transit and shelf life of these trapping systems, are also described. The PROs encapsulation in double- and multiple-layer coatings combined with other technologies can be examined to increase the opportunities for new functional products with amended functionalities opening a novel horizon in food technology.
Collapse
Affiliation(s)
- Sofia Agriopoulou
- Department of Food Science and Technology, University of the Peloponnese, Antikalamos, 24100 Kalamata, Greece;
| | - Slim Smaoui
- Laboratory of Microbial and Enzymatic Biotechnologies and Biomolecules, Center of Biotechnology of Sfax (CBS), University of Sfax, Road of Sidi Mansour Km 6, P.O. Box 1177, Sfax 3018, Tunisia; (S.S.); (M.C.)
| | - Moufida Chaari
- Laboratory of Microbial and Enzymatic Biotechnologies and Biomolecules, Center of Biotechnology of Sfax (CBS), University of Sfax, Road of Sidi Mansour Km 6, P.O. Box 1177, Sfax 3018, Tunisia; (S.S.); (M.C.)
| | - Theodoros Varzakas
- Department of Food Science and Technology, University of the Peloponnese, Antikalamos, 24100 Kalamata, Greece;
| | - Asli Can Karaca
- Department of Food Engineering, Faculty of Chemical and Metallurgical Engineering, Istanbul Technical University, 34469 Maslak, Turkey;
| | - Seid Mahdi Jafari
- Faculty of Food Science and Technology, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan 49138-15739, Iran
- Halal Research Center of IRI, Iran Food and Drug Administration, Ministry of Health and Medical Education, Tehran 14158-45371, Iran
| |
Collapse
|
21
|
Nguyen DT, Han SY, Kozlowski F, Seisenbaeva GA, Kessler VG, Kim BJ, Choi IS. Biphasic water-oil systems for functional augmentation of probiotic Lactobacillus acidophilus nanoencapsulated in luteolin-Fe 3+ shells. Chem Commun (Camb) 2024; 60:5330-5333. [PMID: 38666704 DOI: 10.1039/d4cc01603c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2024]
Abstract
Single-cell nanoencapsulation (SCNE) has great potential in the enhancement of therapeutic effects of probiotic microbes. However, the material scope has been limited to water-soluble compounds to avoid non-biocompatible organic solvents that are harmful to living cells. In this work, the SCNE of probiotic Lactobacillus acidophilus with water-insoluble luteolin and Fe3+ ions is achieved by the vortex-assisted, biphasic water-oil system. The process creates L. acidophilus nanoencapsulated in the luteolin-Fe3+ shells that empower the cells with extrinsic properties, such as resistance to lysozyme attack, anti-ROS ability, and α-amylase-inhibition activity, as well as sustaining viability under acidic conditions. The proposed protocol, embracing water-insoluble flavonoids as shell components in SCNE, will be an advanced add-on to the chemical toolbox for the manipulation of living cells at the single-cell level.
Collapse
Affiliation(s)
- Duc Tai Nguyen
- Department of Chemistry, KAIST, Daejeon 34141, Republic of Korea.
| | - Sang Yeong Han
- Department of Chemistry, KAIST, Daejeon 34141, Republic of Korea.
| | - Filip Kozlowski
- Department of Molecular Sciences, SLU, Uppsala 75007, Sweden
| | | | - Vadim G Kessler
- Department of Molecular Sciences, SLU, Uppsala 75007, Sweden
| | - Beom Jin Kim
- Department of Chemistry, University of Ulsan, Ulsan 44776, Republic of Korea
| | - Insung S Choi
- Department of Chemistry, KAIST, Daejeon 34141, Republic of Korea.
| |
Collapse
|
22
|
Fang T, Liu S. Metal-Phenolic Network Directed Coating of Single Probiotic Cell Followed by Photoinitiated Thiol-Ene Click Fortification to Enhance Oral Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308146. [PMID: 38054771 DOI: 10.1002/smll.202308146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/08/2023] [Indexed: 12/07/2023]
Abstract
Probiotics-based oral therapy has become a promising way to prevent and treat various diseases, while the application of probiotics is primarily restricted by loss of viability due to adverse conditions in the gastrointestinal (GI) tract during oral delivery. Layer-by-layer (LbL) single-cell encapsulation approaches are widely employed to improve the bioavailability of probiotics. However, they are generally time- and labor-intensive owing to multistep operation. Herein, a simple yet efficient LbL technique is developed to coat a model probiotic named Escherichia coli Nissle 1917 (EcN) through polyphenol-Ca2+ network directed allyl-modified gelatin (GelAGE) adsorption followed by cross-linking of GelAGE via photoinitiated thiol-ene click reaction to protect EcN from harsh microenvironments of GI tract. LbL single-cell encapsulation can be performed within 1 h through simple operation. It is revealed that coated EcN exhibits significantly improved viability against acidic gastric fluid and bile salts, and enhanced colonization in the intestinal tract without loss of proliferation capabilities. Furthermore, oral therapy of coated EcN remarkably relieves the pathological symptoms associated with colitis in mice including down-regulating inflammation, repairing epithelial barriers, scavenging reactive oxygen species (ROS), and restoring the homeostasis of gut microbiota. This simplified LbL coating strategy has great potential for various probiotics-mediated biomedical and nutraceutical applications.
Collapse
Affiliation(s)
- Taisong Fang
- Department of Food Science and Nutrition, Innovation Center of Yangtze River Delta, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Songbai Liu
- Department of Food Science and Nutrition, Innovation Center of Yangtze River Delta, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China
| |
Collapse
|
23
|
Han J, McClements DJ, Liu X, Liu F. Oral delivery of probiotics using single-cell encapsulation. Compr Rev Food Sci Food Saf 2024; 23:e13322. [PMID: 38597567 DOI: 10.1111/1541-4337.13322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 02/01/2024] [Accepted: 02/28/2024] [Indexed: 04/11/2024]
Abstract
Adequate intake of live probiotics is beneficial to human health and wellbeing because they can help treat or prevent a variety of health conditions. However, the viability of probiotics is reduced by the harsh environments they experience during passage through the human gastrointestinal tract (GIT). Consequently, the oral delivery of viable probiotics is a significant challenge. Probiotic encapsulation provides a potential solution to this problem. However, the production methods used to create conventional encapsulation technologies often damage probiotics. Moreover, the delivery systems produced often do not have the required physicochemical attributes or robustness for food applications. Single-cell encapsulation is based on forming a protective coating around a single probiotic cell. These coatings may be biofilms or biopolymer layers designed to protect the probiotic from the harsh gastrointestinal environment, enhance their colonization, and introduce additional beneficial functions. This article reviews the factors affecting the oral delivery of probiotics, analyses the shortcomings of existing encapsulation technologies, and highlights the potential advantages of single-cell encapsulation. It also reviews the various approaches available for single-cell encapsulation of probiotics, including their implementation and the characteristics of the delivery systems they produce. In addition, the mechanisms by which single-cell encapsulation can improve the oral bioavailability and health benefits of probiotics are described. Moreover, the benefits, limitations, and safety issues of probiotic single-cell encapsulation technology for applications in food and beverages are analyzed. Finally, future directions and potential challenges to the widespread adoption of single-cell encapsulation of probiotics are highlighted.
Collapse
Affiliation(s)
- Jiaqi Han
- College of Food Science and Engineering, Northwest A&F University, Xianyang, Shaanxi, China
| | - David Julian McClements
- Department of Food Science, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Xianyang, Shaanxi, China
| | - Fuguo Liu
- College of Food Science and Engineering, Northwest A&F University, Xianyang, Shaanxi, China
| |
Collapse
|
24
|
Mosquera FEC, Lizcano Martinez S, Liscano Y. Effectiveness of Psychobiotics in the Treatment of Psychiatric and Cognitive Disorders: A Systematic Review of Randomized Clinical Trials. Nutrients 2024; 16:1352. [PMID: 38732599 PMCID: PMC11085935 DOI: 10.3390/nu16091352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 04/23/2024] [Accepted: 04/28/2024] [Indexed: 05/13/2024] Open
Abstract
In this study, a systematic review of randomized clinical trials conducted from January 2000 to December 2023 was performed to examine the efficacy of psychobiotics-probiotics beneficial to mental health via the gut-brain axis-in adults with psychiatric and cognitive disorders. Out of the 51 studies involving 3353 patients where half received psychobiotics, there was a notably high measurement of effectiveness specifically in the treatment of depression symptoms. Most participants were older and female, with treatments commonly utilizing strains of Lactobacillus and Bifidobacteria over periods ranging from 4 to 24 weeks. Although there was a general agreement on the effectiveness of psychobiotics, the variability in treatment approaches and clinical presentations limits the comparability and generalization of the findings. This underscores the need for more personalized treatment optimization and a deeper investigation into the mechanisms through which psychobiotics act. The research corroborates the therapeutic potential of psychobiotics and represents progress in the management of psychiatric and cognitive disorders.
Collapse
Affiliation(s)
- Freiser Eceomo Cruz Mosquera
- Grupo de Investigación en Salud Integral (GISI), Departamento Facultad de Salud, Universidad Santiago de Cali, Cali 760035, Colombia
| | - Santiago Lizcano Martinez
- Área Servicio de Alimentación, Área Nutrición Clínica Hospitalización UCI Urgencias Y Equipo de Soporte nutricional, Clínica Nuestra, Cali 760041, Colombia;
| | - Yamil Liscano
- Grupo de Investigación en Salud Integral (GISI), Departamento Facultad de Salud, Universidad Santiago de Cali, Cali 760035, Colombia
| |
Collapse
|
25
|
Lei Y, Yan Y, Zhong J, Zhao Y, Xu Y, Zhang T, Xiong H, Chen Y, Wang X, Zhang K. Enterococcus durans 98D alters gut microbial composition and function to improve DSS-induced colitis in mice. Heliyon 2024; 10:e28486. [PMID: 38560132 PMCID: PMC10981110 DOI: 10.1016/j.heliyon.2024.e28486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
Enterococcus durans, is a potential functional strain with the capacity to regulate intestinal health and ameliorate colonic inflammation. However, the strain requires further investigation regarding its safety profile and potential mechanisms of colitis improvement. In this study, the safety of E. durans 98D (Ed) as a potential probiotic was studied using in vitro methods. Additionally, a dextran sulfate sodium (DSS)-induced murine colitis model was employed to investigate its impact on the intestinal microbiota and colitis. In vitro antimicrobial assays revealed Ed sensitivity to common antibiotics and its inhibitory effect on the growth of Escherichia coli O157, Streptococcus pneumoniae CCUG 37328, and Staphylococcus aureus ATCC 25923. To elucidate the functional properties of Ed, 24 weight-matched 6-week-old female C57BL/6J mice were randomly divided into three groups (n = 8): NC group, Con group (DSS), and Ed group (DSS + Ed). Ed administration demonstrated a protective effect on colitis mice, as evidenced by improvements in body weight, colonic length, reduced disease activity index, histological scores, diminished splenomegaly, and decreased goblet cell loss. Furthermore, Ed downregulated the expression of the pro-inflammatory cytokine genes (IL-6, IL-1β, and TNF-α) and upregulated the expression of the anti-inflammatory cytokine gene IL-10. The 16S rRNA gene sequencing revealed significant alterations in microbial α-diversity, with principal coordinate analysis indicating distinct differences in microbial composition among the three groups. At the phylum level, the relative abundance of Actinomycetota significantly increased in the Ed-treated group. At the genus level, Ed treatment markedly elevated the relative abundance of Paraprevotella, Rikenellaceae_RC9, and Odoribacter in DSS-induced colitis mice. In conclusion, Ed exhibits potential as a safe and effective therapeutic agent for DSS-induced colitis by reshaping the colonic microbiota.
Collapse
Affiliation(s)
| | | | - Junyu Zhong
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yitong Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yangbin Xu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Ting Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Hui Xiong
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yulin Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xiaolong Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Ke Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| |
Collapse
|
26
|
Fu Y, Li J, Cai W, Huang Y, Liu X, Ma Z, Tang Z, Bian X, Zheng J, Jiang J, Li C. The emerging tumor microbe microenvironment: From delineation to multidisciplinary approach-based interventions. Acta Pharm Sin B 2024; 14:1560-1591. [PMID: 38572104 PMCID: PMC10985043 DOI: 10.1016/j.apsb.2023.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/20/2023] [Accepted: 11/03/2023] [Indexed: 04/05/2024] Open
Abstract
Intratumoral microbiota has become research hotspots, and emerges as a non-negligent new component of tumor microenvironments (TME), due to its powerful influence on tumor initiation, metastasis, immunosurveillance and prognosis despite in low-biomass. The accumulations of microbes, and their related components and metabolites within tumor tissues, endow TME with additional pluralistic features which are distinct from the conventional one. Therefore, it's definitely necessary to comprehensively delineate the sophisticated landscapes of tumor microbe microenvironment, as well as their functions and related underlying mechanisms. Herein, in this review, we focused on the fields of tumor microbe microenvironment, including the heterogeneity of intratumor microbiota in different types of tumors, the controversial roles of intratumoral microbiota, the basic features of tumor microbe microenvironment (i.e., pathogen-associated molecular patterns (PAMPs), typical microbial metabolites, autophagy, inflammation, multi-faceted immunomodulation and chemoresistance), as well as the multidisciplinary approach-based intervention of tumor microbiome for cancer therapy by applying wild-type or engineered live microbes, microbiota metabolites, antibiotics, synthetic biology and rationally designed biomaterials. We hope our work will provide valuable insight to deeply understand the interplay of cancer-immune-microbial, and facilitate the development of microbes-based tumor-specific treatments.
Collapse
Affiliation(s)
- Yu Fu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Jia Li
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Wenyun Cai
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Yulan Huang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xinlong Liu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zhongyi Ma
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zhongjie Tang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xufei Bian
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Jiayun Jiang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chong Li
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
27
|
Hua Y, Wei Z, Xue C, Si J. Stability and programmed sequential release of Lactobacillus plantarum and curcumin encapsulated in bilayer-stabilized W 1/O/W 2 double emulsion: Effect of pectin as protective shell. Int J Biol Macromol 2024; 265:130805. [PMID: 38490382 DOI: 10.1016/j.ijbiomac.2024.130805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/05/2024] [Accepted: 03/10/2024] [Indexed: 03/17/2024]
Abstract
In order to overcome the problem that traditional W1/O/W2 double emulsions do not have targeted release performance, thereby better meeting the health needs of consumers, ovalbumin fibrils/pectin-based bilayer-stabilized double emulsion (OP-BDE) co-encapsulated with Lactobacillus plantarum and curcumin was constructed with pectin as the outer protective shell, which was expected to be used in the development of novel functional foods. The effects of pectin coating on the viability of Lactobacillus plantarum under conditions including storage, pasteurization, freeze-thaw cycles and in vitro simulated digestion were investigated. Results showed that pectin as protective shell could significantly enhance the tolerance of Lactobacillus plantarum to various environmental factors. Besides, the adsorption of pectin endowed OP-BDE with higher lipolysis and stronger protective effect on curcumin, remarkably improving the photostability and bioaccessibility of curcumin. In addition, in vitro simulated gastrointestinal release study indicated that OP-BDE possessed programmed sequential release property, allowing curcumin and Lactobacillus plantarum to be released in small intestine and colon, respectively. OP-BDE is the first reported co-delivery emulsion system with programmed release characteristic. This study provides new insights into OP-BDE in constructing co-delivery systems and programmed sequential release of active substances, and has potential reference and application value in actual food production.
Collapse
Affiliation(s)
- Yijie Hua
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266404, China
| | - Zihao Wei
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266404, China.
| | - Changhu Xue
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266404, China
| | - Jingyu Si
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266404, China
| |
Collapse
|
28
|
Altemimi AB, Farag HAM, Salih TH, Awlqadr FH, Al-Manhel AJA, Vieira IRS, Conte-Junior CA. Application of Nanoparticles in Human Nutrition: A Review. Nutrients 2024; 16:636. [PMID: 38474764 DOI: 10.3390/nu16050636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Nanotechnology in human nutrition represents an innovative advance in increasing the bioavailability and efficiency of bioactive compounds. This work delves into the multifaceted dietary contributions of nanoparticles (NPs) and their utilization for improving nutrient absorption and ensuring food safety. NPs exhibit exceptional solubility, a significant surface-to-volume ratio, and diameters ranging from 1 to 100 nm, rendering them invaluable for applications such as tissue engineering and drug delivery, as well as elevating food quality. The encapsulation of vitamins, minerals, and antioxidants within NPs introduces an innovative approach to counteract nutritional instabilities and low solubility, promoting human health. Nanoencapsulation methods have included the production of nanocomposites, nanofibers, and nanoemulsions to benefit the delivery of bioactive food compounds. Nutrition-based nanotechnology and nanoceuticals are examined for their economic viability and potential to increase nutrient absorption. Although the advancement of nanotechnology in food demonstrates promising results, some limitations and concerns related to safety and regulation need to be widely discussed in future research. Thus, the potential of nanotechnology could open new paths for applications and significant advances in food, benefiting human nutrition.
Collapse
Affiliation(s)
- Ammar B Altemimi
- Department of Food Science, College of Agriculture, University of Basrah, Basrah 61004, Iraq
- College of Medicine, University of Warith Al-Anbiyaa, Karbala 56001, Iraq
| | - Halgord Ali M Farag
- Halabja Research Center, Halabja Technical College Applied Science, Sulaimani Polytechnic University, Sulaimani 46002, Iraq
- Harem Research Center, Department of Nutrition and Diet Therapy, Harem Hospital, Sulaimani 46001, Iraq
| | - Tablo H Salih
- Halabja Research Center, Halabja Technical College Applied Science, Sulaimani Polytechnic University, Sulaimani 46002, Iraq
- Harem Research Center, Department of Nutrition and Diet Therapy, Harem Hospital, Sulaimani 46001, Iraq
| | - Farhang H Awlqadr
- Halabja Research Center, Halabja Technical College Applied Science, Sulaimani Polytechnic University, Sulaimani 46002, Iraq
| | | | - Italo Rennan Sousa Vieira
- Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-598, RJ, Brazil
| | - Carlos Adam Conte-Junior
- Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-598, RJ, Brazil
| |
Collapse
|
29
|
Cao F, Jin L, Zhang C, Gao Y, Qian Z, Wen H, Yang S, Ye Z, Hong L, Yang H, Tong Z, Cheng L, Ding Y, Wang W, Yu G, Mao Z, Chen X. Engineering Clinically Relevant Probiotics with Switchable "Nano-Promoter" and "Nano-Effector" for Precision Tumor Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2304257. [PMID: 37788635 DOI: 10.1002/adma.202304257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 09/07/2023] [Indexed: 10/05/2023]
Abstract
Probiotics have the potential as biotherapeutic agents for cancer management in preclinical models and human trials by secreting antineoplastic or immunoregulatory agents in the tumor microenvironment (TME). However, current probiotics lack the ability to dynamically respond to unique TME characteristics, leading to limited therapeutic accuracy and efficacy. Although progress has been made in customizing controllable probiotics through synthetic biology, the engineering process is complex and the predictability of production is relatively low. To address this, here, for the first time, this work adopts pH-dependent peroxidase-like (POD-like) artificial enzymes as both an inducible "nano-promoter" and "nano-effector" to engineer clinically relevant probiotics to achieve switchable control of probiotic therapy. The nanozyme initially serves as an inducible "nano-promoter," generating trace amounts of nonlethal reactive oxygen species (ROS) stress to upregulate acidic metabolites in probiotics. Once metabolites acidify the TME to a threshold, the nanozyme switches to a "nano-effector," producing a great deal of lethal ROS to fight cancer. This approach shows promise in subcutaneous, orthotopic, and colitis-associated colorectal cancer tumors, offering a new methodology for modulating probiotic metabolism in a pathological environment.
Collapse
Affiliation(s)
- Fangfang Cao
- Departments of Diagnostic Radiology Surgery Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Lulu Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Chenyin Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yong Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhefeng Qian
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, 310009, China
| | - Hongyang Wen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, 310009, China
| | - Sisi Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, 310003, China
| | - Ziqiang Ye
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Liangjie Hong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Huang Yang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zongrui Tong
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, 310009, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, 310009, China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, 310009, China
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine Zhejiang University, Hangzhou, 310009, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology Surgery Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
30
|
Han M, Yang S, Song J, Gao Z. Layer-by-layer coated probiotics with chitosan and liposomes demonstrate improved stability and antioxidant properties in vitro. Int J Biol Macromol 2024; 258:128826. [PMID: 38123040 DOI: 10.1016/j.ijbiomac.2023.128826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/09/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
Probiotics are of increasing interest for their potential health benefits, but their survival and adhesion in the harsh gastrointestinal environment remain a concern. This study explored a single-cell encapsulation technique to enhance probiotic survival and adhesion in the gastrointestinal tract. We encapsulated probiotics in curcumin-loaded liposomes, further coated them with polymers using layer-by-layer techniques. The coated probiotics were evaluated for survival in simulated gastrointestinal conditions, adhesion to colonic mucus, and scavenging of reactive oxygen species (ROS). The results showed that multi-layer encapsulation increased probiotic size at the nanoscale, enhancing their survival in simulated gastrointestinal conditions. Upon reaching the colon, the shedding of the coating coincided with probiotic proliferation. Additionally, the coated probiotics exhibited increased adhesion to colonic mucus. Moreover, the coating acted as a protective barrier for effectively scavenging reactive oxygen radicals, ensuring probiotic survival in inflammatory environments. This study combines the synergistic effects of probiotics and curcumin, underscoring the promise of single-cell encapsulation techniques in improving the efficacy of probiotics for addressing colitis-related diseases.
Collapse
Affiliation(s)
- Mengzhen Han
- College of Food Science and Engineering, Northwest A&F University, 712100 Yangling, Shaanxi, People's Republic of China
| | - Shuang Yang
- College of Food Science and Engineering, Northwest A&F University, 712100 Yangling, Shaanxi, People's Republic of China
| | - Jiangling Song
- College of Food Science and Engineering, Northwest A&F University, 712100 Yangling, Shaanxi, People's Republic of China
| | - Zhenpeng Gao
- College of Food Science and Engineering, Northwest A&F University, 712100 Yangling, Shaanxi, People's Republic of China.
| |
Collapse
|
31
|
Ding X, Li D, Xu Y, Wang Y, Liang S, Xie L, Yu W, Zhan X, Fu A. Carboxymethyl konjac glucomannan-chitosan complex nanogels stabilized emulsions incorporated into alginate as microcapsule matrix for intestinal-targeted delivery of probiotics: In vivo and in vitro studies. Int J Biol Macromol 2023; 253:126931. [PMID: 37722632 DOI: 10.1016/j.ijbiomac.2023.126931] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/20/2023]
Abstract
In this study, we developed a novel delivery system using carboxymethyl konjac glucomannan-chitosan (CMKGM-CS) nanogels stabilized single and double emulsion incorporated into alginate hydrogel as microcapsule matrix for intestinal-targeted delivery of probiotics. Through in vitro experiments, it was demonstrated that alginate hydrogel provided favorable biocompatible growth conditions for the proliferation of Lactobacillus reuteri (LR). The alginate hydrogel containing single (ASE) or double emulsions (ACG) enhanced the resistance of LR to various adverse environments. Simulated gastrointestinal digestion experiments revealed that the survivability of LR in free, CON, ASE and ACG group decreased by 6.45 log CFU/g, 4.21 log CFU/g, 1.26 log CFU/g and 0.65 log CFU/g, respectively. In vivo studies conducted in mice showed that ACG maintained its integrity during passage through the stomach and released the probiotics in the targeted intestinal area, whereas the pure alginate hydrogels (CON) were prematurely released in the gastrointestinal tract. Moreover, the viable counts of ACG in different intestinal segments (jejunum, ileum, cecum, and colon) were increased by 1.11, 1.42, 1.68, and 1.89 log CFU/g, respectively, after 72 h of oral administration compared to the CON group. This research contributed valuable insights into the development of an effective microbial delivery system with potential applications in the biopharmaceutical and food industries.
Collapse
Affiliation(s)
- Xiaoqing Ding
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture and Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Danlei Li
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture and Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yibin Xu
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture and Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuanyuan Wang
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture and Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shuang Liang
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture and Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lingyu Xie
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture and Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Weiqiang Yu
- Animal Husbandry and Veterinary Services Center of Haiyan, Jiaxing 314300, China.
| | - Xiuan Zhan
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture and Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Aikun Fu
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture and Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
32
|
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: 3] [Impact Index Per Article: 3.0] [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
|
33
|
Akram N, Afzaal M, Saeed F, Ahmad A, Imran A, Ahmed A, Shah YA, Islam F, Alomar SY, Manoharadas S, Nawaz A. Fabrication and Characterization of PVA-WPI Based Nanofiber Mats for Improved Viability of Lactobacillus rhamnosus GG. Foods 2023; 12:3904. [PMID: 37959023 PMCID: PMC10648975 DOI: 10.3390/foods12213904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/09/2023] [Accepted: 10/19/2023] [Indexed: 11/15/2023] Open
Abstract
In the current study, whey protein-based nanofibers were fabricated to encapsulate Lactobacillus rhamnosus. Purposely, different ratios of PVA (polyvinyl alcohol) and WPI (whey protein isolate) were blended to fabricate nanofibers. Nanofiber mats were characterized in terms of particle size, diameter, tensile strength, elongation at break, and loading efficiency. Morphological and molecular characterizations were carried out using scanning electron microscopy (SEM) and Fourier transform infrared (FTIR). Moreover, in vitro viability under simulated gastrointestinal (GI) conditions and thermal stability were also assessed. The results reveal that by increasing the PVA concentration, the conductivity increased while the viscosity decreased. SEM micrographs showed that probiotics were successfully loaded within the nanofiber. The FTIR spectra show strong bonding between the encapsulating materials with the addition of probiotics. In vitro and thermal analyses revealed that the survival of encapsulated probiotics significantly (p < 0.05) improved. In a nutshell, PVA-WPI composite nanofibers have promising potential when used to enhance the viability and stability of probiotics under adverse conditions.
Collapse
Affiliation(s)
- Noor Akram
- Food Safety and Biotechnology Lab, Department of Food Science, Government College University Faisalabad, Faisalabad 38000, Pakistan;
| | - Muhammad Afzaal
- Food Safety and Biotechnology Lab, Department of Food Science, Government College University Faisalabad, Faisalabad 38000, Pakistan;
- Department of Food Science, Government College University Faisalabad, Faisalabad 38000, Pakistan; (F.S.); (A.I.); (F.I.)
| | - Farhan Saeed
- Department of Food Science, Government College University Faisalabad, Faisalabad 38000, Pakistan; (F.S.); (A.I.); (F.I.)
| | - Adnan Ahmad
- Research School of Chemistry, Australian National University, Canberra 2601, Australia;
| | - Ali Imran
- Department of Food Science, Government College University Faisalabad, Faisalabad 38000, Pakistan; (F.S.); (A.I.); (F.I.)
| | - Aftab Ahmed
- Department of Nutritional Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan;
| | - Yasir Abbas Shah
- Natural and Medical Science Research Center, University of Nizwa, Nizwa 616, Oman;
| | - Fakhar Islam
- Department of Food Science, Government College University Faisalabad, Faisalabad 38000, Pakistan; (F.S.); (A.I.); (F.I.)
| | - Suliman Yousef Alomar
- Zoology Department, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Salim Manoharadas
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Asad Nawaz
- Hunan Engineering Technology Research Center for Comprehensive Development and Utilization of Biomass Resources, College of Chemistry and Bioengineering, Hunan University of Science and Engineering, Yongzhou 425199, China
| |
Collapse
|
34
|
Yang Q, Wu Z. Gut Probiotics and Health of Dogs and Cats: Benefits, Applications, and Underlying Mechanisms. Microorganisms 2023; 11:2452. [PMID: 37894110 PMCID: PMC10609632 DOI: 10.3390/microorganisms11102452] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Pets (mostly domestic dogs and cats) play an important role in the daily lives of humans and their health has attracted growing attention from pet owners. The intestinal microbiota, a complex microbial community with barrier-protective, nutritional, metabolic, and immunological functions, is integral to host health. Dysbiosis has been related to a variety of diseases in humans and animals. Probiotics have been used in functional foods and dietary supplements to modulate intestinal microbiota and promote host health, which has been introduced in pet dogs and cats in recent years. Various canine- and feline-derived probiotic strains have been isolated and characterized. The administration of probiotics has shown positive effects on the gut health and can alleviate some intestinal diseases and disorders in dogs and cats, although the underlying mechanisms are largely unresolved. In this review, we summarize the current knowledge on the benefits of probiotics and discuss their possible mechanisms in dogs and cats in order to provide new insights for the further development and application of probiotics in pets.
Collapse
Affiliation(s)
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, Department of Companion Animal Science, China Agricultural University, Beijing 100193, China;
| |
Collapse
|
35
|
Chen C, Zhu Z. Recent Advances in the Nanoshells Approach for Encapsulation of Single Probiotics. Drug Des Devel Ther 2023; 17:2763-2774. [PMID: 37705759 PMCID: PMC10497064 DOI: 10.2147/dddt.s419897] [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: 05/04/2023] [Accepted: 08/16/2023] [Indexed: 09/15/2023] Open
Abstract
The intestine, often referred to as the "second brain" of the human body, houses a vast microbial community that plays a crucial role in maintaining the host's balance and directly impacting overall health. Probiotics, a type of beneficial microorganism, offer various health benefits when consumed. However, probiotics face challenges such as acidic conditions in the stomach, bile acids, enzymes, and other adverse factors before they can colonize the intestinal tissues. At present, pills, dry powder, encapsulation, chemically modified bacteria, and genetically engineered bacteria have emerged as the preferred method for the stable and targeted delivery of probiotics. In particular, the use of nanoshells on the surface of single probiotics has shown promise in regulating their growth and differentiation. These nanoshells can detach from the probiotics' surface upon reaching the intestine, facilitating direct contact between the probiotics and intestinal mucosa. In this perspective, we provide an overview of the current developments in the formation of nanoshells mediated by single probiotics. We also discuss the advantages and disadvantages of different nanocoating strategies and explore future trends in probiotic protection.
Collapse
Affiliation(s)
- Cheng Chen
- The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang, Jiangsu Province, 212300, People’s Republic of China
| | - Ziyu Zhu
- The Affiliated Huai’an Hospital of Xuzhou Medical University and the Second People’s Hospital of Huai’an, Huai’an, 223002, People’s Republic of China
| |
Collapse
|
36
|
Yang J, Li D, Zhang M, Lin G, Hu S, Xu H. From the updated landscape of the emerging biologics for IBDs treatment to the new delivery systems. J Control Release 2023; 361:568-591. [PMID: 37572962 DOI: 10.1016/j.jconrel.2023.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/06/2023] [Accepted: 08/06/2023] [Indexed: 08/14/2023]
Abstract
Inflammatory bowel diseases (IBDs) treatments have shifted from small-molecular therapeutics to the oncoming biologics. The first-line biologics against the moderate-to-severe IBDs are mainly involved in antibodies against integrins, cytokines and cell adhesion molecules. Besides, other biologics including growth factors, antioxidative enzyme, anti-inflammatory peptides, nucleic acids, stem cells and probiotics have also been explored at preclinical or clinical studies. Biologics with variety of origins have their unique potentials in attenuating immune inflammation or gut mucosa healing. Great advances in use of biologics for IBDs treatments have been archived in recent years. But delivering issues for biologic have also been confronted due to their liable nature. In this review, we will focus on biologics for IBDs treatments in the recent publications; summarize the current landscapes of biologics and their promise to control disease progress. Alternatively, the confronted challenges for delivering biologics will also be analyzed. To combat these drawbacks, some new delivering strategies are provided: firstly, designing the functional materials with high affinity toward biologics; secondly, the delivering vehicle systems to encapsulate the liable biologics; thirdly, the topical adhering delivery systems as enema. To our knowledge, this review is the first study to summarize the updated usage of the oncoming biologics for IBDs, their confronted challenges in term of delivery and the potential combating strategies.
Collapse
Affiliation(s)
- Jiaojiao Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Dingwei Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Mengjiao Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Gaolong Lin
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Sunkuan Hu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province 325000, China
| | - Helin Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China.
| |
Collapse
|
37
|
Xie A, Ji H, Liu Z, Wan Y, Zhang X, Xiong H, Nie SP, Wan H. Modified Prebiotic-Based "Shield" Armed Probiotics with Enhanced Resistance of Gastrointestinal Stresses and Prolonged Intestinal Retention for Synergistic Alleviation of Colitis. ACS NANO 2023; 17:14775-14791. [PMID: 37477584 DOI: 10.1021/acsnano.3c02914] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
Oral administration of probiotics is a promising method to alleviate inflammatory bowel diseases (IBDs). However, gastrointestinal environmental sensitivity and inferior intestinal colonization of probiotics hinder the alleviation effect. Here, we developed a simple yet effective modified prebiotic-based "shield" (Fe-TA@mGN) composed of an Fe3+-tannic acid cross-linking network and carboxymethylated β-glucan for arming Escherichia coli Nissle 1917 (EcN@Fe-TA@mGN). The Fe-TA@mGN "shield" not only acted as a dynamic barrier to enhance the gastrointestinal stress resistance ability of EcN but also aided the intestinal colonization of EcN as well as synergized with EcN for the alleviation of dextran sulfate sodium (DSS) induced colitis. More specifically, with the protection of the Fe-TA@mGN "shield", the survival rate of armed EcN could be up to ∼1720 times higher than that of bare EcN after exposure to simulated gastric fluid. Excitingly, the intestinal retention rate of EcN@Fe-TA@mGN was as high as 47.54 ± 6.06% at 16 h post-administration, while almost all bare EcNs were excreted out at 8 h post-administration. With all of the aforementioned attributes, EcN@Fe-TA@mGN efficiently alleviated colitis, verified by the repair of the intestinal barrier and the attenuation of inflammation. Moreover, for EcN@Fe-TA@mGN, mGN synergized with EcN to positively modulate gut microbiota and promote the production of short-chain fatty acids (SCFAs, especially for butyric acid, a primary source for maintaining intestinal health), both of which would further advance the alleviation of colitis. We envision that the strategy developed here will inspire the exploitation of various prebiotics to arm probiotics for the effective alleviation of IBD.
Collapse
Affiliation(s)
- Anqi Xie
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Haihua Ji
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Zheyi Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Yiqun Wan
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang 330031, China
| | - Xuecong Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Huihuang Xiong
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang 330031, China
| | - Shao-Ping Nie
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Hao Wan
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| |
Collapse
|
38
|
Cao F, Jin L, Gao Y, Ding Y, Wen H, Qian Z, Zhang C, Hong L, Yang H, Zhang J, Tong Z, Wang W, Chen X, Mao Z. Artificial-enzymes-armed Bifidobacterium longum probiotics for alleviating intestinal inflammation and microbiota dysbiosis. NATURE NANOTECHNOLOGY 2023; 18:617-627. [PMID: 36973397 DOI: 10.1038/s41565-023-01346-x] [Citation(s) in RCA: 91] [Impact Index Per Article: 91.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 02/03/2023] [Indexed: 06/18/2023]
Abstract
Inflammatory bowel disease can be caused by the dysfunction of the intestinal mucosal barrier and dysregulation of gut microbiota. Traditional treatments use drugs to manage inflammation with possible probiotic therapy as an adjuvant. However, current standard practices often suffer from metabolic instability, limited targeting and result in unsatisfactory therapeutic outcomes. Here we report on artificial-enzyme-modified Bifidobacterium longum probiotics for reshaping a healthy immune system in inflammatory bowel disease. Probiotics can promote the targeting and retention of the biocompatible artificial enzymes to persistently scavenge elevated reactive oxygen species and alleviate inflammatory factors. The reduced inflammation caused by artificial enzymes improves bacterial viability to rapidly reshape the intestinal barrier functions and restore the gut microbiota. The therapeutic effects are demonstrated in murine and canine models and show superior outcomes to traditional clinical drugs.
Collapse
Affiliation(s)
- Fangfang Cao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lulu Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Yong Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongyang Wen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhefeng Qian
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chenyin Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Liangjie Hong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Huang Yang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Jiaojiao Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Zongrui Tong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, Singapore.
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China.
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
39
|
Zhang C, Gao X, Ren X, Xu T, Peng Q, Zhang Y, Chao Z, Jiang W, Jia L, Han L. Bacteria-Induced Colloidal Encapsulation for Probiotic Oral Delivery. ACS NANO 2023; 17:6886-6898. [PMID: 36947056 DOI: 10.1021/acsnano.3c00600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Probiotic oral delivery has crucial implications in biomedical engineering, but its oral bioavailability remains unsatisfactory because of the limited survival and colonization of probiotics in the harsh gastrointestinal tract. Here, a bacteria-induced encapsulation strategy is achieved by assembling metastable colloids to enhance the oral bioavailability of probiotics. The colloids (NTc) composed of amino-modified poly-β-cyclodextrin and tannic acid are formed based on the balance of host-guest interaction-driven attraction and electrostatic repulsion between colloids. Negatively charged probiotics electrostatically attract positively charged NTc to break the balance and induce further assembly surrounding the probiotics. Through a facile one-step mixing, 97% of bacteria are rapidly encapsulated into NTc shells within 10 s, with a high utilization rate of feeding colloids of 91%. More importantly, we show that the compact, thick, and positively charged NTc shells synergistically endow the encapsulated probiotics with strong resistance against simulated gastric fluid with an excellent survival rate of up to 19%, 7500 times superior to the commercial enteric material L100. Moreover, owing to the dynamically noncovalent and self-adaptive nature of host-guest interactions, NTc shells support the proliferation of the encapsulated EcN comparable with that of the naked EcN. In vitro and in vivo experiments also confirm that the NTc-encapsulated probiotics possess durable intestinal adhesion, continuous proliferation activity, enhanced oral bioavailability, good oral biosafety, and excellent therapeutic efficacy in a colitis mouse model. This facile bacteria-induced colloidal encapsulation strategy may extend to various microbes as oral bioagents for treating various diseases.
Collapse
Affiliation(s)
- Chong Zhang
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| | - Xiaorong Gao
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| | - Xinxiu Ren
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| | - Ting Xu
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| | - Qiang Peng
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| | - Yixin Zhang
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| | - Zhenhua Chao
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| | - Wenning Jiang
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| | - Lingyun Jia
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| | - Lulu Han
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, People's Republic of China
| |
Collapse
|
40
|
Wasilewska A, Bielicka M, Klekotka U, Kalska-Szostko B. Nanoparticle applications in food - a review. Food Funct 2023; 14:2544-2567. [PMID: 36799219 DOI: 10.1039/d2fo02180c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
The use of nanotechnology in the food industry raises uncertainty in many respects. For years, achievements of nanotechnology have been applied mainly in biomedicine and computer science, but recently it has also been used in the food industry. Due to the extremely small (nano) scale, the properties and behavior of nanomaterials may differ from their macroscopic counterparts. They can be used as biosensors to detect reagents or microorganisms, monitor bacterial growth conditions, increase food durability e.g. when placed in food packaging, reducing the amount of certain ingredients without changing the consistency of the product (research on fat substitutes is underway), improve the taste of food, make some nutrients get better absorbed by the body, etc. There are companies on the market that are already introducing nanoparticles into the economy to improve their functionality, e.g. baby feeding bottles. This review focuses on the use of nanoparticles in the food industry, both organic (chitosan, cellulose, proteins) and inorganic (silver, iron, zinc oxide, titanium oxide, etc.). The use of nanomaterials in food production requires compliance with all legal requirements regarding the safety and quantity of nano-processed food products described in this review. In the future, new methods of testing nanoparticles should be developed that would ensure the effectiveness of compounds subjected to, for example, nano-encapsulation, i.e. whether the encapsulation process had a positive impact on the specific properties of these compounds. Nanotechnology has revolutionized our approach towards food engineering (from production to processing), food storage and the creation of new materials and products, and the search for new product applications.
Collapse
Affiliation(s)
- A Wasilewska
- University of Bialystok, Faculty of Chemistry, Str. Ciolkowskiego 1K, 15-245, Bialystok, Poland.
- Doctoral School of Exact and Natural Sciences, University of Bialystok, Str. Ciolkowskiego 1K, 15-245 Bialystok, Poland
| | - M Bielicka
- University of Bialystok, Faculty of Chemistry, Str. Ciolkowskiego 1K, 15-245, Bialystok, Poland.
- Doctoral School of Exact and Natural Sciences, University of Bialystok, Str. Ciolkowskiego 1K, 15-245 Bialystok, Poland
| | - U Klekotka
- University of Bialystok, Faculty of Chemistry, Str. Ciolkowskiego 1K, 15-245, Bialystok, Poland.
| | - B Kalska-Szostko
- University of Bialystok, Faculty of Chemistry, Str. Ciolkowskiego 1K, 15-245, Bialystok, Poland.
| |
Collapse
|
41
|
Geng Z, Wang X, Wu F, Cao Z, Liu J. Biointerface mineralization generates ultraresistant gut microbes as oral biotherapeutics. SCIENCE ADVANCES 2023; 9:eade0997. [PMID: 36930714 PMCID: PMC10022893 DOI: 10.1126/sciadv.ade0997] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 02/09/2023] [Indexed: 06/18/2023]
Abstract
Despite the fact that oral microecologics are effective in modulating the gut microbiome, they always suffer from multiple insults during the journey from manufacture to arrival at the intestine. Inspired by the protective mechanism of mineralization, we describe a cytocompatible approach of biointerface mineralization that can generate an ultraresistant and self-removable coating on bacterial surface to solve these challenges. Mineral coating endows bacteria with robust resistances against manufacture-associated oxygen exposure, ultraviolet irradiation, and 75% ethanol. Following oral ingestion, the coating is able to actively neutralize gastric acid and release encapsulated bacteria through spontaneous yet rapid double-decomposition reaction. In addition to acid neutralization, the generated calcium ions can trigger micellar aggregation of bile acid, enabling dual exemptions from the insults of gastric acid and bile acid to achieve uncompromised bacterial viability. Further supported by the therapeutic efficacy of coated bacteria toward colitis mice, biointerface mineralization provides a versatile platform for developing next-generation living oral biotherapeutics.
Collapse
Affiliation(s)
- Zhongmin Geng
- 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
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, 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
| | - Feng Wu
- 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
| | - 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
|
42
|
An B, Wang Y, Huang Y, Wang X, Liu Y, Xun D, Church GM, Dai Z, Yi X, Tang TC, Zhong C. Engineered Living Materials For Sustainability. Chem Rev 2023; 123:2349-2419. [PMID: 36512650 DOI: 10.1021/acs.chemrev.2c00512] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Recent advances in synthetic biology and materials science have given rise to a new form of materials, namely engineered living materials (ELMs), which are composed of living matter or cell communities embedded in self-regenerating matrices of their own or artificial scaffolds. Like natural materials such as bone, wood, and skin, ELMs, which possess the functional capabilities of living organisms, can grow, self-organize, and self-repair when needed. They also spontaneously perform programmed biological functions upon sensing external cues. Currently, ELMs show promise for green energy production, bioremediation, disease treatment, and fabricating advanced smart materials. This review first introduces the dynamic features of natural living systems and their potential for developing novel materials. We then summarize the recent research progress on living materials and emerging design strategies from both synthetic biology and materials science perspectives. Finally, we discuss the positive impacts of living materials on promoting sustainability and key future research directions.
Collapse
Affiliation(s)
- Bolin An
- Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.,CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yanyi Wang
- Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.,CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yuanyuan Huang
- Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.,CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xinyu Wang
- Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.,CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yuzhu Liu
- Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.,CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Dongmin Xun
- Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.,CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - George M Church
- Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston 02115, Massachusetts United States.,Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston 02115, Massachusetts United States
| | - Zhuojun Dai
- Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.,CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiao Yi
- Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.,CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Tzu-Chieh Tang
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston 02115, Massachusetts United States.,Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston 02115, Massachusetts United States
| | - Chao Zhong
- Center for Materials Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.,CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
43
|
A red light-controlled probiotic bio-system for in-situ gut-brain axis regulation. Biomaterials 2023; 294:122005. [PMID: 36701997 DOI: 10.1016/j.biomaterials.2023.122005] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 12/27/2022] [Accepted: 01/13/2023] [Indexed: 01/21/2023]
Abstract
Microbes regulate brain function through the gut-brain axis, deriving the technology to modulate the gut-brain axis in situ by engineered probiotics. Optogenetics offers precise and flexible strategies for controlling the functions of probiotics in situ. However, the poor penetration of most frequently used short wavelength light has limited the application of optogenetic probiotics in the gut. Herein, a red-light optogenetic gut probiotic was applied for drug production and delivery and regulation of the host behaviors. Firstly, a Red-light Optogenetic E. coli Nissle 1917 strain (ROEN) that could respond to red light and release drug product by light-controlled lysis was constructed. The remaining optical power of red light after 3 cm tissue was still able to initiate gene expression of ROEN and produce about approximately 3-fold induction efficiency. To give full play to the in vivo potential of ROEN, its responsive ability of the penetrated red light was tested, and its encapsulation was realized by PH-sensitive alginate microcapsules for further oral administration. The function of ROEN for gut-brain regulation was realized by releasing Exendin-4 fused with anti-neonatal Fc receptor affibody. Neuroprotection and behavioral regulation effects were evaluated in the Parkinson's disease mouse model, after orally administration of ROEN delivering Exendin-4 under optogenetic control in the murine gut. The red-light optogenetic probiotic might be a perspective platform for in situ drug delivery and gut-brain axis regulation.
Collapse
|
44
|
Han SY, Nguyen DT, Kim BJ, Kim N, Kang EK, Park JH, Choi IS. Cytoprotection of Probiotic Lactobacillus acidophilus with Artificial Nanoshells of Nature-Derived Eggshell Membrane Hydrolysates and Coffee Melanoidins in Single-Cell Nanoencapsulation. Polymers (Basel) 2023; 15:polym15051104. [PMID: 36904345 PMCID: PMC10007236 DOI: 10.3390/polym15051104] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
One-step fabrication method for thin films and shells is developed with nature-derived eggshell membrane hydrolysates (ESMHs) and coffee melanoidins (CMs) that have been discarded as food waste. The nature-derived polymeric materials, ESMHs and CMs, prove highly biocompatible with living cells, and the one-step method enables cytocompatible construction of cell-in-shell nanobiohybrid structures. Nanometric ESMH-CM shells are formed on individual probiotic Lactobacillus acidophilus, without any noticeable decrease in viability, and the ESMH-CM shells effectively protected L. acidophilus in the simulated gastric fluid (SGF). The cytoprotection power is further enhanced by Fe3+-mediated shell augmentation. For example, after 2 h of incubation in SGF, the viability of native L. acidophilus is 30%, whereas nanoencapsulated L. acidophilus, armed with the Fe3+-fortified ESMH-CM shells, show 79% in viability. The simple, time-efficient, and easy-to-process method developed in this work would contribute to many technological developments, including microbial biotherapeutics, as well as waste upcycling.
Collapse
Affiliation(s)
- Sang Yeong Han
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon 34141, Republic of Korea
| | - Duc Tai Nguyen
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon 34141, Republic of Korea
| | - Beom Jin Kim
- Department of Chemistry, University of Ulsan, Ulsan 44776, Republic of Korea
- Correspondence: (B.J.K.); (I.S.C.)
| | - Nayoung Kim
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon 34141, Republic of Korea
| | - Eunhye K. Kang
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon 34141, Republic of Korea
| | - Ji Hun Park
- Department of Science Education, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Insung S. Choi
- Center for Cell-Encapsulation Research, Department of Chemistry, KAIST, Daejeon 34141, Republic of Korea
- Correspondence: (B.J.K.); (I.S.C.)
| |
Collapse
|
45
|
Sharma H, Sharma S, Bajwa J, Chugh R, Kumar D. Polymeric carriers in probiotic delivery system. CARBOHYDRATE POLYMER TECHNOLOGIES AND APPLICATIONS 2023. [DOI: 10.1016/j.carpta.2023.100301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2023] Open
|
46
|
Pu Y, Fan X, Zhang Z, Guo Z, Pan Q, Gao W, Luo K, He B. Harnessing polymer-derived drug delivery systems for combating inflammatory bowel disease. J Control Release 2023; 354:1-18. [PMID: 36566845 DOI: 10.1016/j.jconrel.2022.12.044] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/18/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022]
Abstract
The inflammatory bowel disease (IBD) is incurable, chronic, recrudescent disorders in the inflamed intestines. Current clinic treatments are challenged by systemic exposure-induced severe side effects, inefficiency after long-term treatment, and increased risks of infection and malignancy due to immunosuppression. Fortunately, naturally bioactive small molecules, reactive oxygen species scavengers (or antioxidants), and gut microbiota modulators have emerged as promising candidates for the IBD treatment. Polymeric systems have been engineered as a delivery vehicle to improve the bioavailability and efficacy of these therapeutic agents through targeting the mucosa and enhancing intestinal adhesion and retention, and reduce their systemic toxicity. Herein we survey polymer-derived drug delivery systems for combating the IBD. Advanced delivery technologies, therapeutic intervention strategies, and the principles for the construction of hierarchical, mucosa-targeting, and bioresponsive systems are elaborated, providing insights into design and development of from-bench-to-bedside drug delivery polymeric systems for the IBD treatment.
Collapse
Affiliation(s)
- Yuji Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xi Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Zhuangzhuang Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Zhaoyuan Guo
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Qingqing Pan
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Wenxia Gao
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325027, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Functional and Molecular Imaging Key Laboratory of Sichuan Province, Sichuan University, Chengdu 610041, China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
47
|
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: 13] [Impact Index Per Article: 6.5] [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
|
48
|
Ni F, Luo X, Zhao Z, Yuan J, Song Y, Liu C, Huang M, Dong L, Xie H, Cai L, Ren G, Gu Q. Enhancing viability of Lactobacillus plantarum encapsulated by alginate-gelatin hydrogel beads during gastrointestinal digestion, storage and in the mimic beverage systems. Int J Biol Macromol 2022; 224:94-104. [DOI: 10.1016/j.ijbiomac.2022.10.106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/06/2022] [Accepted: 10/11/2022] [Indexed: 11/05/2022]
|
49
|
Zuo P, Metz J, Yu P, Alvarez PJJ. Biofilm-responsive encapsulated-phage coating for autonomous biofouling mitigation in water storage systems. WATER RESEARCH 2022; 224:119070. [PMID: 36096027 DOI: 10.1016/j.watres.2022.119070] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/26/2022] [Accepted: 09/05/2022] [Indexed: 06/15/2023]
Abstract
Biofilms in water storage systems may harbor pathogens that threaten public health. Chemical disinfectants are marginally effective in eradicating biofilms due to limited penetration, and often generate harmful disinfection byproducts. To enhance biofouling mitigation in household water storage tanks, we encapsulated bacteriophages (phages) in chitosan crosslinked with tri-polyphosphate and 3-glycidoxypropyltrimethoxysilane. Phages served as self-propagating green biocides that exclusively infect bacteria. This pH-responsive encapsulation (244 ± 11 nm) enabled autonomous release of phages in response to acidic pH associated with biofilms (corroborated by confocal microscopy with pH-indicator dye SNARF-4F), but otherwise remained stable in pH-neutral tap water for one month. Encapsulated phages instantly bind to plasma-treated plastic and fiberglass surfaces, providing a facile coating method that protects surfaces highly vulnerable to biofouling. Biofilm formation assays were conducted in tap water amended with 200 mg/L glucose to accelerate growth and attachment of Pseudomonas aeruginosa, an opportunistic pathogen commonly associated with biofilms in drinking water distribution and storage systems. Biofilms formation on plastic surfaces coated with encapsulated phages decreased to only 6.7 ± 0.2% (on a biomass basis) relative to the uncoated controls. Likewise, biofilm surface area coverage (4.8 ± 0.2 log CFU/mm2) and live/dead fluorescence ratio (1.80) were also lower than the controls (6.6 ± 0.2 log CFU/mm2 and live/dead ratio of 11.05). Overall, this study offers proof-of-concept of a chemical-free, easily implementable approach to control problematic biofilm-dwelling bacteria and highlights benefits of this bottom-up biofouling control approach that obviates the challenge of poor biofilm penetration by biocides.
Collapse
Affiliation(s)
- Pengxiao Zuo
- Department of Civil and Environmental Engineering, Rice University, Houston, USA; Nanosystems Engineering Research Center for Nanotechnology-Enabled Water Treatment, USA
| | - Jordin Metz
- Department of Chemistry, Rice University, Houston, USA; Nanosystems Engineering Research Center for Nanotechnology-Enabled Water Treatment, USA
| | - Pingfeng Yu
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, China
| | - Pedro J J Alvarez
- Department of Civil and Environmental Engineering, Rice University, Houston, USA; Department of Chemistry, Rice University, Houston, USA; Nanosystems Engineering Research Center for Nanotechnology-Enabled Water Treatment, USA.
| |
Collapse
|
50
|
Sbehat M, Mauriello G, Altamimi M. Microencapsulation of Probiotics for Food Functionalization: An Update on Literature Reviews. Microorganisms 2022; 10:microorganisms10101948. [PMID: 36296223 PMCID: PMC9610121 DOI: 10.3390/microorganisms10101948] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/05/2022] Open
Abstract
Functional foods comprise the largest growing food category due to both consumer demands and health claims by manufacturers. Probiotics are considered one of the best choices for meeting these demands. Traditionally, the food vehicle for introducing probiotics to consumers was dairy products, and to expand the benefits of probiotics for a wider range of consumers, the need to use other food items was essential. To achieve this goal while maximising the benefits of probiotics, protection methods used during food processing were tackled. The microencapsulation of probiotics is a promising methodology for achieving this function. This review highlights the use of the microencapsulation of probiotics in order to functionalise food items that initially were not considered suitable for probiotication, such as baked products, or to increase their functionality such as dairy products. The co-microencapsulation of probiotics with other functional ingredients such polyphenol, prebiotics, or omega-3 is also highlighted.
Collapse
Affiliation(s)
- Maram Sbehat
- Department of Nutrition and Food Technology, An-Najah National University, Nablus P.O. Box 7, Palestine
- Department of Agricultural Sciences, University of Naples Federico II, 80055 Portici, Italy
| | - Gianluigi Mauriello
- Department of Agricultural Sciences, University of Naples Federico II, 80055 Portici, Italy
- Correspondence:
| | - Mohammad Altamimi
- Department of Nutrition and Food Technology, An-Najah National University, Nablus P.O. Box 7, Palestine
| |
Collapse
|