1
|
Cui H, Li J. Hydrogel adhesives for tissue recovery. Adv Colloid Interface Sci 2025; 341:103496. [PMID: 40168713 DOI: 10.1016/j.cis.2025.103496] [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: 10/17/2024] [Revised: 02/11/2025] [Accepted: 03/24/2025] [Indexed: 04/03/2025]
Abstract
Hydrogel adhesives (HAs) are promising and rewarding tools for improving tissue therapy management. Such HAs had excellent properties and potential applications in biological tissues, such as suture replacement, long-term administration, and hemostatic sealing. In this review, the common designs and the latest progress of HAs based on various methodologies are systematically concluded. Thereafter, how to deal with interfacial water to form a robust wet adhesion and how to balance the adhesion and non-adhesion are underlined. This review also provides a brief description of gelation strategies and raw materials. Finally, the potentials of wound healing, hemostatic sealing, controlled drug delivery, and the current applications in dermal, dental, ocular, cardiac, stomach, and bone tissues are discussed. The comprehensive insight in this review will inspire more novel and practical HAs in the future.
Collapse
Affiliation(s)
- Haohao Cui
- Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China; School of Material Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Jingguo Li
- Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China; School of Material Science and Engineering, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
2
|
Li K, Hu Q, Wang L, Wu C, Yang L, Liu G, Wang Y. Engineering of Bioprosthetic Heart Valves with Synergistic Zwitterionic Surface Modification and Zirconium Cross-linking for Improved Biocompatibility and Durability. Acta Biomater 2025:S1742-7061(25)00417-9. [PMID: 40490240 DOI: 10.1016/j.actbio.2025.06.010] [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: 11/13/2024] [Revised: 05/21/2025] [Accepted: 06/05/2025] [Indexed: 06/11/2025]
Abstract
Bioprosthetic heart valves (BHVs) are frequently utilized in surgeries for heart valve replacement to address valvular heart disease (VHD). Despite their widespread use, BHVs still face challenges in clinical applications, such as thrombosis, calcification, immune responses, poor re-endothelialization, infection, component degradation, and mechanical failure, which are largely due to the heterogeneous cross-linking effects. To address these issues, we propose a synergistic engineering strategy based on sequential zwitterionic surface modification and zirconium cross-linking to improve the biocompatibility and durability of BHVs. After surface modification via ring-opening reactions of zwitterionic epoxy copolymers (PGSB) on collagen fibers of decellularized porcine pericardium (D-PP), the zwitterionic PGSB significantly promoted the uniform transfer of zirconium ions (Zr4+) and further coordinated with Zr4+ to achieve homogeneous cross-linking between collagen fibers. Compared to conventional glutaraldehyde (GA)-cross-linked PP, PGSB/Zr-PP showed enhanced anti-thrombotic performance, attenuated immune rejection, accelerated endothelialization, and over 95% reduction in calcification after 90 days of subcutaneous implantation, collectively indicating improved biocompatibility. Furthermore, this homogeneously cross-linked PGSB/Zr-PP exhibited undetectable component degradation and simultaneous improvements in both strength and toughness, all of which are essential for improving the durability of BHVs. Intriguingly, the zwitterionic sulfobetaine groups could be converted into bactericidal quaternary ammonium groups upon coordination with Zr4+, resulting in strong antibacterial and anti-biofilm activities beneficial for preventing life-threatening prosthetic valve endocarditis. More importantly, PGSB/Zr-PP met the ISO 5840-3 standards required for BHV applications in terms of hydrodynamic performance and 200-million-cycle durability. These results demonstrate that PGSB/Zr-PP would be a promising alternative to GA-cross-linked BHVs. STATEMENT OF SIGNIFICANCE: Mainstream glutaraldehyde-cross-linked BHV face persistent clinical challenges, including thrombosis, calcification, immune response, poor re-endothelialization, infection, component degradation, and mechanical failure. Although various non-glutaraldehyde cross-linkers have been investigated, few strategies effectively address these challenges due to the heterogeneous nature of cross-linking. Herein, we present a synergistic engineering strategy based on sequential zwitterionic surface modification and zirconium cross-linking. This strategy produces homogeneously cross-linked BHVs with comprehensive improvements in anti-thrombogenicity, immune compatibility, endothelialization, resistance to calcification and infection, enzymatic stability, and mechanical strength. Notably, the aortic BHV fabricated via this method met the ISO 5840-3 standards for hydrodynamic performance and durability, demonstrating its long-term clinical potential.
Collapse
Affiliation(s)
- Kaijun Li
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, PR China; Key Laboratory of Leather Chemistry and Engineering of Ministry of Education, Sichuan University; Chengdu, 610065, PR China
| | - Qinsheng Hu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, PR China; Department of Orthopedic Surgery, Ya'an People's Hospital, Ya'an 625000, China
| | - Ling Wang
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, PR China; Key Laboratory of Leather Chemistry and Engineering of Ministry of Education, Sichuan University; Chengdu, 610065, PR China
| | - Chengcheng Wu
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, PR China; Key Laboratory of Leather Chemistry and Engineering of Ministry of Education, Sichuan University; Chengdu, 610065, PR China
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, PR China.
| | - Gongyan Liu
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, PR China; Key Laboratory of Leather Chemistry and Engineering of Ministry of Education, Sichuan University; Chengdu, 610065, PR China.
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, PR China
| |
Collapse
|
3
|
Guan Q, Chen J, Liu Z, Xin J, Chen Y, Wang P, Liu J. Biocompatible cloaking of bacteria for effective tumor imaging and therapy. Mater Today Bio 2025; 32:101788. [PMID: 40321697 PMCID: PMC12049847 DOI: 10.1016/j.mtbio.2025.101788] [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: 02/20/2025] [Revised: 04/08/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025] Open
Abstract
Bacteria possess distinctive characteristics, such as tropism, motility, and genetic editability, which make them highly attractive for biomedical applications, such as tumor imaging and therapy. However, the immunogenicity triggered by endotoxins may lead to severe adverse effects and prompt quick elimination in the body, thus restricting their clinical applications. In this study, we described a double-layer coating technique employing tannic acid (TA) and albumin (BSA) for bacteria encapsulation. This compact coating effectively shields endotoxin exposure and inhibits endotoxin leakage from bacteria, exhibiting a favorable safety profile. Moreover, bacteria coated with BSA have superior biocompatibility with their surroundings, which prevents phagocytes from eliminating the bacteria, hence prolonging the reservation in vivo. As bacteria grow, the BSA-TA layer progressively detaches after reaching targeted sites, allowing free bacteria to exploit their own advantages. Importantly, the BSA-TA coating strategy protects bacteria against various environmental assaults without compromising their growth, proliferation, or motility, maintaining their inherent characteristics. In murine tumor models, the BSA-TA-coated bacteria demonstrated enhanced long-term tumor imaging and therapeutic efficacy against tumors. Together, the BSA-TA coating strategy improves the biocompatibility of bacteria and has the capacity to expand the range of bacteria for biomedical applications.
Collapse
Affiliation(s)
- Qing Guan
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Junjie Chen
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Zhaonan Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Jianfeng Xin
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Yu Chen
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Peng Wang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Jun Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| |
Collapse
|
4
|
Wang M, Li D, Ouyang S, Tong B, Chen Y, Ding B, Wang J, Jiang Z, Xu H, Hu S. Hydrogel derived from decellularized pig small intestine submucosa boosted the therapeutic effect of FGF-20 on TNBS-induced colitis in rats via restoring gut mucosal integrity. Mater Today Bio 2025; 32:101783. [PMID: 40321695 PMCID: PMC12049826 DOI: 10.1016/j.mtbio.2025.101783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 04/07/2025] [Accepted: 04/19/2025] [Indexed: 05/08/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by impaired intestinal mucosal barrier function, leading to persistent inflammation and tissue damage. Current therapies often fail to address barrier dysfunction, highlighting the need for innovative treatments. This study developed a novel therapeutic strategy by combining decellularized porcine small intestinal submucosa (D-SIS) with fibroblast growth factor 20 (FGF-20) to promote mucosal repair and restore barrier integrity in a TNBS-induced colitis rat model. The D-SIS-based hydrogel, supplemented with hyaluronic acid (HA), was designed to enhance FGF-20 stability and enable sustained drug release. Results showed that the FGF-20-loaded hydrogel (MAF) exhibited excellent rheological properties, erosion resistance, and controlled drug release, making it suitable for rectal administration. In vitro cell experiments demonstrated that MAF enhanced Caco-2 cell proliferation, migration, and tight junction protein expression, restoring epithelial barrier integrity. In the colitis model, MAF significantly reduced disease activity index (DAI) scores, attenuated inflammation, and restored mucosal morphology. Additionally, MAF promoted goblet cell regeneration, enhanced mucus secretion, and upregulated intestinal stem cell markers, indicating its ability to repair both epithelial and mucus barriers. In conclusion, the MAF hydrogel represents a promising therapeutic approach for UC by combining the regenerative properties of FGF-20 with the bioactive support of D-SIS.
Collapse
Affiliation(s)
- Minmin Wang
- Department of Gastrointestinal Surgery Nursing Unit, Ward 442, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, China
| | - Dingwei Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325035, China
| | - Shenyuan Ouyang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325035, China
| | - Bingjie Tong
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325035, China
| | - Yumo Chen
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325035, China
| | - Bingyu Ding
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325035, China
| | - Jie Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325035, China
| | - Zhijiang Jiang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325035, China
| | - Helin Xu
- 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
| |
Collapse
|
5
|
Zhang Y, Wang Y, Lu Y, Quan H, Wang Y, Song S, Guo H. Advanced oral drug delivery systems for gastrointestinal targeted delivery: the design principles and foundations. J Nanobiotechnology 2025; 23:400. [PMID: 40448152 DOI: 10.1186/s12951-025-03479-8] [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: 03/31/2025] [Accepted: 05/20/2025] [Indexed: 06/02/2025] Open
Abstract
Oral administration has long been considered the most convenient method of drug delivery, requiring minimal expertise and invasiveness. Unlike injections, it avoids discomfort, wound infections, and complications, leading to higher patient compliance. However, the effectiveness of oral delivery is often hindered by the harsh biological barriers of the gastrointestinal tract, which limit the bioaccessibility and bioavailability of drugs. The development of oral drug delivery systems (ODDSs) represents a critical area for the advancement of pharmacotherapy. This review highlights the characteristics and precise targeting mechanisms of ODDSs. It first examines the unique properties of each gastrointestinal compartment, including the stomach, small intestine, intestinal mucus, intestinal epithelial barrier, and colon. Based on these features, it outlines the targeting strategies and design principles for ODDSs aimed at overcoming gastrointestinal barriers to enhance disease treatment. Lastly, the review discusses the challenges and potential future directions for ODDS development, emphasizing their importance for advancing drug delivery technologies and accelerating their future growth.
Collapse
Affiliation(s)
- Yafei Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Yiran Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Yao Lu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Heng Quan
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100089, China
| | - Yuqi Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Sijia Song
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100089, China
| | - Huiyuan Guo
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China.
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100089, China.
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|
6
|
Ma C, Yang J, Fu XN, Luo JY, Liu P, Zeng XL, Li XY, Zhang SL, Zheng S. Microbial characteristics of gut microbiome dysbiosis in patients with chronic liver disease. World J Hepatol 2025; 17:106124. [DOI: 10.4254/wjh.v17.i5.106124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/28/2025] [Accepted: 04/24/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND In this study, we are committed to exploring the characteristics of the gut microbiome in three different stages of chronic liver disease (CLD): Chronic hepatitis B, liver cirrhosis, and hepatocellular carcinoma (HCC).
AIM To delineate the gut microbiota traits in individuals with chronic liver ailments (chronic hepatitis B, cirrhosis, HCC), scrutinizes microbiome alterations during the progression of these diseases, and assesses microbiome disparities among various Child-Pugh categories in cirrhosis sufferers.
METHODS A cohort of 60 CLD patients from the Third People’s Hospital of Yunnan Province were recruited from February to August 2023, together with 37 healthy counterparts. Employing 16SrDNA high-throughput sequencing, we evaluated the diversity and composition of the gut microbiota.
RESULTS Compared to healthy subjects, patients exhibited a reduced presence of Firmicutes and a corresponding decline in butyrate-producing genera. In contrast, an upsurge in Proteobacteria was observed in the diseased cohorts, particularly an increase in Enterobacteriaceae that intensified with the disease's progression. At the genus level, the occurrence of Escherichia_Shigella, Parabacteroides, Streptococcus, Klebsiella, and Enterococcus was higher, with Escherichia_Shigella numbers augmenting as the disease advanced. Furthermore, in cirrhosis patients, an increase in Proteobacteria was noted as liver reserve diminished, alongside a decrease in Ruminococcaceae and Bacteroidaceae.
CONCLUSION The reduced abundance of short-chain fatty acid-producing bacteria in the intestine, alongside the increased abundance of gram-negative bacteria such as Escherichia_Shigella and Parabacteroides, may promote the progression of CLD.
Collapse
Affiliation(s)
- Chi Ma
- Department of Gastroenterology, The Second Affiliated Hospital of Dali University, Kunming 650011, Yunnan Province, China
| | - Juan Yang
- Department of Gastroenterology, The Third People’s Hospital of Yunnan Province, Kunming 650011, Yunnan Province, China
| | - Xin-Nian Fu
- Department of Gastroenterology, The Second Affiliated Hospital of Dali University, Kunming 650011, Yunnan Province, China
| | - Jiang-Yan Luo
- Department of Gastroenterology, The Second Affiliated Hospital of Dali University, Kunming 650011, Yunnan Province, China
| | - Pei Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Dali University, Kunming 650011, Yunnan Province, China
| | - Xue-Li Zeng
- Department of Gastroenterology, The Second Affiliated Hospital of Dali University, Kunming 650011, Yunnan Province, China
| | - Xin-Yi Li
- Department of Gastroenterology, The Second Affiliated Hospital of Dali University, Kunming 650011, Yunnan Province, China
| | - Shun-Ling Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Dali University, Kunming 650011, Yunnan Province, China
| | - Sheng Zheng
- Department of Gastroenterology, The Third People’s Hospital of Yunnan Province, Kunming 650011, Yunnan Province, China
| |
Collapse
|
7
|
Nalinbenjapun S, Sripetthong S, Basit A, Chaimongkolnukul K, Sajomsang W, Ovatlarnporn C. Synthesis and characterization of 5-aminosalicylic acid-azobenzyl-chitosan conjugates-based prodrugs for the colon targeted delivery in dextran sodium sulfate (DSS)-induced ulcerative colitis. Int J Biol Macromol 2025; 318:144734. [PMID: 40441567 DOI: 10.1016/j.ijbiomac.2025.144734] [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: 12/18/2024] [Revised: 05/02/2025] [Accepted: 05/26/2025] [Indexed: 06/16/2025]
Abstract
Site-specific colon drug delivery is crucial in the treatment of colon-localized diseases. In this study, prodrugs of 5-aminosalicylic acid (5-ASA), the drug of choice in ulcerative colitis (UC), were synthesized by conjugating with chitosan having molecular weights (MW) of 30, 80 and 300 kDa to achieve targeted delivery to the colon. FT-IR and UV-Vis analyses confirmed the successful synthesis and varying loading capacities of 5-ASA, with a maximum loading capacity of 11.32 ± 2.0 %. The obtained conjugates (7-9) exhibited gradual drug release characteristics, with up to 30 % of the drug content released after 24 h, in the simulated colonic fluid containing rat gastrointestinal (GI) tract homogenates, demonstrating their colon-specific and slow-release properties. The conjugates were non-toxic to normal human colon epithelial cells at concentrations up to 5-10 μg/mL, suggesting a favorable safety profile. Additionally, conjugate (7) was tested for its efficacy in a mouse model of ulcerative colitis. Necropsy results showed no significant structural changes or lesions in animals treated with conjugate (7). Histopathological analysis revealed mild, multifocal lymphoplasmacytic infiltrates and scattered eosinophils in the lamina propria, indicating a low level of inflammation. Overall, the findings suggest that 5-ASA-azobenzyl-chitosan conjugates hold promise as a potential therapeutic option for ulcerative colitis and other colon localized disorders, with minimal side effects due to reduced systemic exposure.
Collapse
Affiliation(s)
- Sirinporn Nalinbenjapun
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand; Drug Delivery System Research Excellent Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Sasikarn Sripetthong
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand; Drug Delivery System Research Excellent Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Abdul Basit
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand; Drug Delivery System Research Excellent Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand; Department of Medicinal Chemistry, College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, Guangdong, China.
| | | | - Warayuth Sajomsang
- Nanodelivery System Laboratory, National Nanotechnology Center, National Science and Technology Development Agency, Phathum Thani 12120, Thailand
| | - Chitchamai Ovatlarnporn
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand; Drug Delivery System Research Excellent Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand.
| |
Collapse
|
8
|
Wang M, Li J, Ning X, Yang T, Guo X, Dong X, Guo Q, Zhu B, Liu Z. Mechanism of iron (III) ion-induced gels of Ulva lactuca polysaccharides and application in probiotic encapsulation. Int J Biol Macromol 2025; 315:144554. [PMID: 40412704 DOI: 10.1016/j.ijbiomac.2025.144554] [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: 12/25/2024] [Revised: 04/27/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
Ulva lactuca polysaccharides (ULP) have attracted growing interests for their potential applications in the fields of therapeutics, nutraceuticals and food science. However, its gelling properties and applications in probiotic encapsulation are not well understood. In this study, the effects of iron (III) ions (Fe3+) as cross-linking agent to promote the gel formation of ULP and its related mechanisms were investigated. The results showed that Fe3+ could form a dense gelling structure with ULP, while Na+ and Ca2+ did not have similar effects. Rheological analysis indicated that the Fe3+-ULP gel exhibited remarkable resilience under heating conditions, maintaining its gelling characteristics upon thermal cycling up to 85 °C. FT-IR spectroscopy revealed that hydroxyl and carboxyl groups play critical roles in Fe3+-ULP gelation, showing red and blue shifts, respectively with Fe3+ addition in FT-IR absorbance. As for its application in probiotic encapsulation, the encapsulated Fe3+-ULP gel microspheres provide better protection against low pH (2.0) and bile salt (0.3 mg/mL) damage than that for native probiotics (L. gasseri). These findings integrate the use of ULP as both gelling agent and probiotics, offering novel insights into the potential applications of ULP in the field of encapsulation and delivery systems.
Collapse
Affiliation(s)
- Menghui Wang
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Guangdong Engineering Technology Research Center of Aquatic Food Processing and Safety Control, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China
| | - Jinjin Li
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Guangdong Engineering Technology Research Center of Aquatic Food Processing and Safety Control, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China
| | - Xiner Ning
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Guangdong Engineering Technology Research Center of Aquatic Food Processing and Safety Control, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China
| | - Tingqi Yang
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Guangdong Engineering Technology Research Center of Aquatic Food Processing and Safety Control, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China
| | - Xiaoming Guo
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Guangdong Engineering Technology Research Center of Aquatic Food Processing and Safety Control, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China
| | - Xiuping Dong
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Guangdong Engineering Technology Research Center of Aquatic Food Processing and Safety Control, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China; National Engineering Research Center of Seafood, National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Qingbin Guo
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Guangdong Engineering Technology Research Center of Aquatic Food Processing and Safety Control, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China
| | - Beiwei Zhu
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Guangdong Engineering Technology Research Center of Aquatic Food Processing and Safety Control, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China; National Engineering Research Center of Seafood, National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China.
| | - Zhengqi Liu
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Guangdong Engineering Technology Research Center of Aquatic Food Processing and Safety Control, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
9
|
Zhao N, Han YJ, Wang C, Li J, Song LH, Lv LP, Ma P, Deng J, Zhang YY. Two Birds with One Stone: Empowering Probiotic with Nanoenzyme for the Treatment of Inflammatory and Anemia through Oral Administration. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40395049 DOI: 10.1021/acsami.5c04452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
In the context of ulcerative colitis (UC), iron deficiency anemia (IDA) is often presented as a prevalent systemic manifestation. However, there is an absence of an effective strategy for the specific case of UC with IDA. Herein, mendelian randomization (MR) analysis is applied to confirm the causal association between UC and iron-related conditions. Accordingly, we have developed a probiotic-based therapeutic approach that synergistically alleviates inflammation and IDA. Probiotic Escherichia coli Nissle 1917 (EcN) is functionalized by the Fe3O4 nanoenzyme and hyaluronan (HA) through electrostatic layer-by-layer (LBL) adsorption. As expected, the obtained EcN@Fe3O4/HA exhibits excellent properties in vitro, such as gastric acid resistance and ROS-scavenging capability. Upon oral administration, EcN@Fe3O4/HA shows remarkable adhesion in vivo, particularly in inflamed mice. Moreover, EcN@Fe3O4/HA shows a "two birds with one stone" effect in dextran sulfate sodium (DSS)-induced mice. First, it exerts anti-inflammatory effects through promoting the expression of tight junction proteins and regulating the gut microbiota. Second, it addresses the issue of IDA. EcN@Fe3O4/HA effectively ameliorates IDA in DSS-induced mice through iron supplementation, EPO upregulation, and iron homeostasis modulation, resulting in enhanced RBC morphology and elevated cell counts. Therefore, the proposed strategy provides inspiration for future management of diseases and related complications.
Collapse
Affiliation(s)
- Ning Zhao
- Academy of Military Medical Sciences, Beijing 100850, People's Republic of China
| | - Yong-Jiao Han
- Academy of Military Medical Sciences, Beijing 100850, People's Republic of China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, People's Republic of China
| | - Chaojie Wang
- Academy of Military Medical Sciences, Beijing 100850, People's Republic of China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, People's Republic of China
| | - Jiaxu Li
- Academy of Military Medical Sciences, Beijing 100850, People's Republic of China
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, People's Republic of China
| | - Ling-Hui Song
- Academy of Military Medical Sciences, Beijing 100850, People's Republic of China
| | - Li-Ping Lv
- Academy of Military Medical Sciences, Beijing 100850, People's Republic of China
| | - Ping Ma
- Academy of Military Medical Sciences, Beijing 100850, People's Republic of China
| | - Jiang Deng
- Academy of Military Medical Sciences, Beijing 100850, People's Republic of China
| | - Yan-Yu Zhang
- Academy of Military Medical Sciences, Beijing 100850, People's Republic of China
| |
Collapse
|
10
|
Song J, Liu W, Yang L, Sun X, Xie L, Li J, Xu L, Dong A. A NIR-Responsive Deep Penetration Phototherapy Strategy for Treating Infected Skin Defect via Antibacterial Effect and Inflammation Elimination. Adv Healthc Mater 2025:e2500987. [PMID: 40394922 DOI: 10.1002/adhm.202500987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 05/07/2025] [Indexed: 05/22/2025]
Abstract
The increasing severity of antibiotic resistance and the delayed healing of infected wounds have triggered an arduous challenge that threatens human health. Instantly, quiet a few novel, efficient, and safe antibacterial strategies are urgently needed to be explored. In this study, a NIR-activated antibacterial nanocomposite (RB/UCNPs@BP) integrating rose bengal-sensitized upconversion nanoparticles (RB/UCNPs) and black phosphorus (BP) is developed for promoting infection wound healing. The photodynamic therapy (PDT) and photothermal therapy (PTT) are employed here for synergistic antibacterial action, while UCNPs further improve the penetration depth of irradiation and treatment efficiency. More importantly, the typical biodegradability of BP confers reduced resistance on nanocomposites through residual-free antimicrobial methods. The results show that RB/UCNPs@BP significantly inhibits the growth of both Escherichia coli (E.coli) and Staphylococcus aureus (S.aureus) via enhanced PDT and PTT. Besides, the infected wounds achieve better healing by accelerating fibroblast proliferation and migration, reducing inflammatory cell infiltration, and promoting neuronal regeneration and angiogenesis. This study provides a promising and anti-resistant strategy with light-triggered antibacterial and anti-inflammatory activities that can promote the regeneration of infected skin tissue.
Collapse
Affiliation(s)
- Jinze Song
- College of Chemistry and Materials Science, Inner Mongolia Minzu University, Tongliao, 028000, P. R. China
| | - Wenxin Liu
- College of Chemistry and Materials Science, Inner Mongolia Minzu University, Tongliao, 028000, P. R. China
| | - Li Yang
- Department of Functional Laboratory, Tongliao People's Hospital, Tongliao, 028000, P. R. China
| | - Xiao Sun
- Department of Functional Laboratory, Tongliao People's Hospital, Tongliao, 028000, P. R. China
| | - Liangliang Xie
- College of Chemistry and Materials Science, Inner Mongolia Minzu University, Tongliao, 028000, P. R. China
| | - Jiawei Li
- College of Chemistry and Materials Science, Inner Mongolia Minzu University, Tongliao, 028000, P. R. China
| | - Ling Xu
- College of Chemistry and Materials Science, Inner Mongolia Minzu University, Tongliao, 028000, P. R. China
| | - Alideertu Dong
- College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot, 010021, P. R. China
| |
Collapse
|
11
|
Chen Y, Bi S, Zhang X, Chen J, Xin J, Liu Z, Guan Q, Qiu P, Wang P, Liu J. Engineered probiotics remodel the intestinal epithelial barrier and enhance bacteriotherapy for inflammatory bowel diseases. Acta Biomater 2025; 198:467-481. [PMID: 40210182 DOI: 10.1016/j.actbio.2025.04.016] [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: 12/31/2024] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
Inflammatory bowel diseases (IBDs) are often associated with compromised epithelial barriers and dysregulated gut microbiota. In this study, we revealed the synergistic effect that zinc and indole-3-carbinol (I3C) have in restoring the epithelial barrier, and co-localized them on a ZI platform, which was further conjugated to the surface of Escherichia coli Nissle 1917 (EcN). The ZI@EcN formulation effectively delivered ZI to colon tissues and extended its retention in the intestines due to the colonic colonization effect of EcN, thereby promoting the sustained release of zinc and I3C for optimal synergistic effects on epithelial barrier remodeling. The restored epithelium acts as a protective barrier, preventing the infiltration of toxins and pathogens, which significantly reduces inflammation in colonic tissues. Additionally, EcN enriched the gut microbiome, increasing the abundance of beneficial bacteria while reducing that of pathogens, demonstrating its significant efficacy in gut microbiome regulation. In dextran sulfate sodium-induced mouse colitis models, ZI@EcN exhibited substantially improved prophylactic and therapeutic efficacy with favorable safety profiles, highlighting its potential for clinical applications. STATEMENT OF SIGNIFICANCE: This study highlighted the synergistic effects that zinc and indole-3-carbinol, both derived from dietary sources, have on restoring integrity of the intestinal epithelial barrier. A platform (ZI@EcN) was also developed for the targeted delivery and sustained release of zinc and indole-3-carbinol, specifically in colonic tissues, for colitis treatment. This platform not only restores the compromised intestinal epithelial barrier but also regulates the dysbiotic gut microbiota, promoting the recovery of a healthy intestinal microenvironment and showing promise in alleviating complex symptoms in a single formulation. Furthermore, the formulation demonstrated potent prophylactic and therapeutic efficacy against colitis, with favorable safety profiles, and a strong potential for clinical applications.
Collapse
Affiliation(s)
- Yu Chen
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Shijie Bi
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Xiaoli Zhang
- Department of Integrated Traditional Chinese and Western Medicine, the People's Hospital of Huaiyin, Jinan City, Jinan 250000, China
| | - Junjie Chen
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Jianfeng Xin
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Zhaonan Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Qing Guan
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Peiju Qiu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Peng Wang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Jun Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China.
| |
Collapse
|
12
|
Shi JY, Wang YJ, Bao QW, Qin YM, Li PP, Wu QQ, Xia CK, Wu DL, Xie SZ. Polygonatum cyrtonema Hua polysaccharide alleviates ulcerative colitis via gut microbiota-independent modulation of inflammatory immune response. Carbohydr Polym 2025; 356:123387. [PMID: 40049966 DOI: 10.1016/j.carbpol.2025.123387] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/30/2025] [Accepted: 02/13/2025] [Indexed: 05/13/2025]
Abstract
Polygonatum cyrtonema polysaccharides (PCP) exhibit ameliorative effects on colitis. However, whether the protective effects of PCP depend on the gut microbiota and how PCP regulates intestinal immune responses to alleviate colitis remain unclear. Therefore, this study investigated the effect of PCP against colitis focusing on the regulation of intestinal immune response. The PCP structure was reclassified as fructan. PCP treatment significantly reduced the symptoms of colitis. PCP restored IgA, ZO-1, Occludin, and MUC2 expression to enhance intestinal barrier function. Oral PCP administration markedly inhibited excessive inflammation-mediated immune response by modulating inflammatory cytokines secretion and Th17/Tregs cell balance and restored gut microbial composition. Interestingly, PCP still had a significant ameliorating effect on intestinal inflammation in colitis mice with gut microbial depletion by antibiotics. In the Caco-2/RAW264.7 co-culture inflammation model, PCP treatment improved the intestinal epithelial barrier function by regulating the inflammatory immune response through signal transduction pathways. Overall, these findings suggested that the alleviating effects of PCP on colitis are independent of gut microbiota, and that PCP can directly modulate the inflammatory immune response and intestinal barrier function, which in turn regulates gut microbiota. These findings will provide new insights into the action mechanism of natural polysaccharides in relieving colitis.
Collapse
Affiliation(s)
- Jin-Yang Shi
- School of Pharmacy, Anhui Province Key Laboratory of Bioactive Natural Products, Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Yong-Jian Wang
- School of Pharmacy, Anhui Province Key Laboratory of Bioactive Natural Products, Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Qian-Wen Bao
- School of Pharmacy, Anhui Province Key Laboratory of Bioactive Natural Products, Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Ya-Min Qin
- School of Pharmacy, Anhui Province Key Laboratory of Bioactive Natural Products, Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Pei-Pei Li
- School of Pharmacy, Anhui Province Key Laboratory of Bioactive Natural Products, Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Qiao-Qiao Wu
- School of Pharmacy, Anhui Province Key Laboratory of Bioactive Natural Products, Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - Cheng-Kai Xia
- School of Pharmacy, Anhui Province Key Laboratory of Bioactive Natural Products, Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China
| | - De-Ling Wu
- School of Pharmacy, Anhui Province Key Laboratory of Bioactive Natural Products, Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China; Bozhou University, Bozhou, Anhui 236800, China.
| | - Song-Zi Xie
- School of Pharmacy, Anhui Province Key Laboratory of Bioactive Natural Products, Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Anhui University of Chinese Medicine, Hefei, Anhui 230012, China.
| |
Collapse
|
13
|
Wang J, Li J, Yu W, Wang G, Cifuentes A, Ibañez E, Lu W. Microalgal proteins: Extraction, interfacial properties, bioactivities, and future perspectives - A review. Food Chem 2025; 486:144680. [PMID: 40373600 DOI: 10.1016/j.foodchem.2025.144680] [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: 02/19/2025] [Revised: 04/25/2025] [Accepted: 05/07/2025] [Indexed: 05/17/2025]
Abstract
The increasing global demand for protein, coupled with concerns over the environmental sustainability of animal-derived sources, has prompted the search for alternative protein sources. Microalgae have emerged as a promising solution due to their high productivity, protein content, and ability to grow in non-arable environments or photobioreactors. Their proteins, hydrolysates and peptides exhibit diverse bioactivities, including anti-obesity, anti-cancer, antioxidant, and anti-hypertensive effects, as well as functional properties such as emulsification, foaming and gelling. However, their practical utilization is hindered by challenges such as high production costs and environmental sensitivity, particularly in relation to pH, temperature, and light, which can affect their structural stability and functional performance. This review summarizes traditional and innovative extraction techniques, discusses the structure-function relationships of these microalgal components, and highlights their potential applications. Furthermore, it identifies key production and commercialization challenges, proposing strategies to enhance extraction efficiency and environmental stability during processing and storage, thereby facilitating broader industrial implementation.
Collapse
Affiliation(s)
- Junwen Wang
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450003, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin Institute of Technology, Harbin 150001, China
| | - Jiangfei Li
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450003, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin Institute of Technology, Harbin 150001, China
| | - Wenchen Yu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Ge Wang
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China
| | - Alejandro Cifuentes
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, CSIC, Nicolás Cabrera 9, 28049 Madrid, Spain
| | - Elena Ibañez
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, CSIC, Nicolás Cabrera 9, 28049 Madrid, Spain
| | - Weihong Lu
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, China; Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450003, China; National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental Nutrients, Harbin Institute of Technology, Harbin 150001, China.
| |
Collapse
|
14
|
Wang Z, Xu Q, Hou L, He Z, Christian M, Dai X. Food-derived polysaccharides and anti-obesity effects through enhancing adipose thermogenesis: structure-activity relationships, mechanisms, and regulation of gut microecology. Crit Rev Food Sci Nutr 2025:1-22. [PMID: 40351151 DOI: 10.1080/10408398.2025.2500675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Polysaccharides represent a crucial and extensively utilized bioactive fraction in natural products, which are employed in the treatment of metabolic disorders due to their significant therapeutic potential. Recently, food-derived polysaccharides (FPs) have emerged as significant substances in obesity management, valued for their ability to activate thermogenic fat. This review discusses the correlation between the structural features of FPs and their efficacy in combating obesity. Moreover, the molecular mechanism by which FPs regulate thermogenic fat and how the intestinal microecology induces thermogenic fat activity is elucidated. The anti-obesity effects of FPs depend on their structure, including molecular weight, composition, linkages, conformation, and branching. Furthermore, FPs regulate fat thermogenesis via multiple mechanisms, including AMPK, p38, AKT, PGC-1α-FNDC5/irisin, and miRNA signaling pathways. Importantly, gut microbiota, together with its associated metabolites and gut-derived hormones, are pivotal in the regulatory control of brown fat by FPs. This work provides an in-depth examination of how adipose tissue thermogenesis contributes to the anti-obesity effects of FPs, shedding light on their potential in preventing obesity and informing the formulation of natural weight-loss remedies.
Collapse
Affiliation(s)
- Zhenyu Wang
- College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Qiyu Xu
- College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Lijuan Hou
- College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Zhiyong He
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
| | - Mark Christian
- School of Science and Technology, Nottingham Trent University, Clifton, Nottingham, United Kingdom
| | - Xianjun Dai
- College of Life Sciences, China Jiliang University, Hangzhou, China
| |
Collapse
|
15
|
Wang X, Zhang H, Wan Z, Li X, Ibáñez CF, Xie M. A single-cell transcriptomic atlas of all cell types in the brain of 5xFAD Alzheimer mice in response to dietary inulin supplementation. BMC Biol 2025; 23:124. [PMID: 40346662 PMCID: PMC12065180 DOI: 10.1186/s12915-025-02230-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 04/30/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disease that is a major threat to the aging population. Due to lack of effective therapy, preventive treatments are important strategies to limit AD onset and progression, of which dietary regimes have been implicated as a key factor. Diet with high fiber content is known to have beneficial effects on cognitive decline in AD. However, a global survey on microbiome and brain cell dynamics in response to high fiber intake at single-cell resolution in AD mouse models is still missing. RESULTS Here, we show that dietary inulin supplementation synergized with AD progression to specifically increase the abundance of Akkermansia muciniphila in gut microbiome of 5 × Familial AD (FAD) mice. By performing single-nucleus RNA sequencing on different regions of the whole brain with three independent biological replicates, we reveal region-specific changes in the proportion of neuron, astrocyte, and granule cell subpopulations upon inulin supplementation in 5xFAD mice. In addition, we find that astrocytes have more pronounced region-specific diversity than microglia. Intriguingly, such dietary change reduces amyloid-β plaque burden and alleviates microgliosis in the forebrain region, without affecting the spatial learning and memory. CONCLUSIONS These results provide a comprehensive overview on the transcriptomic changes in individual cells of the entire mouse brain in response to high fiber intake and a resourceful foundation for future mechanistic studies on the influence of diet and gut microbiome on the brain during neurodegeneration.
Collapse
Affiliation(s)
- Xiaoyan Wang
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Chinese Institute for Brain Research, Zhongguancun Life Science Park, Beijing, 102206, China
| | - Houyu Zhang
- Chinese Institute for Brain Research, Zhongguancun Life Science Park, Beijing, 102206, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Zhou Wan
- School of Life Sciences, Peking University, Beijing, 100871, China
| | - Xuetong Li
- School of Life Sciences, Peking University, Beijing, 100871, China
| | - Carlos F Ibáñez
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
- Chinese Institute for Brain Research, Zhongguancun Life Science Park, Beijing, 102206, China.
- School of Life Sciences, Peking University, Beijing, 100871, China.
- Peking-Tsinghua Center for Life Sciences, Beijing, 100871, China.
- PKU-IDG/McGovern Institute for Brain Research, Beijing, 100871, China.
- Department of Neuroscience, Karolinska Institute, 17165, Stockholm, Sweden.
| | - Meng Xie
- Peking-Tsinghua Center for Life Sciences, Beijing, 100871, China.
- PKU-IDG/McGovern Institute for Brain Research, Beijing, 100871, China.
- Beijing Key Laboratory of Behavior and Mental Health, School of Psychological and Cognitive Sciences, Peking University, Beijing, 100871, China.
- Department of Medicine Huddinge, Karolinska Institute, 14183, Stockholm, Sweden.
| |
Collapse
|
16
|
Jiménez-González C, Alonso-Peña M, Argos Vélez P, Crespo J, Iruzubieta P. Unraveling MASLD: The Role of Gut Microbiota, Dietary Modulation, and AI-Driven Lifestyle Interventions. Nutrients 2025; 17:1580. [PMID: 40362889 PMCID: PMC12073168 DOI: 10.3390/nu17091580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Revised: 05/01/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
Gut microbiota has a crucial role in the pathophysiology of metabolic-associated steatotic liver disease (MASLD), influencing various metabolic mechanisms and contributing to the development of the disease. Dietary interventions targeting gut microbiota have shown potential in modulating microbial composition and mitigating MASLD progression. In this context, the integration of multi-omics analysis and artificial intelligence (AI) in personalized nutrition offers new opportunities for tailoring dietary strategies based on individual microbiome profiles and metabolic responses. The use of chatbots and other AI-based health solutions offers a unique opportunity to democratize access to health interventions due to their low cost, accessibility, and scalability. Future research should focus on the clinical validation of AI-powered dietary strategies, integrating microbiome-based therapies and precision nutrition approaches. Establishing standardized protocols and ethical guidelines will be crucial for implementing AI in MASLD management, paving the way for a more personalized, data-driven approach to disease prevention and treatment.
Collapse
Affiliation(s)
- Carolina Jiménez-González
- Gastroenterology and Hepatology Department, Clinical and Translational Research in Digestive Diseases, Valdecilla Research Institute (IDIVAL), Marqués de Valdecilla University Hospital, 39011 Santander, Spain; (C.J.-G.); (M.A.-P.); (P.A.V.); (P.I.)
| | - Marta Alonso-Peña
- Gastroenterology and Hepatology Department, Clinical and Translational Research in Digestive Diseases, Valdecilla Research Institute (IDIVAL), Marqués de Valdecilla University Hospital, 39011 Santander, Spain; (C.J.-G.); (M.A.-P.); (P.A.V.); (P.I.)
- Departamento de Anatomía y Biología Celular, Universidad de Cantabria, 39011 Santander, Spain
| | - Paula Argos Vélez
- Gastroenterology and Hepatology Department, Clinical and Translational Research in Digestive Diseases, Valdecilla Research Institute (IDIVAL), Marqués de Valdecilla University Hospital, 39011 Santander, Spain; (C.J.-G.); (M.A.-P.); (P.A.V.); (P.I.)
| | - Javier Crespo
- Gastroenterology and Hepatology Department, Clinical and Translational Research in Digestive Diseases, Valdecilla Research Institute (IDIVAL), Marqués de Valdecilla University Hospital, 39011 Santander, Spain; (C.J.-G.); (M.A.-P.); (P.A.V.); (P.I.)
| | - Paula Iruzubieta
- Gastroenterology and Hepatology Department, Clinical and Translational Research in Digestive Diseases, Valdecilla Research Institute (IDIVAL), Marqués de Valdecilla University Hospital, 39011 Santander, Spain; (C.J.-G.); (M.A.-P.); (P.A.V.); (P.I.)
| |
Collapse
|
17
|
Chaves LS, Oliveira ACP, Oliveira AP, Lopes ALF, Araujo AKS, Pacheco G, Silva KC, Martins FEC, Gomes IAB, Ramos SVS, Viana HTMC, Batista AVF, Oliveira BC, Nicolau LAD, Ribeiro FOS, Castro AV, de Araujo-Nobre AR, Silva DA, Cordeiro LMC, Góis MB, Medeiros JVR. Cashew gum fractions protect intestinal mucosa against shiga toxin-producing Escherichia coli infection: Characterization and insights into microbiota modulation. Int J Biol Macromol 2025; 311:143916. [PMID: 40324507 DOI: 10.1016/j.ijbiomac.2025.143916] [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: 02/03/2025] [Revised: 04/28/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
Diarrheal diseases remain a major public health concern, particularly in regions with poor sanitation. Polysaccharides extracted from natural gums have been investigated as functional agents for intestinal health, and their fractionation enables the production of oligosaccharides with potential prebiotic activity. This study aimed to produce cashew gum (CG) fractions through Smith degradation (CGD48) and partial hydrolysis (CGD24) and to evaluate their ability to modulate and protect the intestinal microbiota. Balb/c mice were administered CG (1200 mg/kg), CGD24 (800 mg/kg), or CGD48 (800 mg/kg) for 10 and 26 days, followed by infection with Shiga toxin-producing Escherichia coli (STEC) (5 × 1010 CFU/mL) for three days. Characterization assays confirmed the fragmentation of CG. Both CGD24 and CGD48 promoted the growth of beneficial bacteria with and without infection and reduced STEC colonization. Furthermore, they preserved mucin levels in the cecum and large intestine and maintained baseline levels of superoxide dismutase (SOD), suggesting protection of the intestinal mucosa. These findings indicate that CG fractions exhibit microbiota-modulating and protective effects against STEC, highlighting their therapeutic potential and the need for further studies to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Letícia S Chaves
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Antonio C P Oliveira
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Ana P Oliveira
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - André L F Lopes
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Andreza K S Araujo
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Gabriella Pacheco
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Katriane C Silva
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Francisco E C Martins
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Isaac A B Gomes
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Sabrine V S Ramos
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Hémilly T M C Viana
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Ana V F Batista
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Beatriz C Oliveira
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Lucas A D Nicolau
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Fábio O S Ribeiro
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Auricélia V Castro
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Alyne Rodrigues de Araujo-Nobre
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Durcilene A Silva
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Lucimara M C Cordeiro
- Department of Biochemistry and Molecular Biology, Federal University of Parana, Curitiba, PR, Brazil
| | - Marcelo B Góis
- Post-Graduation Program in Biosciences and Health, Federal University of Rondonópolis, Rondonópolis, Brazil
| | - Jand V R Medeiros
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil; Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil.
| |
Collapse
|
18
|
Hjørne AP, Mortensen MS, Licht TR, Laursen MF. Loperamide increases mouse gut transit time in a dose-dependent manner with treatment duration-dependent effects on distinct gut microbial taxa. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2025; 6:e7. [PMID: 40336798 PMCID: PMC12056420 DOI: 10.1017/gmb.2025.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/09/2025]
Abstract
Intestinal transit time has been recognized as an important factor in shaping the gut microbiota, although causality remains to be firmly demonstrated. The aim of this study was to evaluate the effect of different loperamide doses on the mouse intestinal transit time and to investigate the effects of increasing transit time on the gut microbial community. Loperamide significantly increased the transit time in a dose-dependent manner. Additionally, we observed a significant difference between the control group and the loperamide-treated groups in the abundance of the bacterial families Bacteroidaceae, Erysipelotrichaceae, Porphyromonadaceae, and Akkermansiaceae after 7 days of loperamide treatment, with the bacterial families responding to the increased transit time at different rates. Fermentation of faeces obtained from the same mice, with or without loperamide, demonstrated that the observed effects on gut microbiota in vivo were not a result of direct interactions between loperamide and the gut microbiota but rather a consequence of loperamide-induced increased intestinal transit time. In the cecum of the mice, we found higher levels of propionate in the high-dose group compared to the control and low-dose groups. Collectively, our findings establish that an altered transit time is causal to changes in the composition and activity of the microbiome.
Collapse
Affiliation(s)
- Anna Pii Hjørne
- National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | - Tine Rask Licht
- National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | | |
Collapse
|
19
|
Zhu Y, Fang Z, Bai J, Wang L, Chen J, Zhang Z, Wang Q, Sheng W, Pan X, Gao Z, Xu D, Wu P, Sun B. Orally Administered Functional Polyphenol-Nanozyme-Armored Probiotics for Enhanced Amelioration of Intestinal Inflammation and Microbiota Dysbiosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411939. [PMID: 40067175 PMCID: PMC12061243 DOI: 10.1002/advs.202411939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/23/2025] [Indexed: 05/04/2025]
Abstract
Maintaining microbiota balance and enhancing the antioxidant performance of nanozyme-based probiotic systems are crucial for effective inflammatory bowel disease (IBD) therapy. Despite significant advancements, developing a green and safe coating technology that functionalizes probiotics with nanozymes while preserving the activity of both components remains a challenge. To address this, chitosan-modified epigallocatechin gallate (EGCG-CS, EC)is synthesized, leveraging the intrinsic adhesive and coordination properties of polyphenols to capture gold nanozymes (AuNPs), forming ECA complexes that enhance nanozyme activity. When coated onto Escherichia coli Nissle 1917 (EcN), the resulting ECA@EcN system effectively scavenged reactive oxygen species (ROS), improving probiotic viability and promoting colon accumulation. Mechanistically, ECA protected EcN by suppressing the activation of the Flagellar Assembly and Branched-Chain Amino Acid Synthesis pathways, ultimately alleviating inflammation and modulating intestinal microbial communities to relieve IBD symptoms. Given the biocompatibility of its components and the environmentally friendly assembly approach, this polyphenol-nanozyme-armored probiotic system represents a promising platform for IBD treatment.
Collapse
Affiliation(s)
- Yong Zhu
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Ziqu Fang
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Jie Bai
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Longhui Wang
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Jiaqing Chen
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Zehua Zhang
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Qiang Wang
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Weiwei Sheng
- Department of General SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Xueyin Pan
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Zhenyuan Gao
- Department of General SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Dengqiu Xu
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Pengkai Wu
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Beicheng Sun
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| |
Collapse
|
20
|
Ofori‐Kwafo A, Sigdel I, Al Mamun E, Zubcevic J, Tang Y. Gut-on-a-chip platforms: Bridging in vitro and in vivo models for advanced gastrointestinal research. Physiol Rep 2025; 13:e70356. [PMID: 40323242 PMCID: PMC12051376 DOI: 10.14814/phy2.70356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/11/2025] [Accepted: 04/22/2025] [Indexed: 05/08/2025] Open
Abstract
The gastrointestinal (GI) tract plays a critical role in nutrient absorption, immune responses, and overall health. Traditional models such as two-dimensional cell cultures have provided valuable insights but fail to replicate the dynamic and complex microenvironment of the human gut. Gut-on-a-chip platforms, which incorporate cells located in the gut into microfluidic devices that simulate peristaltic motion and fluid flow, represent a significant advancement in modeling GI physiology and diseases. This review discusses the evolution of gut-on-a-chip technology, from simple cellular mono-cultures models to more sophisticated systems incorporating bi-cultures and tri-cultures that enable studies of drug metabolism, disease modeling, and gut-microbiome interactions. Although challenges remain, including maintaining long-term cell viability and replicating immune responses, these platforms hold great potential for advancing personalized medicine and improving drug discovery efforts targeting gastrointestinal disorders.
Collapse
Affiliation(s)
- Awurama Ofori‐Kwafo
- Department of Bioengineering, College of EngineeringUniversity of ToledoToledoOhioUSA
| | - Indira Sigdel
- Department of Bioengineering, College of EngineeringUniversity of ToledoToledoOhioUSA
| | - Earshed Al Mamun
- Department of Bioengineering, College of EngineeringUniversity of ToledoToledoOhioUSA
| | - Jasenka Zubcevic
- University of South Florida Center for Microbiome ResearchMicrobiomes InstituteTampaFloridaUSA
- Department of Neurosurgery and Brain RepairUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Yuan Tang
- Department of Bioengineering, College of EngineeringUniversity of ToledoToledoOhioUSA
| |
Collapse
|
21
|
Zhong H, Jiang J, Hussain M, Zhang H, Chen L, Guan R. The Encapsulation Strategies for Targeted Delivery of Probiotics in Preventing and Treating Colorectal Cancer: A Review. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2500304. [PMID: 40192333 PMCID: PMC12079478 DOI: 10.1002/advs.202500304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/01/2025] [Indexed: 05/16/2025]
Abstract
Colorectal cancer (CRC) ranks as the third most prevalent cancer worldwide. It is associated with imbalanced gut microbiota. Probiotics can help restore this balance, potentially reducing the risk of CRC. However, the hostile environment and constant changes in the gastrointestinal tract pose significant challenges to the efficient delivery of probiotics to the colon. Traditional delivery methods are often insufficient due to their low viability and lack of targeting. To address these challenges, researchers are increasingly focusing on innovative encapsulation technologies. One such approach is single-cell encapsulation, which involves applying nanocoatings to individual probiotic cells. This technique can improve their resistance to the harsh gastrointestinal environment, enhance mucosal adhesion, and facilitate targeted release, thereby increasing the effectiveness of probiotic delivery. This article reviews the latest developments in probiotic encapsulation methods for targeted CRC treatment, emphasizing the potential benefits of emerging single-cell encapsulation techniques. It also analyzes and compares the advantages and disadvantages of current encapsulation technologies. Furthermore, it elucidates the underlying mechanisms through which probiotics can prevent and treat CRC, evaluates the efficacy and safety of probiotics in CRC treatment and adjuvant therapy, and discusses future directions and potential challenges in the targeted delivery of probiotics for CRC treatment and prevention.
Collapse
Affiliation(s)
- Hao Zhong
- College of Food Science and TechnologyZhejiang University of TechnologyHangzhou310014China
| | - Jin Jiang
- College of Food Science and TechnologyZhejiang University of TechnologyHangzhou310014China
| | - Muhammad Hussain
- College of Food Science and TechnologyZhejiang University of TechnologyHangzhou310014China
- Moganshan Institute ZJUTKangqianDeqing313200China
| | - Haoxuan Zhang
- College of Food Science and TechnologyZhejiang University of TechnologyHangzhou310014China
| | - Ling Chen
- Sanya Branch of Hainan Academy of Inspection and TestingSan Ya572011China
| | - Rongfa Guan
- College of Food Science and TechnologyZhejiang University of TechnologyHangzhou310014China
- Moganshan Institute ZJUTKangqianDeqing313200China
| |
Collapse
|
22
|
Yang N, Jike X, Zhang M, Jiang T, Lei H. Synthesis, characterization of thiolated hyaluronic acid and evaluation of its encapsulation effects on Limosilactobacillus reuteri HR7. Int J Biol Macromol 2025; 310:143486. [PMID: 40280531 DOI: 10.1016/j.ijbiomac.2025.143486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/14/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Hyaluronic acid (HA) was thiol-modified by adding different concentrations of L-Cysteine (1 mmol, 2 mmol, 3 mmol, 4 mmol, 5 mmol and 6 mmol) and the resulting polymers (HA-SH) were characterized. The results of FTIR, 1H NMR, XRD, SEM and DSC all confirmed the success of thiol modification, accompanied by free thiol group content of 3169.51-3913.44 μmol/g. Sulfur element was only detected in HA-SH, accounting for 1.12 %-1.23 %. The Mw and particle size were decreased after thiol modification, representing a more uniform polysaccharide conformation, which was beneficial for the encapsulation of probiotics. Rheological analysis showed that the hydrogel prepared by HA displayed viscoelastic fluid properties, while the hydrogels prepared by HA-SH exhibited solid-like gel properties, indicating enhanced gelation properties after thiol modification. Subsequently, the hydrogels were applied to probiotics encapsulation to explore the effects on gastrointestinal tolerance. A higher encapsulation efficiency was observed in HA-SH hydrogel with enhanced gastrointestinal tolerance, increasing by 38.15 % on average. These results demonstrated that thiolation was a good strategy for polysaccharide modification and hydrogel formed by HA-SH was a more promising encapsulation and delivery system for probiotics compared with HA.
Collapse
Affiliation(s)
- Nana Yang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| | - Xiaolan Jike
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| | - Mengmeng Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| | - Tian Jiang
- Shanghai Helplifes Technology Co., Ltd, Shanghai 201702, China.
| | - Hongjie Lei
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
23
|
Yilmaz EG, Hacıosmanoğlu N, Jordi SBU, Yilmaz B, Inci F. Revolutionizing IBD research with on-chip models of disease modeling and drug screening. Trends Biotechnol 2025; 43:1062-1078. [PMID: 39523166 DOI: 10.1016/j.tibtech.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/30/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024]
Abstract
Inflammatory bowel disease (IBD) comprises chronic inflammatory conditions with complex mechanisms and diverse manifestations, posing significant clinical challenges. Traditional animal models and ethical concerns in human studies necessitate innovative approaches. This review provides an overview of human intestinal architecture in health and inflammation, emphasizing the role of microfluidics and on-chip technologies in IBD research. These technologies allow precise manipulation of cellular and microbial interactions in a physiologically relevant context, simulating the intestinal ecosystem microscopically. By integrating cellular components and replicating 3D tissue architecture, they offer promising models for studying host-microbe interactions, wound healing, and therapeutic approaches. Continuous refinement of these technologies promises to advance IBD understanding and therapy development, inspiring further innovation and cross-disciplinary collaboration.
Collapse
Affiliation(s)
- Eylul Gulsen Yilmaz
- UNAM-National Nanotechnology Research Center, Bilkent University, 06800 Ankara, Turkey; Institute of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey
| | - Nedim Hacıosmanoğlu
- UNAM-National Nanotechnology Research Center, Bilkent University, 06800 Ankara, Turkey; Institute of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey
| | - Sebastian Bruno Ulrich Jordi
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, 3010, Bern, Switzerland; Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, 3008, Bern, Switzerland
| | - Bahtiyar Yilmaz
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, 3010, Bern, Switzerland; Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, 3008, Bern, Switzerland.
| | - Fatih Inci
- UNAM-National Nanotechnology Research Center, Bilkent University, 06800 Ankara, Turkey; Institute of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey.
| |
Collapse
|
24
|
Zhu D, Ma X, Wang J, Chen T, Yang J, Liu Y, Lin Z, Wu M, Hu TY, Zhang Y. A Sequential Release Micro-nano System for Colitis Therapy via Gut Microbiota and Immune Regulation. Angew Chem Int Ed Engl 2025; 64:e202424409. [PMID: 39980315 DOI: 10.1002/anie.202424409] [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: 12/31/2024] [Revised: 02/19/2025] [Accepted: 02/19/2025] [Indexed: 02/22/2025]
Abstract
Commencing with the breakdown of the intestinal barrier, various pathogenic factors, such as dysbiosis of the gut microbiota, harmful inflammatory cytokines, and immune system imbalance, collectively contribute to the development of colitis. Numerous interventions focusing on single factors have been developed to provide short-term therapeutic benefits, but the continued existence of unresolved pathogenic factors can lead to disease exacerbation. Here we have designed a multicomponent system-inulin microspheres encapsulating selenium-containing nanomicelles, aiming to tackle the multiple factors associated with colitis. This micro-nano drug delivery platform achieves sequential release of drugs in the inflamed colon, with each component of the system functioning independently and jointly. The outer layer of inulin supplies nutrients for probiotics. The inner core comprises selenocystamine and 3-oxolithocholic acid, which polarize macrophages towards an anti-inflammatory phenotype and regulate adaptive immunity by inhibiting TH17-cell differentiation, respectively. In an acute colitis mouse model, this therapeutic system ameliorates colonic inflammation, enhances the abundance of gut microbiota, and modulates the mucosal immune system, showing potential in preventing colitis.
Collapse
Affiliation(s)
- Dongdong Zhu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xiaocao Ma
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Jingguo Wang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Tiantian Chen
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Jiahui Yang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yan Liu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Zhun Lin
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Minhao Wu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Tony Y Hu
- Center of Cellular and Molecular Diagnosis, Tulane University School of Medicine, New Orleans, Louisiana, 70112, United States
| | - Yuanqing Zhang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| |
Collapse
|
25
|
Ni P, Li L, Du K, Nov P, Wang D, Wang C, Kou Q, Li Y, Zhang Y, Zheng C, Fu W, Li J. Unveiling the immunological terrain of pancreatic ductal adenocarcinoma: strategies to prompt immunotherapy from Mendelian randomization. Discov Oncol 2025; 16:613. [PMID: 40279021 PMCID: PMC12031697 DOI: 10.1007/s12672-025-02250-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is challenging to treat due to its immunosuppressive tumor microenvironment (TME) and resistance to immune checkpoint inhibitors. This study aims to discover new therapeutic targets and predictive biomarkers for PDAC. METHODS Using Mendelian randomization, we studied causal relationships between PDAC and an array of immune cell traits, bacterial traits, inflammatory factors, and blood metabolites. We employed large genome-wide association study datasets and the two-sample MR approach for the investigation. RESULTS Our results highlight suggestive evidence of associations between PDAC and distinct immune cell phenotypes, revealing nuanced alterations across monocytes, T-cells, B-cells, dendritic cells, and myeloid-derived suppressor cells. Our study provides a granular view of the PDAC-immune interface, identifying key immune cell traits and their associations with PDAC. For instance, our findings suggest a detrimental reduction in various monocyte traits, alongside a decrease in B-cell populations. Conversely, certain T-cell subsets showed increased associations, indicating potential targets for immunotherapeutic strategies. The bacterial trait associations, particularly with Collinsella and Ruminococcus torques, highlight the gut microbiome's influence on immune modulation and PDAC pathogenesis. Additionally, the traits concerning Interleukin-12 subunit beta levels and T-cell surface glycoprotein CD5 levels further indicate their function of this complex interaction. CONCLUSIONS This study enhances our understanding of PDAC's resistance to immunotherapies and highlights the potential of personalized immunotherapy and metabolic pathway modulation in PDAC treatment. Our findings provide supportive evidence for research and clinical translation.
Collapse
Affiliation(s)
- Peizan Ni
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Guangzhou, 510280, Guangdong, China
| | - Lilin Li
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Guangzhou, 510280, Guangdong, China
| | - KunPeng Du
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Guangzhou, 510280, Guangdong, China
| | - Pengkhun Nov
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Guangzhou, 510280, Guangdong, China
| | - Duanyu Wang
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Guangzhou, 510280, Guangdong, China
| | - Changqian Wang
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Guangzhou, 510280, Guangdong, China
| | - Qianzi Kou
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Guangzhou, 510280, Guangdong, China
| | - Ying Li
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Guangzhou, 510280, Guangdong, China
| | - Yangfeng Zhang
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Guangzhou, 510280, Guangdong, China
| | - Chongyang Zheng
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Guangzhou, 510280, Guangdong, China
| | - Wen Fu
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Guangzhou, 510280, Guangdong, China
| | - Jiqiang Li
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, No. 253, Industrial Avenue, Guangzhou, 510280, Guangdong, China.
| |
Collapse
|
26
|
Moțățăianu A, Mănescu IB, Șerban G, Ion V, Bălașa R, Andone S. The Effects of a Mediterranean Diet on Metabolic Hormones and Cytokines in Amyotrophic Lateral Sclerosis Patients: A Prospective Interventional Study. Nutrients 2025; 17:1437. [PMID: 40362746 PMCID: PMC12073196 DOI: 10.3390/nu17091437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Revised: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Amyotrophic lateral sclerosis (ALS) is a prevalent neurodegenerative disease but lacks effective treatments. Dietary interventions, notably the Mediterranean diet, promise to modulate disease pathways. This study aimed to investigate the impact of the Mediterranean diet on gut hormones and cytokines in patients with amyotrophic lateral sclerosis (ALS). Methods: We conducted a 12-month, single-center prospective study on a total of 44 ALS patients. After a 6-month observation period, the patients were placed on a dairy-free Mediterranean diet for the next 6 months. We evaluated the patients at baseline (T0), 6 months (T1), and 12 months (T2). We measured the ALS Functional Rating Scale-Revised (ALSFRS-R) scores and a panel of metabolic hormones and cytokines. Results: The ALSFRS-R scores declined over 12 months (37.59 ± 6.32 at T0 vs. 30.23 ± 8.91 at T2, p < 0.001), indicating expected disease progression with no significant difference in the rate of decline before and after the dietary intervention. The leptin levels significantly decreased from T0 to T1 (T0: 4956 ± 3994 pg/mL vs. T1: 3196 ± 2807 pg/mL, p = 0.038). The insulin and GLP-1 levels showed significant drops at T2 (insulin T0: 480 ± 369 vs. T2: 214 ± 213 pmol/L, p < 0.01; GLP-1 T0: 118 ± 76 vs. T2: 60 ± 57 pg/mL, p < 0.01). C-peptide increased at T2 (T0: 3814 ± 1967 vs. T2: 9532 ± 4000 pg/mL, p < 0.001). Among the cytokines, the levels of IL-12P70, IL-13, IL-9, and IL-2 significantly decreased from T0 to T2 (all p < 0.05), while IL-17A and TNFα significantly increased between T1 and T2 (p < 0.01). Conclusions: The Mediterranean diet intervention in ALS patients modulated several metabolic hormones and cytokines but with no evidence of impacting the disease's evolution or of a slowed clinical progression. These findings suggest a potential role for dietary intervention, particularly the Mediterranean diet, in modulating gut hormones and cytokines in ALS patients, but its impact on disease course is unclear. Future randomized studies are needed to confirm these changes and to determine whether dietary intervention can have any benefit in ALS.
Collapse
Affiliation(s)
- Anca Moțățăianu
- Department of Neurology, University of Medicine, Pharmacy, Science and Technology of Târgu Mureș ‘George Emil Palade’, 540142 Târgu Mureș, Romania
- 1st Neurology Clinic, Mures County Clinical Emergency Hospital, 540136 Târgu Mureș, Romania
| | - Ion Bogdan Mănescu
- Department of Laboratory Medicine, University of Medicine, Pharmacy, Science and Technology of Târgu Mureș ‘George Emil Palade’, 540142 Târgu Mureș, Romania
| | - Georgiana Șerban
- Doctoral School, University of Medicine, Pharmacy, Science and Technology of Târgu Mureș ‘George Emil Palade’, 540142 Târgu Mureș, Romania
| | - Valentin Ion
- Faculty of Pharmacy, Department of Analytical Chemistry and Drug Analysis, University of Medicine, Pharmacy, Science and Technology of Târgu Mureș ‘George Emil Palade’, 540142 Târgu Mureș, Romania
- Drug Testing Laboratory, University of Medicine, Pharmacy, Science and Technology of Târgu Mureș ‘George Emil Palade’, 540142 Târgu Mureș, Romania
| | - Rodica Bălașa
- Department of Neurology, University of Medicine, Pharmacy, Science and Technology of Târgu Mureș ‘George Emil Palade’, 540142 Târgu Mureș, Romania
- 1st Neurology Clinic, Mures County Clinical Emergency Hospital, 540136 Târgu Mureș, Romania
| | - Sebastian Andone
- Department of Neurology, University of Medicine, Pharmacy, Science and Technology of Târgu Mureș ‘George Emil Palade’, 540142 Târgu Mureș, Romania
- 1st Neurology Clinic, Mures County Clinical Emergency Hospital, 540136 Târgu Mureș, Romania
| |
Collapse
|
27
|
Zhao Z, Xu Y, Hu Y. Acid-resistant chemotactic DNA micromotors for probiotic delivery in inflammatory bowel disease. Nat Commun 2025; 16:3778. [PMID: 40263286 PMCID: PMC12015548 DOI: 10.1038/s41467-025-59172-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 04/13/2025] [Indexed: 04/24/2025] Open
Abstract
Microcapsules composed of synthetic polymeric matrices have attracted considerable attention in delivering oral probiotics. However, existing polymeric microcapsules demonstrate inadequate acid resistance and adaptability, as well as deficiency in the inflamed colon-specificity and uncontrolled release of probiotics therein. Herein, a DNA microcapsule is prepared as a probiotic-transporting micromotor through photo-crosslinking of hyaluronic acid methacrylate and acrydite-modified A-/C-rich oligomers within the microfludically generated droplets in the presence of nitric oxide-cleavable crosslinker and gas donor manganese carbonyl (MnCO). As the microcapsules traverse stomach, duodenum, and ultimately colon, the formation and dissociation of A-motif and i-motif structures instigate a reversible shrinking-swelling transition of microcapsules to preserve probiotic viability. Subsequently, the microcapsules exhibit chemotaxis towards inflamed colon site, driven by a gas-generating reaction between MnCO and elevated reactive oxygen species. Following disintegration of the microcapsules, triggered by endogenous nitric oxide, probiotics are released to reshape the dysbiosis of intestinal microflora. This advanced delivery system offers significant promise for the effective clinical management of inflammatory bowel disease.
Collapse
Affiliation(s)
- Zinan Zhao
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, 201804, P. R. China
| | - Yao Xu
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, 201804, P. R. China
| | - Yong Hu
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, Shanghai, 201804, P. R. China.
| |
Collapse
|
28
|
Garg G, Trisal A, Singh AK. Unlocking the therapeutic potential of gut microbiota for preventing and treating aging-related neurological disorders. Neuroscience 2025; 572:190-203. [PMID: 40073931 DOI: 10.1016/j.neuroscience.2025.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 03/03/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
Billions of microorganisms inhabit the human gut and maintain overall health. Recent research has revealed the intricate interaction between the brain and gut microbiota through the microbiota-gut-brain axis (MGBA) and its effect on neurodegenerative disorders (NDDs). Alterations in the gut microbiota, known as gut dysbiosis, are linked to the development and progression of several NDDs. Studies suggest that the gut microbiota may be a viable target for improving cognitive health and reducing hallmarks of brain aging. Numerous pathways including hypothalamic-pituitary-adrenal axis stimulation, neurotransmitter release disruption, system-wide inflammation, and increased intestinal and blood-brain barrier permeability connect gut dysbiosis to neurological conditions. Metabolites produced by the gut microbiota influence neural processes that affect brain function. Clinical interventions depend on the capacity to understand the equilibrium between beneficial and detrimental gut microbiota, as it affects both neurodegeneration and neuroprotection. The importance of the gut microbiota and its metabolites during brain aging and the development of neurological disorders is summarized in this review. Moreover, we explored the possible therapeutic effects of the gut microbiota on age-related NDDs. Highlighting various pathways that connect the gut and the brain, this review identifies several important domains where gut microbiota-based interventions could offer possible solutions for age-related NDDs. Furthermore, prebiotics and probiotics are discussed as effective alternatives for mitigating indirect causes of gut dysbiosis. These therapeutic interventions are poised to play a significant role in improving dysbiosis and NDDs, paving the way for further research.
Collapse
Affiliation(s)
- Geetika Garg
- Department of Zoology, Savitribai Phule Pune University, Pune 411007, India
| | - Anchal Trisal
- Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Abhishek Kumar Singh
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal 576 104, India.
| |
Collapse
|
29
|
Li P, Zhu L, Song C, Wu M, Zhu X, He S, Hu B, Chen Z, Liu Z, Luo B, Liu Y, Yang X, Hu J. Triple-Functional Probiotics with Intracellularly Synthesized Selenium Nanoparticles for Colitis Therapy by Regulating the Macrophage Phenotype and Modulating Gut Microbiota. ACS NANO 2025; 19:14213-14232. [PMID: 40192063 DOI: 10.1021/acsnano.5c00574] [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: 04/16/2025]
Abstract
The dysregulated macrophage phenotype, as the main cause of colitis, not only enhanced oxidative stress to exacerbate inflammatory responses but was closely related with gut microbial dysbiosis. It was needed to simultaneously address the three issues for the effective treatment of colitis, but it was not satisfied. Here, we developed "three-birds-one-stone" probiotics, named Se@EcN-C2/A2, for colitis treatment. Escherichia coli Nissle 1917 (EcN), a clinically approved probiotic, was used to intracellularly synthesize selenium (Se) nanoparticles by biomineralization, giving Se@EcN. Coating glycol chitosan and sodium alginate on the surface of Se@EcN (Se@EcN-C2/A2) endowed probiotics with high resistance to the harsh gastrointestinal tract environment and strong adhesion and targeting ability to the inflamed site of the colon to facilitate the uptake by M1 macrophages. Se@EcN-C2/A2 was metabolized to SeCys2 and MetSeCys to be involved in the synthesis of GPX2 and TXNRD1, which led to reaction oxygen species clearance to inhibit Toll-like receptor and nuclear factor κB signaling pathways to suppress inflammatory response and polarize M1 macrophages to M2 phenotypes by activating PI3K/AKT signaling pathways. In DSS-induced colitis mice, Se@EcN-C2/A2 exerted satisfactory therapeutic and prophylactic effects, including scavenging oxidative stress and regulating macrophage phenotypes to suppress inflammatory response and restore gut barrier functions. Moreover, the living probiotic EcN in the colon effectively regulated microbial dysbiosis by decreasing the abundance of Escherichia-Shigella and increasing the abundance of Lactobacillus and Bifidobacterium.
Collapse
Affiliation(s)
- Puze Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lichong Zhu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Cheng Song
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Meichan Wu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xuan Zhu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Suting He
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bo Hu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zehao Chen
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhi Liu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ban Luo
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yan Liu
- Department of Gastroenterology, Wuhan Eighth Hospital, Wuhan 430010, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jun Hu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Jiangxia Laboratory, Wuhan 430200, China
| |
Collapse
|
30
|
Liu X, Mao B, Tang X, Zhang Q, Zhao J, Chen W, Cui S. Bacterial viability retention in probiotic foods: a review. Crit Rev Food Sci Nutr 2025:1-23. [PMID: 40215221 DOI: 10.1080/10408398.2025.2488228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Probiotics offer substantial health benefits, leading to their increased consumption in various food products. The viability of probiotics is a critical factor that influences the nutritional and therapeutic efficacy of these foods. However, as probiotics often lose viability during production and oral administration, effective preservation and encapsulation technologies are needed to overcome this challenge. This review elucidates the diverse sources and incorporation strategies of probiotics, while systematically analyzing the effects of water transformation (ice front velocity, glass transition temperature, and collapse temperature), processing conditions (food matrix, temperature, and dissolved oxygen), and gastrointestinal challenges (gastric fluid, digestive enzymes, and bile salts) on probiotic viability. Effective strategies to strengthen probiotic viability encompass three primary domains: fermentation processes, production techniques, and encapsulation methods. Specifically, these include meticulous fermentation control (nitrogen sources, lipids, and carbon sources), pre-stress treatments (pre-cooling, heat shock, NaCl stress, and acid stress), optimized lyoprotectant selection (carbohydrates, proteins, and polyols), synergistic freeze-drying technologies (infrared technology, spray drying, and microwave), bulk encapsulation approaches (polysaccharide or protein-based microencapsulation), and single-cell encapsulation methods (self-assembly and surface functionalization). Despite these advancements, targeting specific probiotics and food matrices remains challenging, necessitating further research to enhance probiotic viability.
Collapse
Affiliation(s)
- Xuewu Liu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Bingyong Mao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xin Tang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Qiuxiang Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Research Laboratory for Maternal-Infant Microbiota and Health, Jiangnan University, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Shumao Cui
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- International Joint Research Laboratory for Maternal-Infant Microbiota and Health, Jiangnan University, Wuxi, China
| |
Collapse
|
31
|
Li H, Chen J, Chen Z, Liao J. Predicting immune status and gene mutations in stomach adenocarcinoma patients based on inflammatory response-related prognostic features. Discov Oncol 2025; 16:497. [PMID: 40205166 PMCID: PMC11982005 DOI: 10.1007/s12672-025-02210-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Stomach adenocarcinoma (STAD) is an aggressive malignant tumor. Herein, we characterized the prognosis based on inflammatory response-related features and evaluated their potential impact on survival and immune status of STAD patients. METHODS Inflammation-related genes obtained from public databases were used to analyze the inflammatory response scores of STAD samples. The differentially expressed genes (DEGs) between STAD and adjacent gastric tissue were then analyzed using the "limma" package. Genes associated with STAD prognosis were obtained from the intersection of inflammation-related genes and DEGs. The key genes screened by last absolute shrinkage and selection operator (LASSO) Cox and stepwise regression analyses were used to construct prognostic models and nomograms. The tumor immune dysfunction exclusion (TIDE) algorithm was used to assess potential differences in immunotherapy response between high- and low-risk groups and to explore gene mutation signatures using the R software maftools package. In addition, GSEA was used to predict pathway characteristics between different subgroups. Finally, scratch and transwell assays were performed to explore the role of SERPINE1 in STAD cells. RESULTS We found that a high-inflammatory group was associated with poor prognosis in STAD patients. 14 inflammation-related DEGs out of 126 DEGs were identified to be associated with the prognosis of STAD patients, and the prognostic models and nomograms constructed from the subsequently identified key genes (SLC7A1, CD82, SERPINE1 ROS1 and SLC7A2) demonstrated a good predictive performance in terms of prognosis of STAD. Patients in the STAD high-risk group had higher StromalScore and TIDE scores. It was also found that patients in the STAD low-risk group may have a higher mutation burden. Enrichment analysis revealed significant enrichment of epithelial-mesenchymal transition, angiogenesis and KRAS pathways in the high-risk group. In-vitro experiments showed that down-regulation of SERPINE1 attenuated the migratory and invasive abilities of AGS cells. CONCLUSION This study provides new insights into prognostic prediction and immunotherapy for STAD from the perspective of the inflammatory response.
Collapse
Affiliation(s)
- Huanjun Li
- Medical Oncology, Dongguan Institute of Clinical Cancer Research, Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan People's Hospital, Dongguan, 523888, China
| | - Jingtang Chen
- Medical Oncology, Dongguan Institute of Clinical Cancer Research, Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan People's Hospital, Dongguan, 523888, China
| | - Zhiliang Chen
- General Surgery Department, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan People's Hospital, Dongguan, 523888, China
| | - Jingsheng Liao
- Medical Oncology, Dongguan Institute of Clinical Cancer Research, Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Dongguan People's Hospital, Dongguan, 523888, China.
| |
Collapse
|
32
|
Cao D, Xiang J, Chen X, Liu P, Zhao P, Wang J, Li R, Li K, Chen S, Liu G. Biodegradable Cellulose Acetate Nanofibrous Membranes with Self-Sustaining Electrostatic Effect for Efficient and Stable Air Purification. ACS APPLIED MATERIALS & INTERFACES 2025; 17:22025-22034. [PMID: 40155205 DOI: 10.1021/acsami.5c00794] [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: 04/01/2025]
Abstract
Amid the global pursuit of carbon neutrality and the pressing challenge of severe air pollution, degradable cellulose acetate (CA) materials hold great potential in the field of air filtration. However, their weak polarity and poor antibacterial properties limit their widespread application in this field. Herein, we developed CA-based nanofibrous membranes (CAPZ NFMs) with antimicrobial properties, which achieved efficient and stable filtration performance through a self-sustaining electrostatic effect driven by polarity. CAPZ NFMs were fabricated by electrospinning a solution that contained CA, highly polar zwitterionic copolymers (PSG), and biocompatible Zr4+. The zwitterionic groups of PSG increased the polarity of CAPZ NFMs to 19.62 mN·m-1, significantly surpassing that of pristine CA NFMs (2.94 mN·m-1). This enhancement granted CAPZ NFMs a surface potential of 2.07 kV, which enabled a PM0.3 filtration efficiency of 99.56% while maintaining a low pressure drop of 79 Pa. Notably, CAPZ NFMs maintained superior performance under high humidity conditions and 6 months of outdoor storage. Additionally, Zr4+ coordinated with the zwitterionic groups of PSG to form quaternary ammonium groups, endowing CAPZ NFMs with broad-spectrum antibacterial efficacy of over 99.99%. This work could provide new strategies for developing next-generation biodegradable, high-electrostatic filtration materials.
Collapse
Affiliation(s)
- Dan Cao
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, PR China
- National Engineering Research Center of Clean Technology in Leather Industry, Sichuan University, Chengdu 610065, PR China
| | - Jun Xiang
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, PR China
- National Engineering Research Center of Clean Technology in Leather Industry, Sichuan University, Chengdu 610065, PR China
| | - Xi Chen
- The Collaborative Innovation Center for Eco-Friendly and Fire-Safety Polymeric Materials (MoE), National Engineering Laboratory of Eco-Friendly Polymeric Materials (Sichuan), State Key Laboratory of Polymer Materials Engineering, College of Chemistry, Sichuan University, Chengdu 610065, PR China
| | - Ping Liu
- College of Graduate and Professional Studies, Trine University, Reston, Virginia 20191, United States
| | - Peng Zhao
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, PR China
- National Engineering Research Center of Clean Technology in Leather Industry, Sichuan University, Chengdu 610065, PR China
| | - Junchao Wang
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, PR China
- National Engineering Research Center of Clean Technology in Leather Industry, Sichuan University, Chengdu 610065, PR China
| | - Rui Li
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, PR China
- National Engineering Research Center of Clean Technology in Leather Industry, Sichuan University, Chengdu 610065, PR China
| | - Kaijun Li
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, PR China
- National Engineering Research Center of Clean Technology in Leather Industry, Sichuan University, Chengdu 610065, PR China
| | - Sheng Chen
- National Engineering Research Center of Clean Technology in Leather Industry, Sichuan University, Chengdu 610065, PR China
| | - Gongyan Liu
- College of Biomass Science and Engineering, Sichuan University, Chengdu 610065, PR China
- National Engineering Research Center of Clean Technology in Leather Industry, Sichuan University, Chengdu 610065, PR China
| |
Collapse
|
33
|
Xing H, Liu X, Wang J, Zhou T, Jin X, Qiu R, Lu Y, Liu C, Song Y. Magnetically targeted delivery of probiotics for controlled residence and accumulation in the intestine. NANOSCALE 2025; 17:8588-8598. [PMID: 40072455 DOI: 10.1039/d4nr04753b] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
The effectiveness of orally delivered probiotics in treating gastrointestinal diseases is restricted by inadequate gut retention. In this study, we present a magnetically controlled strategy for probiotic delivery, which enables controlled accumulation and residence of probiotics in the intestine. The magnetically controlled probiotic is established by attaching amino-modified iron oxide (Fe3O4-NH3+ NPs) to polydopamine-coated Lacticaseibacillus rhamnosus GG (LGG@P) through electrostatic self-assembly and named as LGG@P@Fe3O4. In a simulated gastrointestinal environment, LGG@P@Fe3O4 maintains both structural stability and probiotic viability. Furthermore, the LGG@P@Fe3O4 clusters can be easily manipulated by an external magnetic field, inducing directional movement and aggregation. In vitro simulations demonstrated significant accumulation and retention of LGG@P@Fe3O4 under a magnetic field, with the optical density (OD) value of the suspension decreasing from ∼1.17 to ∼0.29. In contrast, the OD value of the suspension without a magnetic field remained at its original level (∼1.15). In a mouse model with intragastrically administered LGG@P@Fe3O4, the group exposed to a magnet exhibited stronger gut fluorescence after 24 h. The magnetically controlled probiotic delivery strategy offers an easy manufacturing and feasible method to enhance the effectiveness of probiotics in treating gastrointestinal diseases.
Collapse
Affiliation(s)
- Hanye Xing
- Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China.
| | - Xingyu Liu
- Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China.
| | - Ju Wang
- Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China.
| | - Tao Zhou
- Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China.
| | - Xiangxiang Jin
- Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China.
| | - Rui Qiu
- Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China.
| | - Yang Lu
- Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China.
| | - Changhong Liu
- Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China.
| | - Yonghong Song
- Anhui Province Key Laboratory of Advanced Catalytic Materials and Reaction Engineering, School of Chemistry and Chemical Engineering, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China.
| |
Collapse
|
34
|
Liu A, Ma J, Liu Z, Qiu T, Zhao Q, Li G, Liang X, Li Q. "Shield" Armed Programmable Probiotics Harboring α-Aminoadipate Aminotransferase Gene Regulate Tryptophan Metabolism and Gut Microbiota to Alleviate the Inflammatory Bowel Disease. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:7920-7932. [PMID: 40116595 DOI: 10.1021/acs.jafc.4c13017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
Current treatment of inflammatory bowel disease (IBD) relies on anti-inflammatory and immunosuppressive agents. However, this concept is considered outdated due to its restricted efficacy and unavoidable side effects. Herein, a polynorepinephrine-coated programmable probiotic expressing α-aminoadipate aminotransferase (NE-EcN-pA) was constructed to improve the levels of kynurenic acid and xanthurenic acid in the intestine by modulating the endogenous tryptophan metabolism. The NE layer could protect EcN-pA against the harsh environment of the gastrointestinal tract, enhancing its survival and colonization. In UC mice, oral administration of NE-EcN-pA effectively alleviated intestinal inflammation and restored the intestinal epithelial barrier owing to the activation of the aryl hydrocarbon receptor pathway. Furthermore, NE-EcN-pA promoted the diversity of intestinal flora, improved the imbalance of flora, and enhanced the content of short-chain fatty acids in the colon. Overall, NE-EcN-pA can regulate endogenous tryptophan metabolism and gut microbiota, showing promise in the treatment of gastrointestinal disorders.
Collapse
Affiliation(s)
- Aijiang Liu
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jun Ma
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Zengguang Liu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Tianyuan Qiu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Qixuan Zhao
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Guangquan Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130012, China
| | - Xiao Liang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Quanshun Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| |
Collapse
|
35
|
Kim TT, Malu D, He D, Hu Y, Kim J. Development of Bioorthogonally Degradable Tough Hydrogels Using Enamine N-Oxide Based Crosslinkers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414692. [PMID: 40018818 PMCID: PMC11962699 DOI: 10.1002/adma.202414692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/20/2025] [Indexed: 03/01/2025]
Abstract
Inducibly degradable polymers present new opportunities to integrate tough hydrogels into a wide range of biomaterials. Rapid and inducible degradation enables fast transition in material properties without sacrificing material integrity prior to removal. In pursuit of bioorthogonal chemical modalities that will enable inducible polymer degradation in biologically relevant environments, enamine N-oxide crosslinkers are developed for double network acrylamide-based polymer/alginate hydrogels. Bioorthogonal dissociation initiated by the application of aqueous diboron solution through several delivery mechanisms effectively lead to polymer degradation. Their degradation by aqueous B2(OH)4 solution results in a fracture energy half-life of <10 min. The biocompatibility of the degradable hydrogels and B2(OH)4 reagent is assessed, and the removability of strongly adhered tough hydrogels on mice skin is evaluated. Thermoresponsive PNiPAAm/Alg hydrogels are fabricated and application of the hydrogels as a chemically inducible degradable intraoral wound dressing is demonstrated. It is demonstrated through in vivo maximum tolerated dose studies that diboron solution administered to mice by oral gavage is well tolerated. Successful integration of enamine N-oxides within the tough double network hydrogels as chemically degradable motifs demonstrates the applicability of enamine N-oxides in the realm of polymer chemistry and highlights the importance of chemically induced bioorthogonal dissociation reactions for materials science.
Collapse
Affiliation(s)
- Thomas T. Kim
- School of Chemistry and BiochemistryGeorgia Institute of TechnologyAtlantaGA30332USA
- Department of Cancer BiologyDana‐Farber Cancer InstituteBostonMA02215USA
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical SchoolBostonMA02115USA
| | - Deep Malu
- George W. Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
- School of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Dongjing He
- George W. Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
- School of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Yuhang Hu
- George W. Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
- School of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Justin Kim
- School of Chemistry and BiochemistryGeorgia Institute of TechnologyAtlantaGA30332USA
- Department of Cancer BiologyDana‐Farber Cancer InstituteBostonMA02215USA
- Department of Biological Chemistry and Molecular PharmacologyHarvard Medical SchoolBostonMA02115USA
| |
Collapse
|
36
|
Kaden T, Alonso‐Román R, Stallhofer J, Gresnigt MS, Hube B, Mosig AS. Leveraging Organ-on-Chip Models to Investigate Host-Microbiota Dynamics and Targeted Therapies for Inflammatory Bowel Disease. Adv Healthc Mater 2025; 14:e2402756. [PMID: 39491534 PMCID: PMC12004439 DOI: 10.1002/adhm.202402756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/29/2024] [Indexed: 11/05/2024]
Abstract
Inflammatory bowel disease (IBD) is an idiopathic gastrointestinal disease with drastically increasing incidence rates. Due to its multifactorial etiology, a precise investigation of the pathogenesis is extremely difficult. Although reductionist cell culture models and more complex disease models in animals have clarified the understanding of individual disease mechanisms and contributing factors of IBD in the past, it remains challenging to bridge research and clinical practice. Conventional 2D cell culture models cannot replicate complex host-microbiota interactions and stable long-term microbial culture. Further, extrapolating data from animal models to patients remains challenging due to genetic and environmental diversity leading to differences in immune responses. Human intestine organ-on-chip (OoC) models have emerged as an alternative in vitro model approach to investigate IBD. OoC models not only recapitulate the human intestinal microenvironment more accurately than 2D cultures yet may also be advantageous for the identification of important disease-driving factors and pharmacological interventions targets due to the possibility of emulating different complexities. The predispositions and biological hallmarks of IBD focusing on host-microbiota interactions at the intestinal mucosal barrier are elucidated here. Additionally, the potential of OoCs to explore microbiota-related therapies and personalized medicine for IBD treatment is discussed.
Collapse
Affiliation(s)
- Tim Kaden
- Dynamic42 GmbH07745JenaGermany
- Institute of Biochemistry IICenter for Sepsis Control and CareJena University Hospital07747JenaGermany
| | - Raquel Alonso‐Román
- Department of Microbial Pathogenicity MechanismsLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Junior Research Group Adaptive Pathogenicity StrategiesLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
| | | | - Mark S. Gresnigt
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Junior Research Group Adaptive Pathogenicity StrategiesLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
| | - Bernhard Hube
- Department of Microbial Pathogenicity MechanismsLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Institute of MicrobiologyFaculty of Biological SciencesFriedrich Schiller University07743JenaGermany
| | - Alexander S. Mosig
- Institute of Biochemistry IICenter for Sepsis Control and CareJena University Hospital07747JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
| |
Collapse
|
37
|
Wang R, Wang Z, Zhang M, Zhong D, Zhou M. Application of photosensitive microalgae in targeted tumor therapy. Adv Drug Deliv Rev 2025; 219:115519. [PMID: 39955076 DOI: 10.1016/j.addr.2025.115519] [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: 11/27/2024] [Revised: 01/02/2025] [Accepted: 01/18/2025] [Indexed: 02/17/2025]
Abstract
Microalgae present a novel and multifaceted approach to cancer therapy by modulating the tumor-associated microbiome (TAM) and the tumor microenvironment (TME). Through their ability to restore gut microbiota balance, reduce inflammation, and enhance immune responses, microalgae contribute to improved cancer treatment outcomes. As photosynthetic microorganisms, microalgae exhibit inherent anti-tumor, antioxidant, and immune-regulating properties, making them valuable in photodynamic therapy and tumor imaging due to their capacity to generate reactive oxygen species. Additionally, microalgae serve as effective drug delivery vehicles, leveraging their biocompatibility and unique structural properties to target the TME more precisely. Microalgae-based microrobots further expand their therapeutic potential by autonomously navigating complex biological environments, offering a promising future for precision-targeted cancer treatments. We position microalgae as a multifunctional agent capable of modulating TAM, offering novel strategies to enhance TME and improve the efficacy of cancer therapies.
Collapse
Affiliation(s)
- Ruoxi Wang
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310029, China; Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China; Zhejiang University-Ordos City Etuoke Banner Joint Research Center, Haining 314400, China
| | - Zhouyue Wang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China
| | - Min Zhang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China
| | - Danni Zhong
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | - Min Zhou
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310029, China; Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China; Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China; Zhejiang University-Ordos City Etuoke Banner Joint Research Center, Haining 314400, China.
| |
Collapse
|
38
|
Kovynev A, Charchuta MM, Begtašević A, Ducarmon QR, Rensen PCN, Schönke M. Combination of dietary fiber and exercise training improves fat loss in mice but does not ameliorate MASLD more than exercise alone. Am J Physiol Gastrointest Liver Physiol 2025; 328:G399-G410. [PMID: 40033967 DOI: 10.1152/ajpgi.00317.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/02/2025] [Accepted: 02/20/2025] [Indexed: 03/05/2025]
Abstract
Lifestyle interventions, such as diet and exercise, are currently the main therapies against metabolic dysfunction-associated steatotic liver disease (MASLD). However, not much is known about the combined impact of fiber and exercise on the modulation of gut-liver axis and MASLD amelioration. Here, we studied the impact of the combination of exercise training and a fiber-rich diet on the amelioration of MASLD. Male APOE*3-Leiden.CETP mice were fed a high-fat high-cholesterol diet with or without the addition of fiber (10% inulin) and exercise trained on a treadmill, or remained sedentary. Exercise training and fiber supplementation reduced fat mass gain and lowered plasma glucose levels. Only the combination treatment, however, induced fat loss and decreased plasma triglyceride and cholesterol levels compared with sedentary control mice. Exercise training with and without the addition of fiber had a similar ameliorating effect on the MASLD score. Only exercise without fiber decreased the hepatic expression of inflammatory markers. Fiber diet was mainly responsible for remodeling the gut microbial composition, with an increase in the relative abundance of the short-chain fatty acid (SCFA)-producing genera Anaerostipes and Muribaculaceae, whereas, surprisingly, exercise training alone and with fiber resulted in the highest increase of SCFA production. Overall, the combination of exercise training and dietary fiber decreases fat mass and improves glucose and lipid homeostasis but does not have an additional synergistic positive effect on liver health compared with exercise training alone.NEW & NOTEWORTHY The combination of dietary fiber intake and exercise training has a synergetic beneficial effect on the metabolic health, resulting in fat loss, lowered blood glucose, and lowered plasma lipid levels in mice with steatotic liver disease. However, fiber supplementation, despite a positive remodulation of the gut-liver axis, does not have an additional positive effect on liver health compared with exercise training alone.
Collapse
Affiliation(s)
- Artemiy Kovynev
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Mikołaj M Charchuta
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Amina Begtašević
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Quinten R Ducarmon
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Milena Schönke
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
39
|
Wang Z, Zhang C. Nanomaterials for targeted therapy of kidney diseases: Strategies and advances. Mater Today Bio 2025; 31:101534. [PMID: 39990736 PMCID: PMC11846943 DOI: 10.1016/j.mtbio.2025.101534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/21/2025] [Accepted: 01/28/2025] [Indexed: 02/25/2025] Open
Abstract
The treatment and management of kidney diseases pose a significant global burden. Due to the presence of blood circulation barriers and glomerular filtration barriers, drug therapy for kidney diseases faces challenges such as poor renal targeting, short half-life, and severe systemic side effects, severely hindering therapeutic progress. Therefore, the research and development of kidney-targeted therapeutic agents is of great clinical significance. In recent years, the application of nanotechnology in the field of nephrology has shown potential for revolutionizing the diagnosis and treatment of kidney diseases. Carefully designed nanomaterials can exhibit optimal biological characteristics, influencing various aspects such as circulation, retention, targeting, and excretion. Rationally designing and modifying nanomaterials based on the anatomical structure and pathophysiological environment of the kidney to achieve highly specific kidney-targeted nanomaterials or nanodrug delivery systems is both feasible and promising. Based on the targeted therapy of kidney diseases, this review discusses the advantages and limitations of current nanomedicine in the targeted therapy of kidney diseases, and summarizes the application and challenges of current renal active/passive targeting strategies, in order to further promote the development of kidney-targeted nanomedicine through a preliminary summary of previous studies and future prospects.
Collapse
Affiliation(s)
- Zhiwen Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
40
|
Wei S, Yang D, Shou Z, Zhang Y, Zheng S, Zan X, Li L, Zhang C. Proanthocyanidin capsules remodel the ROS microenvironment via regulating MAPK signaling for accelerating diabetic wound healing. Mater Today Bio 2025; 31:101467. [PMID: 39896292 PMCID: PMC11786704 DOI: 10.1016/j.mtbio.2025.101467] [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: 09/27/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 02/04/2025] Open
Abstract
Defective diabetic wound healing is a major clinical challenge, where hyperglycemia at the wound site induces excessive reactive oxygen species (ROS) which activate the MAPK pathway (particularly p38 MAPK), resulting in sustained release of inflammatory factors and cellular damage/apoptosis. Polyphenols are efficient ROS scavengers which reduce the level of inflammation at the wound site and promote wound healing, but the low bioavailability limits their biomedical application. This study developed a simple and highly efficient method for preparing proanthocyanidin (PC) capsules through hydrogen bonding and hydrophobic interactions among PC molecules. PC capsules can continuously scavenge free radicals and release proanthocyanidins, significantly enhancing their bioavailability. A single dose of PC capsules accelerates wound healing in diabetic mice by regulating the p38 MAPK signaling cascade, reducing inflammatory mediator concentration, inhibiting cell apoptosis, and remodeling the wound microenvironment. This research makes an important contribution to the field of enhancing polyphenol bioavailability for wound healing and reveals the potential of modulating the MAPK pathway for treating other inflammation and oxidative stress-related diseases.
Collapse
Affiliation(s)
- Shaoyin Wei
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, Zhejiang, China
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong, 250100, China
| | - Dong Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, Zhejiang, China
| | - Zeyu Shou
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yipiao Zhang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, No. 18, Chaowang Road, Gongshu District, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
- Zhejiang Provincial Key Laboratory of TCM for Innovative R&D and Digital Intelligent Manufacturing of TCM Great Health Products, Huzhou, 313200, China
| | - Shengwu Zheng
- Wenzhou Celecare Medical Instruments Co. Ltd, Wenzhou, 325000, China
| | - Xingjie Zan
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, Zhejiang, China
| | - Lianxin Li
- Department of Orthopaedics Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan City, Shandong Province, 250021, China
| | - Chunwu Zhang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, Zhejiang, China
| |
Collapse
|
41
|
Wu Z, Xu K, Huang R, Wang X, Teng JLL, Yu X, Jin L, Li Q, Leung KCF, Wong HM, Li X. Cyborg microbe biohybrids with metal-organic coating layers: Strategies, functionalisation and potential applications. Mater Today Bio 2025; 31:101642. [PMID: 40161925 PMCID: PMC11950775 DOI: 10.1016/j.mtbio.2025.101642] [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: 01/03/2025] [Revised: 02/10/2025] [Accepted: 03/06/2025] [Indexed: 04/02/2025] Open
Abstract
The integration of living microbes, specifically bacteria and fungi, with metal-organic nanocoatings has led to the recent development of cyborg microbe biohybrids, which show excellent adaptability and functionality for a wide range of potential applications in biotechnology and medicine. This review discusses the strategies, functionalisation, and applications of these biohybrids, which are categorised into two types of coatings: metal-organic frameworks (MOFs) and metal-phenolic networks (MPNs). Key advances in their synthetic approaches via in-situ and pre-synthesised coatings are crucially addressed, and yet the methodology details and specific advantages are highlighted. Despite the notable advancements, there are various limitations and challenges, such as determination of the long-term viability and stability of the biohybrids, insufficient work on their theranostic applications and essentially scaling-up difficulties for industrial and clinical translation. The latest advancements in the biohybrids and related technology have established a critical foundation for enhancing innovative studies through the strong interdisciplinary teamwork.
Collapse
Affiliation(s)
- Zichen Wu
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, PR China
| | - Ke Xu
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, PR China
| | - Regina Huang
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, PR China
| | - Xinna Wang
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong SAR, PR China
| | - Jade Lee-Lee Teng
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, PR China
| | - Xiaolin Yu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, PR China
| | - Lijian Jin
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, PR China
| | - Quanli Li
- Institute of Oral Science, Department of Stomatology, Longgang Otorhinolaryngology Hospital, No. 3004L Longgang Avenue, Shenzhen, PR China
- Key Lab of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Meishan Road, Hefei, PR China
| | - Ken Cham-Fai Leung
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong SAR, PR China
| | - Hai Ming Wong
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, PR China
| | - Xuan Li
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, PR China
| |
Collapse
|
42
|
Yu Q, Yang J, Liu L, Huang Y, Wang E, Li D, Yuan H. One-step immobilization of chitosanase on microcrystalline cellulose using a carbohydrate binding module family 2. Carbohydr Polym 2025; 353:123291. [PMID: 39914986 DOI: 10.1016/j.carbpol.2025.123291] [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: 11/07/2024] [Revised: 01/05/2025] [Accepted: 01/16/2025] [Indexed: 05/07/2025]
Abstract
Enzyme immobilization technology holds significant value in biocatalysis. Carbohydrate-binding modules (CBMs), with their specific binding to natural polysaccharides, offer a highly promising immobilization method. In the present study, the binding ability with their natural substrates and heterologous expression levels of four CBMs using fluorescent protein tagging were studied, revealing that CBM2r presented the highest immobilization efficiency and expression level. Using the Design of Experiments (DOE), the immobilization conditions for mCherry-CBM2r were optimized, achieving a protein loading of 2.45 wt% on Avicel under optimal conditions: a solid-liquid ratio of 1:30, NaCl concentration of 108 mM, protein concentration of 6 mg/mL, and incubation time of 120 min. Subsequently, CBM2r gene was fused with chitosanase gene from Bacillus subtilis (BsCsn) and expressed in Escherichia coli for establishing a novel one-step immobilization of fusion enzymes mediated by CBM2r on microcrystalline cellulose. The immobilized CBM2r-BsCsn-Avicel was used for batch hydrolysis of high-concentration chitosan to produce chito-oligosaccharides, with the enzyme retaining 96 % substrate degradation efficiency over seven cycles and achieving a space-time yield of 232.8 kg/m3/h. This study provides a simple, cost-effective, environment friendly, and competitive biocatalytic immobilization strategy.
Collapse
Affiliation(s)
- Qijun Yu
- State Key Laboratory of Animal Biotech Breeding, and Key Laboratory of Soil Microbiology, Ministry of Agriculture, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jinshui Yang
- State Key Laboratory of Animal Biotech Breeding, and Key Laboratory of Soil Microbiology, Ministry of Agriculture, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Liang Liu
- State Key Laboratory of Animal Biotech Breeding, and Key Laboratory of Soil Microbiology, Ministry of Agriculture, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yaru Huang
- State Key Laboratory of Animal Biotech Breeding, and Key Laboratory of Soil Microbiology, Ministry of Agriculture, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Entao Wang
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Dongmei Li
- State Key Laboratory of Animal Biotech Breeding, and Key Laboratory of Soil Microbiology, Ministry of Agriculture, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Hongli Yuan
- State Key Laboratory of Animal Biotech Breeding, and Key Laboratory of Soil Microbiology, Ministry of Agriculture, College of Biological Sciences, China Agricultural University, Beijing, China.
| |
Collapse
|
43
|
Wu Q, Lu Z, Wang L, Peng S, Wang Z, Qiu Y, Liao Z, Wang Y, Qin X. Konjac glucomannan/xanthan gum hydrogels loaded with metal-phenolic networks encapsulated probiotic to promote infected wound healing. Carbohydr Polym 2025; 353:123243. [PMID: 39914948 DOI: 10.1016/j.carbpol.2025.123243] [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: 11/11/2024] [Revised: 01/02/2025] [Accepted: 01/06/2025] [Indexed: 05/07/2025]
Abstract
Probiotic hydrogel systems have been reported to promote healing of infected wounds by secreting functional bioactive secondary metabolites (BSM) of probiotics. Herein, Bacillus subtilis (B. subtilis), a probiotic, are encapsulated via a metal-phenolic networks (MPNs) and loaded into konjac glucomannan/xanthan gum-based hydrogels for wound repair. This MPNs were designed and composed of Bletilla striata polysaccharide, procyanidin, Ca2+, which can enhance the cross-linking through hydrogen bonding to form the KGXM-PCB@Bsubtilis hydrogel, protects the probiotic from antibiotics and prevents B. subtilis from escaping into the wound microenvironment, thereby avoiding exposure to a possible threat. Moreover, the KGXM-PCB@Bsubtilis hydrogel not only exhibits superior mechanical characteristics and biocompatibility, but also shows excellent antimicrobial, antioxidant and anti-inflammatory properties that can inhibit the growth of Staphylococcus aureus, remove the active oxygens, and promote cell migration. In vivo experiments showed that after treatment with the KGXM-PCB@Bsubtilis hydrogel, the wound healing rate reached 98.31 % on day 14, and collagen deposition was highly expressed (81.11 ± 2.20 %), which promoted wound healing and regeneration of new tissue. This study provides new ideas for developing wound dressings based on living bacterial hydrogels.
Collapse
Affiliation(s)
- Qingxin Wu
- School of Biological Engineering, Zhuhai Campus of Zunyi Medical University, Guangdong 519000, China
| | - Zhengfeng Lu
- School of Biological Engineering, Zhuhai Campus of Zunyi Medical University, Guangdong 519000, China
| | - Lantao Wang
- School of Biological Engineering, Zhuhai Campus of Zunyi Medical University, Guangdong 519000, China
| | - Shuting Peng
- School of Biological Engineering, Zhuhai Campus of Zunyi Medical University, Guangdong 519000, China
| | - Zhengxiao Wang
- School of Biological Engineering, Zhuhai Campus of Zunyi Medical University, Guangdong 519000, China
| | - Yan Qiu
- School of Biological Engineering, Zhuhai Campus of Zunyi Medical University, Guangdong 519000, China
| | - Zhengping Liao
- School of Biological Engineering, Zhuhai Campus of Zunyi Medical University, Guangdong 519000, China
| | - Yarong Wang
- School of Biological Engineering, Zhuhai Campus of Zunyi Medical University, Guangdong 519000, China
| | - Xiaofei Qin
- School of Biological Engineering, Zhuhai Campus of Zunyi Medical University, Guangdong 519000, China.
| |
Collapse
|
44
|
Liu J, Wang H, Zhang C. Acteoside-Loaded Self-Healing Hydrogel Enhances Skin Wound Healing through Modulation of Hair Follicle Stem Cells. Cell Mol Bioeng 2025; 18:163-183. [PMID: 40290106 PMCID: PMC12018657 DOI: 10.1007/s12195-025-00845-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/18/2025] [Indexed: 04/30/2025] Open
Abstract
Background Skin wound healing is a complex biological process involving cellular, molecular, and physiological events. Traditional treatments often fail to provide optimal outcomes, particularly for chronic wounds. Objectives This study aimed to develop a self-healing hydrogel loaded with Acteoside, a bioactive compound with antioxidant and anti-inflammatory properties, to enhance skin wound healing. Methods Using transcriptomic analysis, Rab31 was identified as a key target of Acteoside in regulating hair follicle stem cells (HFSCs). In vitro assays demonstrated that Acteoside promotes HFSC proliferation, migration, and differentiation by upregulating Rab31 expression. The self-healing hydrogel was prepared using quaternized chitosan derivatives, which exhibited excellent mechanical properties, antibacterial, and antioxidant activities. Results In vivo studies in a mouse model showed that Acteoside-loaded hydrogel significantly accelerated wound healing, promoting skin regeneration and improving wound closure. Conclusions This research highlights the potential of Acteoside-loaded self-healing hydrogels as an innovative therapeutic strategy for enhancing skin wound healing. By modulating HFSC activity, this hydrogel offers a promising solution for improving healing outcomes in challenging wound environments. Graphical Abstract Schematic representation of an injectable self-healing hydrogel loaded with the phenylethanoid compound acteoside for regulating the proliferation and differentiation of HFSCs to mediate the healing of skin wounds. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-025-00845-2.
Collapse
Affiliation(s)
- Junyu Liu
- Shandong Ruixin Pharmaceutical Company, Yishui County, Intersection of Tengfei East Road and East Second Ring Road, Linyi, 276400 Shandong Province China
| | - Hua Wang
- Shandong Luoxin Pharmaceutical Group Stock Company, Linyi, 276017 China
| | - Caihua Zhang
- Shandong Ruixin Pharmaceutical Company, Yishui County, Intersection of Tengfei East Road and East Second Ring Road, Linyi, 276400 Shandong Province China
| |
Collapse
|
45
|
Tufail MA, Schmitz RA. Exploring the Probiotic Potential of Bacteroides spp. Within One Health Paradigm. Probiotics Antimicrob Proteins 2025; 17:681-704. [PMID: 39377977 PMCID: PMC11925995 DOI: 10.1007/s12602-024-10370-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2024] [Indexed: 03/21/2025]
Abstract
Probiotics are pivotal in maintaining or restoring the balance of human intestinal microbiota, a crucial factor in mitigating diseases and preserving the host's health. Exploration into Bacteroides spp. reveals substantial promise in their development as next-generation probiotics due to their profound interaction with host immune cells and capability to regulate the microbiome's metabolism by significantly impacting metabolite production. These beneficial bacteria exhibit potential in ameliorating various health issues such as intestinal disorders, cardiovascular diseases, behavioral disorders, and even cancer. Though it's important to note that a high percentage of them are as well opportunistic pathogens, posing risks under certain conditions. Studies highlight their role in modifying immune responses and improving health conditions by regulating lymphocytes, controlling metabolism, and preventing inflammation and cancer. The safety and efficacy of Bacteroides strains are currently under scrutiny by the European Commission for authorization in food processing, marking a significant step towards their commercialization. The recent advancements in bacterial isolation and sequencing methodologies, coupled with the integration of Metagenome-Assembled Genomes (MAGs) binning from metagenomics data, continue to unveil the potential of Bacteroides spp., aiding in the broader understanding and application of these novel probiotics in health and disease management.
Collapse
Affiliation(s)
- Muhammad Aammar Tufail
- Institut für Allgemeine Mikrobiologie, Christian-Albrechts-Universität zu Kiel, 24118, Kiel, Germany.
| | - Ruth A Schmitz
- Institut für Allgemeine Mikrobiologie, Christian-Albrechts-Universität zu Kiel, 24118, Kiel, Germany.
| |
Collapse
|
46
|
Zhao YY, Wu ZJ, Du Y, Han QQ, Bai YY, Liu B, Li J. Gut microbiome and serum metabolites in neuropathic pain: The PPARα perspective. Behav Brain Res 2025; 482:115442. [PMID: 39864460 DOI: 10.1016/j.bbr.2025.115442] [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: 12/03/2024] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 01/28/2025]
Abstract
Neuropathic pain (NP) is a chronic disease state centred on neuroinflammation with a high prevalence and limited effective treatment options. Peroxisome proliferator-activated receptor α (PPARα) has emerged as a promising target for NP management due to its anti-inflammatory properties. Recent evidence highlights the critical role of the gut microbiome and its metabolites in NP pathogenesis. This study aimed to investigate whether PPARα modulates the development and alleviation of NP by influencing gut microbial communities and serum metabolites. 16S rDNA sequencing and liquid chromatography-mass spectrometry (LC-MS/MS) untargeted metabolomics analyses performed 14 days after the establishment of a chronic constriction injury (CCI) pain model in C57BL/6 J mice showed significant changes in gut microbial and metabolite levels in CCI mice. Intraperitoneal injection of the PPARα agonist GW7647 (5 mg/kg) significantly attenuated mechanical allodynia and thermal hyperalgesia in CCI mice, whereas injection of the PPARα antagonist GW6471 (20 mg/kg) produced the opposite effect. Immunofluorescence analysis revealed that GW7647 effectively suppressed microglial activation. Additionally, PPARα agonist and antagonist treatments markedly altered the composition and abundance of intestinal microbial communities in CCI mice. Further serum LC-MS/MS analysis identified 258 potential serum metabolic biomarkers, many of which correlated with changes in gut microbial composition. These findings demonstrate that PPARα influences serum metabolite profiles by modulating gut microbiota composition, which subsequently affects NP progression. This study provides novel insights into the mechanisms underlying NP and suggests potential therapeutic avenues targeting PPARα and gut microbiota.
Collapse
Affiliation(s)
- Yu-Ying Zhao
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Zi-Jun Wu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Yue Du
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Qing-Qing Han
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Yuan-Yuan Bai
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Bin Liu
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China; Center for Critical Care Medicine, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin 300020, China.
| | - Jing Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China.
| |
Collapse
|
47
|
Balnadupete A, Moideen FM, Varughese A, Mugaranja K, M JT, Charavu R, Bhandary Y. Beyond the pill: incrimination of nuclear factor-kappa B and their targeted phytomedicine for pulmonary fibrosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04067-1. [PMID: 40137966 DOI: 10.1007/s00210-025-04067-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
Pulmonary fibrosis (PF) is a slow and irreparable damage of the lung caused by the accumulation of scar tissue, which eventually results in organ dysfunction and fatality from gas exchange failure. One of the extensively studied inflammatory pathways in PF is the NF-κB signalling pathway, which is reportedly involved in epithelial-mesenchymal transition, myofibroblast differentiation, and other cellular processes. Additionally, studies have evidence that NF-κB signalling pathways can be employed as a potential target for developing therapeutic agents against PF. In the current scenario, FDA-approved drugs, nintedanib and pirfenidone, have been used for the treatment of PF with potential side effects. Recently, the usage of bioactive compounds has attracted attention in the treatment of PF. This review focuses on the involvement of the NF-κB signalling pathway in PF and the significance of phytocompounds in regulating the NF-κB pathway. Both the in vitro and in vivo studies reveal that NF-κB-targeted plant-based bioactive compounds significantly ameliorate the PF condition as well as improve the health condition. Databases such as Scopus, PubMed, and Web of Science were used to conduct literature surveys and compile data on all the bioactive compounds. In conclusion, the plant-derived bioactive compounds are potent enough to target the NF-κB with its biological properties, and this could be a highly effective therapeutic strategy for PF in the future.
Collapse
Affiliation(s)
- Akarsha Balnadupete
- Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore, 575018, Karnataka, India
| | - Fathimath Muneesa Moideen
- Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore, 575018, Karnataka, India
| | - Aleena Varughese
- Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore, 575018, Karnataka, India
| | - Kirana Mugaranja
- Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore, 575018, Karnataka, India
| | - Jeena T M
- Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore, 575018, Karnataka, India
| | - Rakshitha Charavu
- Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore, 575018, Karnataka, India
| | - Yashodhar Bhandary
- Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore, 575018, Karnataka, India.
- Specialized Research Unit, Yenepoya Medical College & Hospital, Yenepoya (Deemed to Be University), Deralakatte, Mangalore, Karnataka, 575018, India.
| |
Collapse
|
48
|
Hu W, Wang Y, Han J, Zhang W, Chen J, Li X, Wang L. Microfluidic organ-on-a-chip models for the gut-liver axis: from structural mimicry to functional insights. Biomater Sci 2025; 13:1624-1656. [PMID: 40019226 DOI: 10.1039/d4bm01273a] [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: 03/01/2025]
Abstract
The gut-liver axis plays a crucial role in maintaining metabolic balance and overall human health. It orchestrates various processes, such as blood flow, nutrient transfer, metabolite processing, and immune cell communication between the two organs. Traditional methods, such as animal models and two-dimensional (2D) cell cultures, are insufficient in fully replicating the intricate functions of the gut-liver axis. The emergence of microfluidic technology has revolutionized this field, facilitating the development of organ-on-a-chip (OOC) systems. These systems are capable of mimicking the complex structures and dynamic environments of the gut and liver in vitro and incorporating sensors for real-time monitoring. In this article, we review the latest progress in gut-on-a-chip (GOC) and liver-on-a-chip (LOC) systems, as well as the integrated gut-liver-on-a-chip (GLOC) models. Our focus lies in the simulation of physiological parameters, three-dimensional (3D) structural mimicry, microbiome integration, and multicellular co-culture. All these aspects are essential for constructing accurate in vitro models of the gut and liver. Furthermore, we explore the current applications of OOC technology in the study of the gut and liver, including its use in disease modeling, toxicity testing, and drug screening. Finally, we discuss the challenges that remain and outline potential future directions for advancing GOC and LOC development in vitro.
Collapse
Affiliation(s)
- Wanlin Hu
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Yushen Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Junlei Han
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Wenhong Zhang
- College of Mechanical Engineering, Donghua University, Shanghai 201620, China
| | - Jun Chen
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Xinyu Li
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
| | - Li Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| |
Collapse
|
49
|
Deng J, Hu Y, Zhu P, Yu Y, Chen Q, Wu H, Zha Z, Wang H, Ma Y. Probiotic Delivery for Editing of the Gut Microbiota to Mitigate Colitis and Maintain Hepatic Homeostasis Via Gut-Liver Axis. ACS NANO 2025; 19:10500-10514. [PMID: 40047584 DOI: 10.1021/acsnano.5c00325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Inflammatory bowel disease (IBD) compromises the intestinal barrier and disrupts gut microbiota, impacting liver function via the gut-liver axis, which in turn influences the intestinal microbiota through lipid metabolites exacerbating IBD. This study introduced a probiotic-based treatment using Lactobacillus acidophilus encapsulated in tungsten ion-loaded mesoporous polydopamine (LA@WMPDA) to ameliorate colitis and balance enterohepatic homeostasis. After oral administration, the encapsulation could protect Lactobacillus acidophilus, scavenge reactive oxygen/nitrogen species, and the released tungsten ions would inhibit abnormal Enterobacteriaceae growth during colitis, consequently restoring the intestinal barrier and regulating the gut microbiota. Nontargeted metabolomics and transcriptomics analyses showed increased short-chain fatty acids and indole derivatives, and decreased hepatic lipid metabolism. Pathways associated with immune response, cell migration and death, and response to bacterium showed significant down-regulation in the colon and liver transcriptome analysis. Thus, this study provided a pioneered paradigm for IBD treatment and highlighted the regulation of liver-related metabolic functions via the gut-liver axis.
Collapse
Affiliation(s)
- Junwei Deng
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Yaoyu Hu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Pengfei Zhu
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, P. R. China
| | - Yi Yu
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, P. R. China
| | - Qian Chen
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Haitao Wu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, P. R. China
| | - Zhengbao Zha
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, P. R. China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, P. R. China
| | - Yan Ma
- Research Center for Translational Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, P. R. China
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, P. R. China
| |
Collapse
|
50
|
Keung WS, Zhang WH, Luo HY, Chan KC, Chan YM, Xu J. Correlation between the structures of natural polysaccharides and their properties in regulating gut microbiota: Current understanding and beyond. Carbohydr Polym 2025; 352:123209. [PMID: 39843110 DOI: 10.1016/j.carbpol.2024.123209] [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: 11/18/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 01/24/2025]
Abstract
Natural polysaccharides have complex structural properties and a wide range of health-promoting effects. Accumulating evidence suggests that the effects are significantly mediated through fermentation by gut microbiota. In recent years, the relationship between the structures of natural polysaccharides and their properties in regulating gut microbiota has garnered significant research attention as researchers attempt to precisely understand the role of gut microbiota in the bioactivities of natural polysaccharides. Progress in this niche, however, remains limited. In this review, we first provide an overview of current research investigating this structure-property relationship. We then present a detailed correlation analysis between the structural characteristics of 159 purified natural polysaccharides and their effects on gut microbiota reported over the past two decades. The analysis revealed that diverse gut bacteria show specific correlations with the molecular weight, glycosidic linkages, and monosaccharide composition of natural polysaccharides. Multifaceted molecular mechanisms, including carbohydrate binding, enzymatic degradation, and cross-feeding, were proposed to be collectively involved in these correlations. Finally, we offer our perspective on future studies to further improve our understanding of the relationship between polysaccharide structure and gut microbiota regulation.
Collapse
Affiliation(s)
- Wing-Shan Keung
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong
| | - Wei-Hao Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong
| | - Han-Yan Luo
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong
| | - Kam-Chun Chan
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong
| | - Yui-Man Chan
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong
| | - Jun Xu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong.
| |
Collapse
|