1
|
Xiong M, Sun W. Research progress of probiotics and their protective strategy in the field of inflammatory bowel disease treatment: A review. Medicine (Baltimore) 2024; 103:e40401. [PMID: 39495980 PMCID: PMC11537665 DOI: 10.1097/md.0000000000040401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/17/2024] [Indexed: 11/06/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic intestinal inflammatory disease characterized by recurrent episodes and difficult-to-cure symptoms. Although the pathogenesis of IBD is closely related to host genetic susceptibility, intestinal microbiota, environmental factors, and immune responses, leading to mucosal damage and increased intestinal permeability. Intestinal mucosal injury in IBD patients causes pathogenic bacteria and pathogenic factors to invade the intestine, leading to disturb the structure and metabolic products of intestinal flora. Researchers have found that probiotics, as live microbial agents, can effectively inhibit the growth of pathogenic bacteria, regulate intestinal flora, optimize intestinal microecology, restore intestinal homeostasis, and promote intestinal mucosal repairing. During the oral delivery process, probiotics are susceptible to adverse physiological factors, leading to reduced bioavailability. Additionally, the oxidative stress microenvironment induced by intestinal mucosal damage makes it difficult for probiotics to colonize the intestinal tract of IBD patients, thereby affecting their probiotic effect. This research mainly introduces and reviews the advantages and disadvantages of probiotics and their protective strategies in the treatment of IBD, and prospects the future development trends of probiotics and their protective strategies. Probiotics can effectively inhibit the growth of harmful microorganisms, regulate the structure of the intestinal microbiota, and promote mucosal repairing, thereby reducing immune stress and alleviating intestinal inflammation, providing a new perspective for the treatment of IBD. The development of single-cell encapsulation technology not only effectively maintaining the biological activity of probiotics during oral delivery, but also endowing probiotics with additional biological functions naturally achieved through surface programming, which has multiple benefits for intestinal health.
Collapse
Affiliation(s)
- Ming Xiong
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Wanlei Sun
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| |
Collapse
|
2
|
Zhu X, Zhi Y, Heng X, Zhou L, Liu C, Zhao Y, Wang Y, Liu J, Huang J. Optimization of a gelatin/carboxymethylcellulose-based probiotic microcapsule and its application in preventing dextran sodium sulfate-induced colitis in mice. J Food Sci 2024; 89:7976-7991. [PMID: 39366777 DOI: 10.1111/1750-3841.17372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/09/2024] [Accepted: 08/22/2024] [Indexed: 10/06/2024]
Abstract
Oral administration of probiotics has demonstrated substantial potential in alleviating colitis. However, most of the ingested microorganisms struggle to survive the harsh conditions of the gastrointestinal tract, leading to decreased efficacy. In the present study, using double emulsification (W1/O/W2) and complex coacervation methods, we developed a gelatin/carboxymethyl cellulose (CMC)-based probiotic microcapsule and analyzed the efficacy of encapsulated probiotics in preventing dextran sodium sulfate (DSS)-induced colitis in mice. Our results reveal that nearly 90% of the encapsulated probiotics remained viable after 30-day storage at 4°C and approximately 38.1% of viable bacteria (4.0 × 108 cfu/g) survived after 4-h simulated gastrointestinal digestion. In a DSS-induced colitis model, pretreatment with probiotics exerted significant protective effects, with the bacterial microcapsule-treated group having superior outcomes to those of the bacterial suspension plus empty carrier group. Probiotic treatments, especially those administered in the encapsulated form, significantly increased fecal short-chain fatty acid contents, and altered the intestinal microbial composition. The family Muribaculaceae, dominant bacteria in the mouse gut, may be the key microorganism involved in the BM regulation process. Our study presents an alternative approach to treating colitis using probiotics. PRACTICAL APPLICATION: The encapsuled probiotic showed remarkable storage stability at 4°C, maintained good vitality after simulated digestion, and gained superior outcomes in preventing colitis. Our results offer an alternative approach for the probiotic preparations aiming to prevent the intestinal inflammation.
Collapse
Affiliation(s)
- Xiao Zhu
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei, China
| | - Yiming Zhi
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei, China
| | - Xiaoyi Heng
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei, China
| | - Lihui Zhou
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei, China
| | - Chang Liu
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei, China
| | - Yiwen Zhao
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei, China
| | - Yijun Wang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei, China
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, China
- Anhui Provincial Joint Construction Key Laboratory of Industrial New-Style Tea Beverage Green Manufacturing, Anhui Agricultural University, Hefei, China
| | - Jun Liu
- School of Food Science and Engineering, Yangzhou University, Yangzhou, China
| | - Jinbao Huang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei, China
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, China
- Anhui Provincial Joint Construction Key Laboratory of Industrial New-Style Tea Beverage Green Manufacturing, Anhui Agricultural University, Hefei, China
| |
Collapse
|
3
|
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
|
4
|
Liu M, Ma S, Zhi J, Wang M, Xu Y, Kim YR, Luo K. Bioinspired core-shell microparticle for dual-delivery of prebiotic and probiotic for the treatment of ulcerative colitis. J Control Release 2024; 376:566-576. [PMID: 39426504 DOI: 10.1016/j.jconrel.2024.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/08/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
Lactiplantibacillus plantarum (LP) is a well-known probiotic strain that has a beneficial effect in preventing ulcerative colitis. However, delivering a sufficient number of viable LP to the colon still face challenges due to its vulnerability to the highly complex intestinal flora ecosystem. Herein, we present a centrifuge-driven micronozzle system designed for double-layered core-shell alginate microcapsules (DAM), which can serve as an effective carrier for dual delivery of resistant starch nanoparticles (RSNP, prebiotic) and LP (probiotics) for the treatment of colitis. This system enables precise loading of LP and RSNP within the core and shell regions of DAM, respectively. The resulting LP/RS@DAM exhibited a high encapsulation efficiency of LP (108 CFU per bead), in which the dense distribution of RSNP in the shell effectively protected LP against acidic conditions (pH 2) and maintained the cell viability up to 52 % even after long-term storage for 30 days. Furthermore, LP/RS@DAM effectively enhances the production of short-chain fatty acids, leading to a reduction in inflammatory cytokines and restoration of intestinal microbial diversity in dextran sulfate sodium (DSS)-induced colitis. We believe that this innovative approach would offer a potential solution for improving colitis management and paving the way for tailored therapeutic interventions in gastrointestinal disorders.
Collapse
Affiliation(s)
- Mengyao Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong Province 266003, China
| | - Shuang Ma
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong Province 266003, China
| | - Jinglei Zhi
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong Province 266003, China
| | - Mingming Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong Province 266003, China
| | - Ying Xu
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong Province 266003, China
| | - Young-Rok Kim
- Institute of Life Science and Resources & Department of Food Science and Biotechnology, College of Life Sciences, Kyung Hee University, Yongin 17104, South Korea
| | - Ke Luo
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong Province 266003, China.
| |
Collapse
|
5
|
Tang S, Wang W, Wang Y, Gao Y, Dai K, Zhang W, Wu X, Yuan X, Jin C, Zan X, Zhu L, Geng W. Sustained release of 5-aminosalicylic acid from azoreductase-responsive polymeric prodrugs for prolonged colon-targeted colitis therapy. J Nanobiotechnology 2024; 22:468. [PMID: 39103846 DOI: 10.1186/s12951-024-02724-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 07/16/2024] [Indexed: 08/07/2024] Open
Abstract
Ulcerative colitis (UC) is a challenging inflammatory gastrointestinal disorder, whose therapies encounter limitations in overcoming insufficient colonic retention and rapid systemic clearance. In this study, we report an innovative polymeric prodrug nanoformulation for targeted UC treatment through sustained 5-aminosalicylic acid (5-ASA) delivery. Amphiphilic polymer-based 13.5 nm micelles were engineered to incorporate azo-linked 5-ASA prodrug motifs, enabling cleavage via colonic azoreductases. In vitro, micelles exhibited excellent stability under gastric/intestinal conditions while demonstrating controlled 5-ASA release over 24 h in colonic fluids. Orally administered micelles revealed prolonged 24-h retention and a high accumulation within inflamed murine colonic tissue. At an approximately 60% dose reduction from those most advanced recent studies, the platform halted DSS colitis progression and outperformed standard 5-ASA therapy through a 77-97% suppression of inflammatory markers. Histological analysis confirmed intact colon morphology and restored barrier protein expression. This integrated prodrug nanoformulation addresses limitations in colon-targeted UC therapy through localized bioactivation and tailored pharmacokinetics, suggesting the potential of nanotechnology-guided precision delivery to transform disease management.
Collapse
Affiliation(s)
- Sicheng Tang
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
- University of Chinese Academy of Sciences, Wenzhou Institute, Wenzhou, 325001, Zhejiang, China.
| | - Wenchao Wang
- Department of Pain, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yijian Wang
- Department of Pain, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yuhan Gao
- University of Chinese Academy of Sciences, Wenzhou Institute, Wenzhou, 325001, Zhejiang, China
- School of Pharmacy, Wenzhou Medical University, Wenzhou, 325001, Zhejiang, China
| | - Keke Dai
- Department of Pain, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Wenjing Zhang
- Department of Pain, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xudong Wu
- Department of Pain, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xiaodie Yuan
- University of Chinese Academy of Sciences, Wenzhou Institute, Wenzhou, 325001, Zhejiang, China
| | - Chaofan Jin
- University of Chinese Academy of Sciences, Wenzhou Institute, Wenzhou, 325001, Zhejiang, China
| | - Xingjie Zan
- University of Chinese Academy of Sciences, Wenzhou Institute, Wenzhou, 325001, Zhejiang, China
| | - Limeng Zhu
- University of Chinese Academy of Sciences, Wenzhou Institute, Wenzhou, 325001, Zhejiang, China.
| | - Wujun Geng
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Medical University, Wenzhou, 325001, Zhejiang, China.
| |
Collapse
|
6
|
Lin X, Jiao R, Cui H, Yan X, Zhang K. Physiochemically and Genetically Engineered Bacteria: Instructive Design Principles and Diverse Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403156. [PMID: 38864372 PMCID: PMC11321697 DOI: 10.1002/advs.202403156] [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: 03/26/2024] [Revised: 04/18/2024] [Indexed: 06/13/2024]
Abstract
With the comprehensive understanding of microorganisms and the rapid advances of physiochemical engineering and bioengineering technologies, scientists are advancing rationally-engineered bacteria as emerging drugs for treating various diseases in clinical disease management. Engineered bacteria specifically refer to advanced physiochemical or genetic technologies in combination with cutting edge nanotechnology or physical technologies, which have been validated to play significant roles in lysing tumors, regulating immunity, influencing the metabolic pathways, etc. However, there has no specific reviews that concurrently cover physiochemically- and genetically-engineered bacteria and their derivatives yet, let alone their distinctive design principles and various functions and applications. Herein, the applications of physiochemically and genetically-engineered bacteria, and classify and discuss significant breakthroughs with an emphasis on their specific design principles and engineering methods objective to different specific uses and diseases beyond cancer is described. The combined strategies for developing in vivo biotherapeutic agents based on these physiochemically- and genetically-engineered bacteria or bacterial derivatives, and elucidated how they repress cancer and other diseases is also underlined. Additionally, the challenges faced by clinical translation and the future development directions are discussed. This review is expected to provide an overall impression on physiochemically- and genetically-engineered bacteria and enlighten more researchers.
Collapse
Affiliation(s)
- Xia Lin
- Central Laboratory and Department of UltrasoundSichuan Academy of Medical SciencesSichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of ChinaNo. 32, West Second Section, First Ring RoadChengduSichuan610072China
| | - Rong Jiao
- Central Laboratory and Department of UltrasoundSichuan Academy of Medical SciencesSichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of ChinaNo. 32, West Second Section, First Ring RoadChengduSichuan610072China
| | - Haowen Cui
- Central Laboratory and Department of UltrasoundSichuan Academy of Medical SciencesSichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of ChinaNo. 32, West Second Section, First Ring RoadChengduSichuan610072China
| | - Xuebing Yan
- Department of OncologyAffiliated Hospital of Yangzhou University. No.368Hanjiang Road, Hanjiang DistrictYangzhouJiangsu Province225012China
| | - Kun Zhang
- Central Laboratory and Department of UltrasoundSichuan Academy of Medical SciencesSichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of ChinaNo. 32, West Second Section, First Ring RoadChengduSichuan610072China
| |
Collapse
|
7
|
Feng P, Bai X, Ma X, Kong H, Yang R. Interfacial-engineered living drugs with "ON/OFF" switching for oral delivery. NANOSCALE 2024; 16:13399-13406. [PMID: 38953700 DOI: 10.1039/d4nr01927j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Living drugs offer a new frontier in medicine, paving the way for personalized and potentially curative treatments. A customized living drug generally requires specialized technologies for highly effective and selective delivery to lesion locations. In this study, we explored an interfacial engineering method for living drugs by wrapping them with a "stealth coating", achieving "ON/OFF" switching of the communications between probiotics and the gastrointesinal (GI) tract. This maximized the bioactivity of living drugs following oral administration to exempt acidic insults and then significantly improved the retention through the gastrointestinal tract. With the notable ability to improve oral availability, the interfacial-engineered living drugs represent remarkable effects for enhanced oral delivery and treatment efficacy in the dextran sulfate sodium (DSS)-induced acute colitis model. We believe that this work has the potential to revolutionize medicine by precisely targeting and increasing curative activity in the future of disease treatment.
Collapse
Affiliation(s)
- Pingping Feng
- College of Chemistry and Molecular Engineering, Peking University, Beijing, No. 292 Chengfu Road, Haidian District, Beijing, 100871 P. R. China.
| | - Xuefei Bai
- College of Chemistry, Beijing Normal University, No. 19, Xinjiekouwai Street, Haidian District, Beijing 100875, P. R. China.
| | - Xiaofei Ma
- College of Chemistry, Beijing Normal University, No. 19, Xinjiekouwai Street, Haidian District, Beijing 100875, P. R. China.
| | - Han Kong
- College of Chemistry, Beijing Normal University, No. 19, Xinjiekouwai Street, Haidian District, Beijing 100875, P. R. China.
| | - Rui Yang
- College of Chemistry, Beijing Normal University, No. 19, Xinjiekouwai Street, Haidian District, Beijing 100875, P. R. China.
| |
Collapse
|
8
|
Wang K, Liu F, Muchu B, Deng J, Peng J, Xu Y, Li F, Ouyang M. E3 ubiquitin ligase RNF180 mediates the ALKBH5/SMARCA5 axis to promote colon inflammation and Th17/Treg imbalance in ulcerative colitis mice. Arch Pharm Res 2024; 47:645-658. [PMID: 39060657 DOI: 10.1007/s12272-024-01507-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 07/06/2024] [Indexed: 07/28/2024]
Abstract
SMARCA5, a protein in the SWI/SNF family, has been previously implicated in the development of ulcerative colitis (UC) through methylation. However, the specific molecular mechanisms by which SMARCA5 contributes to colonic inflammation and the imbalance between Th17 and Treg cells remain unclear. This study was designed to explore these molecular mechanisms. A UC mouse model was established using dextran sulfate sodium induction, followed by measurements of mouse weight, disease activity index (DAI) score, colon length, pathological changes in the colon, and FITC-dextran concentration. The levels of IL-17a, IFN-γ, IL-6, TNF-α, TGF-β, and IL-10 were measured, along with the protein expression of ZO-1 and Occludin. Flow cytometry was used to assess the presence of IL-17 + CD4 + (Th17 +) cells and FOXP3 + CD25 + CD4 + (Treg +) cells in the spleen and mesenteric lymph nodes of UC mice. We observed that SMARCA5 and RNF180 were increased, while ALKBH5 was downregulated in UC mouse colon tissue. SMARCA5 or RNF180 knockdown or ALKBH5 overexpression ameliorated the colon inflammation and Th17/Treg cell imbalance in UC mice, shown by increased body weight, colon length, FOXP3 + CD25 + CD4 + T cells, and the levels of ZO-1, Occludin, TGF-β, IL-10, and FOXP3. It decreased DAI scores, IL-17 + CD4 + T cells, and levels of IL-17a, IFN-γ, IL-6, TNF-α, and ROR-γt. ALKBH5 inhibited SMARCA5 expression via m6A modification, while RNF180 reduced ALKBH5 expression via ubiquitination. Our findings indicate that RNF180 aggravated the colon inflammation and Th17/Treg cell imbalance in UC mice by regulating the ALKBH5/SMARCA5 axis.
Collapse
MESH Headings
- Animals
- Male
- Mice
- AlkB Homolog 5, RNA Demethylase/metabolism
- Chromosomal Proteins, Non-Histone/metabolism
- Chromosomal Proteins, Non-Histone/genetics
- Colitis, Ulcerative/immunology
- Colitis, Ulcerative/pathology
- Colitis, Ulcerative/chemically induced
- Colitis, Ulcerative/metabolism
- Dextran Sulfate/toxicity
- Disease Models, Animal
- Inflammation/metabolism
- Inflammation/pathology
- Inflammation/immunology
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Th17 Cells/immunology
- Th17 Cells/metabolism
- Ubiquitin-Protein Ligases/metabolism
- Ubiquitin-Protein Ligases/genetics
Collapse
Affiliation(s)
- Kailing Wang
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Fan Liu
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Budumu Muchu
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Jiawen Deng
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Jing Peng
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Yan Xu
- Department of Health Management Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Fujun Li
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Miao Ouyang
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
9
|
Liang J, Dai W, Liu C, Wen Y, Chen C, Xu Y, Huang S, Hou S, Li C, Chen Y, Wang W, Tang H. Gingerenone A Attenuates Ulcerative Colitis via Targeting IL-17RA to Inhibit Inflammation and Restore Intestinal Barrier Function. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400206. [PMID: 38639442 PMCID: PMC11267284 DOI: 10.1002/advs.202400206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/27/2024] [Indexed: 04/20/2024]
Abstract
Ulcerative colitis (UC) is a complicated and recurrent intestinal disease. Currently available drugs for UC treatment are scarce, therefore, novel therapeutic drugs for the UC are urgently to be developed. Gingerenone A (GA) is a phenolic compound known for its anti-inflammatory effect, but its effect on UC remains unknown. Here, it is shown that GA protects mice against UC, which is closely associated with inhibiting intestinal mucosal inflammation and enhancing intestinal barrier integrity in vivo and in vitro. Of note, RNA sequencing analysis demonstrates an evident correlation with IL-17 signaling pathway after GA treatment, and this effect is further corroborated by Western blot. Mechanistically, GA directly interacts with IL-17RA protein through pull-down, surface plasmon resonance analysis and molecular dynamics simulation. Importantly, lentivirus-mediated IL-17RA/Act1 knock-down or GA co-treatment with brodalumab/ixekizumab significantly impairs the protective effects of GA against DSS-induced inflammation and barrier dysfunction, suggesting a critical role of IL-17RA signaling for GA-mediated protection against UC. Overall, these results indicate that GA is an effective agent against UC mainly through the direct binding of IL-17RA to inhibit inflammatory signaling activation.
Collapse
Affiliation(s)
- Jian Liang
- School of Pharmaceutical SciencesState Key Laboratory of Traditional Chinese Medicine SyndromeGuangzhou University of Chinese MedicineGuangzhou510006China
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhou510060China
- Dongguan Institute of Guangzhou University of Chinese MedicineDongguan523808China
| | - Weigang Dai
- Center of Ganstric CancerThe First Affiliated HospitalSun Yat‐Sen UniversityGuangzhou510062China
| | - Chuanghui Liu
- School of Pharmaceutical SciencesState Key Laboratory of Traditional Chinese Medicine SyndromeGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Yifan Wen
- School of Pharmaceutical SciencesState Key Laboratory of Traditional Chinese Medicine SyndromeGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Chen Chen
- School of Pharmaceutical SciencesState Key Laboratory of Traditional Chinese Medicine SyndromeGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Yifei Xu
- Shenzhen Traditional Chinese Medicine HospitalThe Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen518033China
| | - Song Huang
- School of Pharmaceutical SciencesState Key Laboratory of Traditional Chinese Medicine SyndromeGuangzhou University of Chinese MedicineGuangzhou510006China
- Dongguan Institute of Guangzhou University of Chinese MedicineDongguan523808China
| | - Shaozhen Hou
- School of Pharmaceutical SciencesState Key Laboratory of Traditional Chinese Medicine SyndromeGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Chun Li
- School of Pharmaceutical SciencesState Key Laboratory of Traditional Chinese Medicine SyndromeGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Yongming Chen
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhou510060China
| | - Wei Wang
- School of Pharmaceutical SciencesState Key Laboratory of Traditional Chinese Medicine SyndromeGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Hailin Tang
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhou510060China
| |
Collapse
|
10
|
Park B, Han G, Jin DY, Gil KC, Shin D, Lee J, Park JY, Jang H, Park D, Lee S, Kim K, Yang Y, Kim Y, Kim JS, Kim SH, Shim MK. Mucoadhesive Mesalamine Prodrug Nanoassemblies to Target Intestinal Macrophages for the Treatment of Inflammatory Bowel Disease. ACS NANO 2024; 18:16297-16311. [PMID: 38867457 DOI: 10.1021/acsnano.4c05544] [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: 06/14/2024]
Abstract
While mesalamine, a 5-aminosalicylic acid (5-ASA), is pivotal in the management of inflammatory bowel disease (IBD) through both step-up and top-down approaches in clinical settings, its widespread utilization is limited by low bioavailability at the desired site of action due to rapid and extensive absorption in the upper gastrointestinal (GI) tract. Addressing mesalamine's pharmacokinetic challenges, here, we introduce nanoassemblies composed exclusively of a mesalamine prodrug that pairs 5-ASA with a mucoadhesive and cathepsin B-cleavable peptide. In an IBD model, orally administered nanoassemblies demonstrate enhanced accumulation and sustained retention in the GI tract due to their mucoadhesive properties and the epithelial enhanced permeability and retention (eEPR) effect. This retention enables the efficient uptake by intestinal pro-inflammatory macrophages expressing high cathepsin B, triggering a burst release of the 5-ASA. This cascade fosters the polarization toward an M2 macrophage phenotype, diminishes inflammatory responses, and simultaneously facilitates the delivery of active agents to adjacent epithelial cells. Therefore, the nanoassemblies show outstanding therapeutic efficacy in inhibiting local inflammation and contribute to suppressing systemic inflammation by restoring damaged intestinal barriers. Collectively, this study highlights the promising role of the prodrug nanoassemblies in enhancing targeted drug delivery, potentially broadening the use of mesalamine in managing IBD.
Collapse
Affiliation(s)
- Byeongmin Park
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Geonhee Han
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Do Young Jin
- Department of Nano-Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Ki Cheol Gil
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Dongwon Shin
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Jongwon Lee
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Jung Yeon Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Hochung Jang
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Daeho Park
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Sangmin Lee
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Kwangmeyung Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Yoosoo Yang
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Yongju Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Jun-Seob Kim
- Department of Nano-Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Sun Hwa Kim
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Man Kyu Shim
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| |
Collapse
|
11
|
Ma L, Ma Y, Gao Q, Liu S, Zhu Z, Shi X, Dai F, Reis RL, Kundu SC, Cai K, Xiao B. Mulberry Leaf Lipid Nanoparticles: a Naturally Targeted CRISPR/Cas9 Oral Delivery Platform for Alleviation of Colon Diseases. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307247. [PMID: 38243871 DOI: 10.1002/smll.202307247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/14/2023] [Indexed: 01/22/2024]
Abstract
Oral treatment of colon diseases with the CRISPR/Cas9 system has been hampered by the lack of a safe and efficient delivery platform. Overexpressed CD98 plays a crucial role in the progression of ulcerative colitis (UC) and colitis-associated colorectal cancer (CAC). In this study, lipid nanoparticles (LNPs) derived from mulberry leaves are functionalized with Pluronic copolymers and optimized to deliver the CRISPR/Cas gene editing machinery for CD98 knockdown. The obtained LNPs possessed a hydrodynamic diameter of 267.2 nm, a narrow size distribution, and a negative surface charge (-25.6 mV). Incorporating Pluronic F127 into LNPs improved their stability in the gastrointestinal tract and facilitated their penetration through the colonic mucus barrier. The galactose end groups promoted endocytosis of the LNPs by macrophages via asialoglycoprotein receptor-mediated endocytosis, with a transfection efficiency of 2.2-fold higher than Lipofectamine 6000. The LNPs significantly decreased CD98 expression, down-regulated pro-inflammatory cytokines (TNF-α and IL-6), up-regulated anti-inflammatory factors (IL-10), and polarized macrophages to M2 phenotype. Oral administration of LNPs mitigated UC and CAC by alleviating inflammation, restoring the colonic barrier, and modulating intestinal microbiota. As the first oral CRISPR/Cas9 delivery LNP, this system offers a precise and efficient platform for the oral treatment of colon diseases.
Collapse
Affiliation(s)
- Lingli Ma
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Ya Ma
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Qiang Gao
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Shengsheng Liu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Zhenhua Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xiaoxiao Shi
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Fangyin Dai
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Rui L Reis
- Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Barco, Guimaraes, 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, 4800-058, Portugal
| | - Subhas C Kundu
- Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Barco, Guimaraes, 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, 4800-058, Portugal
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Bo Xiao
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| |
Collapse
|
12
|
Zhu L, Yu T, Wang W, Xu T, Geng W, Li N, Zan X. Responsively Degradable Nanoarmor-Assisted Super Resistance and Stable Colonization of Probiotics for Enhanced Inflammation-Targeted Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308728. [PMID: 38241751 DOI: 10.1002/adma.202308728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/09/2023] [Indexed: 01/21/2024]
Abstract
Manipulation of the gut microbiota using oral microecological preparations has shown great promise in treating various inflammatory disorders. However, delivering these preparations while maintaining their disease-site specificity, stability, and therapeutic efficacy is highly challenging due to the dynamic changes associated with pathological microenvironments in the gastrointestinal tract. Herein, a superior armored probiotic with an inflammation-targeting capacity is developed to enhance the efficacy and timely action of bacterial therapy against inflammatory bowel disease (IBD). The coating strategy exhibits suitability for diverse probiotic strains and has negligible influence on bacterial viability. This study demonstrates that these armored probiotics have ultraresistance to extreme intraluminal conditions and stable mucoadhesive capacity. Notably, the HA-functionalized nanoarmor equips the probiotics with inflamed-site targetability through multiple interactions, thus enhancing their efficacy in IBD therapy. Moreover, timely "awakening" of ingested probiotics through the responsive transferrin-directed degradation of the nanoarmor at the site of inflammation is highly beneficial for bacterial therapy, which requires the bacterial cells to be fully functional. Given its easy preparation and favorable biocompatibility, the developed single-cell coating approach provides an effective strategy for the advanced delivery of probiotics for biomedical applications at the cellular level.
Collapse
Affiliation(s)
- Limeng Zhu
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325000, China
- Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tiantian Yu
- Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Wenchao Wang
- Department of Pain, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Tong Xu
- College of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Wujun Geng
- Department of Pain, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Na Li
- Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Xingjie Zan
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325000, China
- Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| |
Collapse
|
13
|
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
|
14
|
Yu C, Xu Y, Wei Y, Guo Y, Wang Y, Song P, Yu J. Gut microbiota and liver metabolomics reveal the potential mechanism of Lactobacillus rhamnosus GG modulating the liver toxicity caused by polystyrene microplastics in mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:6527-6542. [PMID: 38151562 DOI: 10.1007/s11356-023-31564-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/11/2023] [Indexed: 12/29/2023]
Abstract
Microplastics (MPs) are known to cause liver toxicity as they can spread through the food chain. Most researches on their toxicity have focused on individual organs, neglecting the crucial "gut-liver axis"-a bidirectional communication pathway between the gut and liver. Probiotics have shown promise in modulating the effects of environmental pollutants. In this study, we exposed mice to Lactobacillus rhamnosus GG (LGG, 100 mg/kg b.w./d) and/or polystyrene microplastics (PS-MPs, 5 mg/kg b.w./d) for 28 d via gavage to investigate how probiotics influence live toxicity through the gut-liver axis. Our results demonstrated that PS-MPs induced liver inflammation (increased IL-6 and TNF-α) and disrupted lipid metabolism. However, when combined with LGG, these effects were alleviated. LGG also improved colon health, rectifying ciliary defects and abnormal mucus secretion caused by PS-MPs. Furthermore, LGG improved gut microbiota dysbiosis induced by PS-MPs. Metabolomics and gene expression analysis (Cyp7a1 and Cyp7b1) indicated that LGG modulated bile acid metabolism. In summary, LGG appears to protect the liver by maintaining gut homeostasis, enhancing gut barrier integrity, and reducing the liver inflammation. These findings confirm the potential of LGG to modulate liver toxicity caused by PS-MPs through the gut-liver axis, offering insights into probiotics' application for environmental pollutant detoxification.
Collapse
Affiliation(s)
- Changhao Yu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Xuelin Road, Nanjing, 210023, People's Republic of China
| | - Yawen Xu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Xuelin Road, Nanjing, 210023, People's Republic of China
| | - Yiping Wei
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Xuelin Road, Nanjing, 210023, People's Republic of China
| | - Yuxue Guo
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Xuelin Road, Nanjing, 210023, People's Republic of China
| | - Yi Wang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Xuelin Road, Nanjing, 210023, People's Republic of China
| | - Ping Song
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Xuelin Road, Nanjing, 210023, People's Republic of China
| | - Jing Yu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Xuelin Road, Nanjing, 210023, People's Republic of China.
| |
Collapse
|
15
|
Kuang X, Liu Y, Luo H, Li Q, Wu F, Fan C, Liu J. Triggerable Prodrug Nanocoating Enables On-Demand Activation of Microbial and Small-Molecular Therapeutics for Combination Treatment. J Am Chem Soc 2023; 145:26932-26946. [PMID: 37988674 DOI: 10.1021/jacs.3c10015] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
The synergy of living microbial and small-molecular therapeutics has been widely explored for treating a variety of diseases, while current combination strategies often suffer from low bioavailability, heterogeneous spatiotemporal distribution, and premature drug release. Here, the use of a triggerable prodrug nanocoating is reported to enable the on-demand activation of microbial and small-molecular therapeutics for combination treatment. As a proof-of-concept study, a reactive oxygen species-responsive aromatic thioacetal linker is employed to prepare cationic chitosan-drug conjugates, which can form a nanocoating on the surface of living bacteria via electrostatic interaction. Following administration, the wrapped bacteria can be prevented from in vivo insults by the shielding effect of the nanocoating and be co-delivered with the conjugated drug in a spatiotemporally synchronous manner. Upon reaching the lesion site, the upgraded reactive oxygen species trigger in situ cleavage of the thioacetal linker, resulting in the release of the conjugated drug and a linker-derived therapeutic cinnamaldehyde. Meanwhile, a charge reversal achieved by the generation of negatively charged thiolated chitosan induces the dissociation of the nanocoating, leading to synchronous release of the living bacteria. The adequate activation of the combined therapeutics at the lesion site exhibits superior synergistic treatment efficacy, as demonstrated by an in vivo assessment using a mouse model of colitis. This work presents an appealing approach to combine living microbial and small-molecular therapeutics for advanced therapy of diseases.
Collapse
Affiliation(s)
- Xiao Kuang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ying Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Huilong Luo
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Qian Li
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Feng Wu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Chunhai Fan
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
16
|
Qiu J, Xiang S, Sun M, Tan M. Preparation of Polysaccharide-Protein Hydrogels with an Ultrafast Self-Healing Property as a Superior Oral Delivery System of Probiotics. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:18842-18856. [PMID: 37978937 DOI: 10.1021/acs.jafc.3c05898] [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: 11/19/2023]
Abstract
Oral administration of probiotic supplements can effectively regulate intestinal disorders. However, harsh gastrointestinal conditions greatly limit the bioavailability of probiotics. In this work, biomass-derived polysaccharide-protein hydrogels (Dex-sBSA hydrogels) were constructed as an oral probiotic delivery system. The hydrogel encapsulation significantly promoted the growth and proliferation of probiotics and protected them from gastric acid, bile salts, reactive oxygen species, and antibiotics. In vivo experiments demonstrated that the hydrogel encapsulation significantly enhanced the bioavailability of probiotics, of which the cell number in the intestine, colon, and cecum was 35 times, 8 times, and 203 times higher than the free one, respectively. Attributed to the superior ultrafast self-healing property, the Dex-sBSA hydrogel successfully prevented the probiotics from quick elimination and prolonged the retention time in the gut, providing great possibilities for colonization and proliferation. These results clearly indicate the great potential of the Dex-sBSA hydrogel as a superior oral delivery system for probiotics.
Collapse
Affiliation(s)
- Jiaqi Qiu
- State Key Lab of Marine Food Processing & Safety Control, Dalian Polytechnic University, Dalian 116034, P. R. China
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, P. R. China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Siyuan Xiang
- State Key Lab of Marine Food Processing & Safety Control, Dalian Polytechnic University, Dalian 116034, P. R. China
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, P. R. China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Miyao Sun
- State Key Lab of Marine Food Processing & Safety Control, Dalian Polytechnic University, Dalian 116034, P. R. China
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, P. R. China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Mingqian Tan
- State Key Lab of Marine Food Processing & Safety Control, Dalian Polytechnic University, Dalian 116034, P. R. China
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, P. R. China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, P. R. China
| |
Collapse
|
17
|
Peng P, Feng T, Yang X, Nie C, Yu L, Ding R, Zhou Q, Jiang X, Li P. Gastrointestinal Microenvironment Responsive Nanoencapsulation of Probiotics and Drugs for Synergistic Therapy of Intestinal Diseases. ACS NANO 2023; 17:14718-14730. [PMID: 37490035 DOI: 10.1021/acsnano.3c02646] [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: 07/26/2023]
Abstract
The gut microbiota are prominent in preserving intestinal environmental homeostasis and managing human health, and their dysbiosis has been directly related to many kinds of intestinal diseases. Probiotics-based therapy appears as a promising approach for the treatment of gut microbiota dysbiosis, while it always suffers from limited bioavailability and therapeutic effect after oral administration. Herein, we presented a facile and safe strategy to treat colitis by nanoencapsulation of probiotics and an anti-inflammatory agent, 5-aminosalicylic acid (5-ASA), within the gastrointestinal microenvironment responsive alginate polysaccharide. Because of acid resistance, the alginate-based coating protected probiotics from the harsh gastric condition. The coating could be disintegrated to release probiotics and 5-ASA upon arriving in the intestinal tract, where the pH is normally higher than 5. In the dextran sulfate sodium-induced colitis mouse model, probiotics recovered their bioactivities and acted together with anti-inflammatory 5-ASA to alleviate colitis by upregulating microbiota richness and diversity, reducing expression of proinflammatory cytokines, and restoring intestinal barriers. This work demonstrated the synergistic therapy of intestinal diseases based on alginate-encapsulated probiotics and a clinical drug, which provided an extensive method to improve the therapeutic effect of oral microecologics.
Collapse
Affiliation(s)
- Pandi Peng
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Ningbo Institute & Chongqing Technology Innovation Center, Northwestern Polytechnical University (NPU), Xi'an, 710072 China
| | - Tao Feng
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Ningbo Institute & Chongqing Technology Innovation Center, Northwestern Polytechnical University (NPU), Xi'an, 710072 China
| | - Xue Yang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Ningbo Institute & Chongqing Technology Innovation Center, Northwestern Polytechnical University (NPU), Xi'an, 710072 China
| | - Chaofan Nie
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Ningbo Institute & Chongqing Technology Innovation Center, Northwestern Polytechnical University (NPU), Xi'an, 710072 China
| | - Luofeng Yu
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Ningbo Institute & Chongqing Technology Innovation Center, Northwestern Polytechnical University (NPU), Xi'an, 710072 China
| | - Rui Ding
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Ningbo Institute & Chongqing Technology Innovation Center, Northwestern Polytechnical University (NPU), Xi'an, 710072 China
| | - Qian Zhou
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Ningbo Institute & Chongqing Technology Innovation Center, Northwestern Polytechnical University (NPU), Xi'an, 710072 China
| | - Xueqing Jiang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Ningbo Institute & Chongqing Technology Innovation Center, Northwestern Polytechnical University (NPU), Xi'an, 710072 China
| | - Peng Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Ningbo Institute & Chongqing Technology Innovation Center, Northwestern Polytechnical University (NPU), Xi'an, 710072 China
| |
Collapse
|
18
|
Liu Y, Yu W, Wang Q, Cao Z, Li J. Artificially engineered bacteria to treat gastrointestinal disease and cancer. Drug Discov Today 2023; 28:103667. [PMID: 37302541 DOI: 10.1016/j.drudis.2023.103667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/26/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
Therapeutics based on living organisms provide a roadmap for next-generation biomedicine. Bacteria have an essential role in the development, regulation, and treatment of gastrointestinal disease and cancer through similar mechanisms. However, primitive bacteria lack the stability to overcome complex drug delivery barriers, and their multifunctionality in reinforcing both conventional and emerging therapeutics is limited. Artificially engineered bacteria (ArtBac) with modified surfaces and genetic functions show promise for tackling these problems. Herein, we discuss recent applications of ArtBac as living biomedicine for the treatment of gastrointestinal diseases and tumors. Future perspectives are given to guide the rational design of ArtBac toward safe multifunctional medicine.
Collapse
Affiliation(s)
- Yong Liu
- School of Science, Hainan University, Haikou 570228, China
| | - Wenqin Yu
- School of Life Sciences, Hainan University, Haikou 570228, China
| | - Qian Wang
- College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, 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.
| | - Juanjuan Li
- School of Life Sciences, Hainan University, Haikou 570228, China.
| |
Collapse
|
19
|
Fang Y, Luo X, Xu Y, Liu Z, Mintz RL, Yu H, Yu X, Li K, Ju E, Wang H, Tang Z, Tao Y, Li M. Sandwich-Structured Implants to Obstruct Multipath Energy Supply and Trigger Self-Enhanced Hypoxia-Initiated Chemotherapy Against Postsurgical Tumor Recurrence and Metastasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023:e2300899. [PMID: 37156756 PMCID: PMC10401165 DOI: 10.1002/advs.202300899] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/03/2023] [Indexed: 05/10/2023]
Abstract
As a currently common strategy to treat cancer, surgical resection may cause tumor recurrence and metastasis due to residual postoperative tumors. Herein, an implantable sandwich-structured dual-drug depot is developed to trigger a self-intensified starvation therapy and hypoxia-induced chemotherapy sequentially. The two outer layers are 3D-printed using a calcium-crosslinked mixture ink containing soy protein isolate, polyvinyl alcohol, sodium alginate, and combretastatin A4 phosphate (CA4P). The inner layer is one patch of poly (lactic-co-glycolic acid)-based electrospun fibers loaded with tirapazamine (TPZ). The preferentially released CA4P destroys the preexisting blood vessels and prevents neovascularization, which obstructs the external energy supply to cancer cells but aggravates hypoxic condition. The subsequently released TPZ is bioreduced to cytotoxic benzotriazinyl under hypoxia, further damaging DNA, generating reactive oxygen species, disrupting mitochondria, and downregulating hypoxia-inducible factor 1α, vascular endothelial growth factor, and matrix metalloproteinase 9. Together these processes induce apoptosis, block the intracellular energy supply, counteract the disadvantage of CA4P in favoring intratumor angiogenesis, and suppress tumor metastasis. The in vivo and in vitro results and the transcriptome analysis demonstrate that the postsurgical adjuvant treatment with the dual-drug-loaded sandwich-like implants efficiently inhibits tumor recurrence and metastasis, showing great potential for clinical translation.
Collapse
Affiliation(s)
- Youqiang Fang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, P. R. China
| | - Xing Luo
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, P. R. China
| | - Yanteng Xu
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, P. R. China
| | - Zheng Liu
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, P. R. China
| | - Rachel L Mintz
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Haiyang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Xuan Yu
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, P. R. China
- Department of Ultrasound, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, P. R. China
| | - Kai Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, P. R. China
- Department of Ultrasound, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, P. R. China
| | - Enguo Ju
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, P. R. China
| | - Haixia Wang
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, P. R. China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, P. R. China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, P. R. China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, P. R. China
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, P. R. China
| |
Collapse
|