51
|
Huang Y, Cai H, Liu H, Wang L, Feng G, Ding Z, Fei Y, Li A, Fang J. Probiotic nanocomposite materials with excellent resistance, inflammatory targeting, and multiple efficacies for enhanced treatment of colitis in mice. J Nanobiotechnology 2025; 23:188. [PMID: 40055678 PMCID: PMC11889847 DOI: 10.1186/s12951-025-03240-1] [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: 11/02/2024] [Accepted: 02/18/2025] [Indexed: 05/13/2025] Open
Abstract
The occurrence of inflammatory bowel disease (IBD) is relevant to impaired intestinal mucosal barrier and disordered gut microbiota, subsequently leading to excessive production of reactive oxygen species (ROS) and elevated levels of inflammatory factors. Traditional therapies focus on inhibiting inflammation, but the vast majority involve non-targeted systemic administration, whose long-term use may result in potential side effects. Oral microbial therapy has exhibited great application prospects currently in IBD treatment; however, its progress has been slowed by issues with deficient bioavailability, poor targeting of colitis, and low therapeutic efficacy. Consequently, it is exceedingly desirable to develop a strategy by which probiotics can be endowed with additional anti-inflammatory and antioxidant properties, as well as enhanced targeting of the inflamed intestine. Herein, we present an innovative therapeutic strategy for encapsulating probiotic Bacillus coagulans spores with rosmarinic acid (RA) and silk fibroin (SF). Probiotics in spore morphology possessed strong gastrointestinal environmental resistance; RA alleviated oxidative damage by scavenging ROS and inhibited inflammatory responses; SF assisted probiotics release and colonize in the inflamed intestine. We demonstrated the therapeutic efficacy of probiotic composite materials in a colitis mouse model, which significantly alleviated a series of colitis symptoms, inhibited inflammatory cytokine storms, restored the balance of the gut microbiota, and downregulated inflammation-related signaling pathways. We are optimistic that the utilization of therapeutic nanocoating to modify probiotics will open up novel avenues for future microbial therapy targeting IBD.
Collapse
Affiliation(s)
- Yuewen Huang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, China
| | - Hongting Cai
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, China
| | - Huipeng Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, China
| | - Lixiang Wang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, China
| | - Guangfu Feng
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, China.
| | - Zizi Ding
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, China
| | - Yanquan Fei
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, China
| | - Aike Li
- Feed Research Institute/Grain Quality & Nutrition Institute, Key Laboratory of Grain and Oil Biotechnology of National Food and Strategic Reserves Administration, Academy of National Food and Strategic Reserves Administration, Beijing, 100037, China.
| | - Jun Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410128, China.
| |
Collapse
|
52
|
Wang W, Chen Y, Wang Y, Wang Y, Zhang W, Dai K, Geng W, Tang S. Azo-linked 5-ASA-coumarin prodrug: Fluorescent tracking for colonic drug release in UC treatment. Talanta 2025; 284:127277. [PMID: 39608145 DOI: 10.1016/j.talanta.2024.127277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/12/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024]
Abstract
Theranostic prodrugs that enable real-time, non-invasive monitoring of drug release and biodistribution are highly desirable for optimizing therapeutic efficacy and guiding personalized medication. Herein, we report a colon-targeted theranostic prodrug system (P1) for the simultaneous delivery and tracking of 5-aminosalicylic acid (5-ASA) in the treatment of ulcerative colitis (UC). P1 comprises a fluorescent 7-amino-4-methylcoumarin (7-AMC) reporter covalently linked to 5-ASA via an azo bond, which quenches the fluorescence of 7-AMC until P1 is activated by azoreductases in the colonic microenvironment. This selective activation triggers the release of 5-ASA and the revival of 7-AMC fluorescence, enabling real-time monitoring of drug delivery. To improve the solubility and targeted delivery of P1, it was encapsulated within polymeric micelles (PM) that selectively adhere to the positively charged, inflamed colonic tissues. In vitro studies confirmed the stability, biocompatibility, and selective activation of P1 under simulated colonic conditions. Notably, in a mouse model of UC, the P1-loaded PM achieved targeted delivery of 5-ASA to the inflamed colon, resulting in effective attenuation of colitis symptoms. Importantly, the in situ activation of P1 allowed for the real-time, non-invasive visualization of drug release and biodistribution, providing valuable insights for treatment optimization. This theranostic prodrug approach offers a promising strategy for the simultaneous therapy and tracking of 5-ASA delivery in UC treatment, with the potential to facilitate personalized medication and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Wenchao Wang
- Department of Pain, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and BrainHealth), Wenzhou Medical University, Zhejiang, China
| | - Yingjie Chen
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China; University of Chinese Academy of Sciences, Wenzhou Institute, Zhejiang, China
| | - Yuan Wang
- University of Chinese Academy of Sciences, Wenzhou Institute, Zhejiang, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Zhejiang, China
| | - Yijian Wang
- Department of Pain, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and BrainHealth), Wenzhou Medical University, Zhejiang, China
| | - Wenjing Zhang
- Department of Pain, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and BrainHealth), Wenzhou Medical University, Zhejiang, China
| | - Keke Dai
- Department of Pain, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and BrainHealth), Wenzhou Medical University, Zhejiang, China
| | - Wujun Geng
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and BrainHealth), Wenzhou Medical University, Zhejiang, China.
| | - Sicheng Tang
- University of Chinese Academy of Sciences, Wenzhou Institute, Zhejiang, China.
| |
Collapse
|
53
|
Magalhães MI, Azevedo MJ, Castro F, Oliveira MJ, Costa ÂM, Sampaio Maia B. The link between obesity and the gut microbiota and immune system in early-life. Crit Rev Microbiol 2025; 51:264-284. [PMID: 38651972 DOI: 10.1080/1040841x.2024.2342427] [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: 12/23/2023] [Revised: 04/04/2024] [Accepted: 04/06/2024] [Indexed: 04/25/2024]
Abstract
In early-life, the gut microbiota is highly modifiable, being modulated by external factors such as maternal microbiota, mode of delivery, and feeding strategies. The composition of the child's gut microbiota will deeply impact the development and maturation of its immune system, with consequences for future health. As one of the main sources of microorganisms to the child, the mother represents a crucial factor in the establishment of early-life microbiota, impacting the infant's wellbeing. Recent studies have proposed that dysbiotic maternal gut microbiota could be transmitted to the offspring, influencing the development of its immunity, and leading to the development of diseases such as obesity. This paper aims to review recent findings in gut microbiota and immune system interaction in early-life, highlighting the benefits of a balanced gut microbiota in the regulation of the immune system.
Collapse
Affiliation(s)
- Maria Inês Magalhães
- Doctoral Program in Biomedical Sciences, ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- Nephrology and Infectious Diseases R&D group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- nBTT, NanoBiomaterials for Targeted Therapies group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FMDUP - Faculdade de Medicina Dentária da Universidade do Porto, Porto, Portugal
| | - Maria João Azevedo
- Nephrology and Infectious Diseases R&D group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- nBTT, NanoBiomaterials for Targeted Therapies group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FMDUP - Faculdade de Medicina Dentária da Universidade do Porto, Porto, Portugal
- Academic Center for Dentistry Amsterdam (ACTA), Universiteit van Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Flávia Castro
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Maria José Oliveira
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Ângela M Costa
- Tumor and Microenvironment Interactions group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Benedita Sampaio Maia
- Nephrology and Infectious Diseases R&D group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- nBTT, NanoBiomaterials for Targeted Therapies group, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FMDUP - Faculdade de Medicina Dentária da Universidade do Porto, Porto, Portugal
| |
Collapse
|
54
|
Hasannezhad H, Bakhshi A, Mozafari MR, Naghib SM. A review of chitosan role in milk bioactive-based drug delivery, smart packaging and biosensors: Recent advances and developments. Int J Biol Macromol 2025; 294:139248. [PMID: 39740715 DOI: 10.1016/j.ijbiomac.2024.139248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 12/21/2024] [Accepted: 12/25/2024] [Indexed: 01/02/2025]
Abstract
Chitosan, a versatile biopolymer derived from chitin, is increasingly recognized in the milk industry for its multifunctional applications in drug delivery, smart packaging, and biosensor development. This review provides a comprehensive analysis of recent advances in chitosan production techniques. These include chemical, biological, and novel methods such as deep eutectic solvents (DES), microwave-assisted approaches, and laser-assisted processes. Surface modification strategies to enhance its functional properties are also discussed. The review highlights the development of various chitosan-based nanocarriers, including nanoparticles, nanofibers, nanogels, and nanocomposites. It emphasizes their stability when combined with milk bioactive ingredients like lipids, peptides, lactose, and minerals. The gastrointestinal fate and safety of chitosan nanoparticles are critically evaluated, showcasing their potential for safe consumption in dairy-related applications. In drug delivery systems, chitosan exhibits excellent compatibility with milk-derived carbohydrates, proteins, and minerals, enabling the development of innovative drug delivery platforms. Additionally, its incorporation into smart packaging materials enhances the shelf-life and quality of dairy products. Chitosan-based biosensors offer precise contaminant detection in the milk industry by enabling precise detection of contaminants such as Bisphenol A, melamine, bacteria, drugs, antibiotics, toxins, heavy metals, and allergens, thus ensuring food safety and quality. Emerging trends, including the integration of artificial intelligence, advanced gene editing, and multifunctional chitosan, are discussed, offering insights into future personalized delivery systems and merging food and drug technologies. The review concludes by highlighting gaps in current research and offering recommendations for future exploration. These suggestions aim to optimize chitosan's unique properties to address key challenges in the milk industry. This article serves as a valuable resource for researchers, industry professionals, and policymakers aiming to innovate within the dairy sector using chitosan-based technologies.
Collapse
Affiliation(s)
- Hossein Hasannezhad
- Biomaterials and Tissue Engineering Research Group, Interdisciplinary Technologies Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran; Department of Food Science and Technology, Faculty of Agriculture and Food Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ali Bakhshi
- Biomaterials and Tissue Engineering Research Group, Interdisciplinary Technologies Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran; Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran, Iran
| | - M R Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran, Iran.
| |
Collapse
|
55
|
Zheng Y, Gao D, Xie H, Geng H. Epicatechin protects mice against OVA-induced asthma through inhibiting airway inflammation and modulating gut microbiota. Cell Signal 2025; 127:111609. [PMID: 39826677 DOI: 10.1016/j.cellsig.2025.111609] [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: 11/25/2024] [Revised: 12/21/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Allergic asthma is a chronic airway inflammatory reaction that seriously affects people's quality of life and even endangers their lives. The aim of this study was to explore the role of epicatechin (EC) on asthma and its potential mechanism. A mice model of allergic asthma was established by intraperitoneal injection of ovalbumin (OVA) with aluminum hydrogen solution, and nebulized inhalation of OVA to stimulate. EC (10, 20, 40 mg/kg) was administered 30 min before nebulization for three consecutive days. The results showed that EC attenuated OVA-induced lung injury, inflammatory cell infiltration, IgE, and inflammatory cytokine production. EC also inhibited OVA-induced NF-κB activation and increased Nrf2 and HO-1 expression. 16S rRNA sequencing analysis demonstrated that at genus level, EC significantly increased the abundance of Lachnospiraceae_NK4A136_group, Ligilactobacillus, Alloprevotella. Meanwhile, EC inhibited the abundance of Clostridia UCG-014, Helicobacter, Paramuribaculum, and Escherichia-Shigella. In conclusion, EC can effectively alleviate the symptoms of asthma in mice, which may through regulating the composition of gut microbiota and inhibiting inflammatory response.
Collapse
Affiliation(s)
- Yang Zheng
- Department of Gynecology, China-Japan Union Hospital of Jilin University, 126 Sendai Street, Changchun, Jilin 130033, China
| | - Dengyu Gao
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, 126 Sendai Street, Changchun, Jilin 130033, China
| | - Hongyang Xie
- Department of Gynecology, China-Japan Union Hospital of Jilin University, 126 Sendai Street, Changchun, Jilin 130033, China.
| | - Huafeng Geng
- Department of Gynecology, China-Japan Union Hospital of Jilin University, 126 Sendai Street, Changchun, Jilin 130033, China.
| |
Collapse
|
56
|
Zhao Y, Zhu M, Ling Y, Zhao Y, Lu X, Chu B, He Y, Wang H. A DNA Nanopatch-Bacteriophage System Targeting Streptococcus Gallolyticus for Inflammatory Bowel Disease Treatment and Colorectal Cancer Prevention. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2417334. [PMID: 39924920 DOI: 10.1002/adma.202417334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/28/2025] [Indexed: 02/11/2025]
Abstract
Persistent inflammation in inflammatory bowel disease (IBD) increases Streptococcus gallolyticus (Sg) colonization, increasing the risk of colorectal cancer progression via the Sg-activated cyclooxygenase-2 (COX-2) pathway and β-catenin upregulation. This study presents Sg-specific bacteriophages modified with DNA nanopatches (DNPs@P) designed to treat IBD and prevent Sg-induced malignancy. The DNPs are composed of DNA origami nanosheets and phage capture strands. The DNPs scavenge reactive oxygen species, enhancing the therapeutic efficacy of the phages while targeting and lysing pathogenic bacteria. Coating with an enteric polymer, DNPs@P ensures effective delivery in the gastrointestinal tract. These findings demonstrate significant restoration of colonic length, reduced inflammation, and improved gut microbiota diversity compared with current clinical treatments. Additionally, DNPs@P effectively prevents colonic tumourigenesis in mouse models. This approach presents a promising strategy for treating gastrointestinal diseases by remodeling the gut microenvironment, addressing a critical gap in current therapies.
Collapse
Affiliation(s)
- Yadan Zhao
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Mengna Zhu
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Yufan Ling
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, P. R. China
| | - Yingying Zhao
- Department of Ultrasound, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Xing Lu
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Binbin Chu
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Yao He
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
- Macao Translational Medicine Center, Macau University of Science and Technology, Taipa, Macau SAR, 999078, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, Macau SAR, 999078, China
| | - Houyu Wang
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| |
Collapse
|
57
|
Utpal BK, Sutradhar B, Zehravi M, Sweilam SH, Panigrahy UP, Urs D, Fatima AF, Nallasivan PK, Chhabra GS, Sayeed M, Alshehri MA, Rab SO, Khan SL, Emran TB. Polyphenols in wound healing: unlocking prospects with clinical applications. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2459-2485. [PMID: 39453503 DOI: 10.1007/s00210-024-03538-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024]
Abstract
Wound healing is a multifaceted, complex process that factors like aging, metabolic diseases, and infections may influence. The potentiality of polyphenols, natural compounds, has shown anti-inflammatory and antimicrobial properties in promoting wound healing and their potential applications in wound management. The studies reviewed indicate that polyphenols have multiple mechanisms that promote wound healing. This involves enhancing antioxidant defenses, reducing oxidative stress, modulating inflammatory responses, improving healing times, reducing infection rates, and enhancing tissue regeneration in clinical trials and in vivo and in vitro studies. Polyphenols have been proven to be effective in managing hard-to-heal wounds, especially in diabetic and elderly populations. Polyphenols have shown significant benefits in promoting angiogenesis and stimulating collagen synthesis. Polyphenol treatment has been demonstrated to have therapeutic effects in wound healing and chronic wound management. Their ability to regulate key healing processes makes them suitable for new wound care products and treatments. Future research should enhance formulations and delivery methods to optimize polyphenols' bioavailability and therapeutic efficacy in wound management approaches.
Collapse
Affiliation(s)
- Biswajit Kumar Utpal
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, 1207, Bangladesh.
| | - Baishakhi Sutradhar
- Department of Microbiology, Gono University (Bishwabidyalay), Nolam, Mirzanagar, Savar, Dhaka, 1344, Bangladesh
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, Buraydah, 51418, Saudi Arabia.
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, Cairo, 11829, Egypt
| | - Uttam Prasad Panigrahy
- Faculty of Pharmaceutical Science, Assam Down Town University, Gandhi Nagar, Sankar Madhab Path, Panikhaiti, Guwahati, Assam, 781026, India
| | - Deepadarshan Urs
- Inflammation Research Laboratory, Department of Studies & Research in Biochemistry, Mangalore University, Jnana Kaveri Post Graduate Campus, Kodagu, Karnataka, India
| | - Ayesha Farhath Fatima
- Department of Pharmaceutics, Anwarul Uloom College of Pharmacy, New Mallepally, Hyderabad, India
| | - P Kumar Nallasivan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Karpagam Academy of Higher Education, Pollachi Main Road, Eachanari, Coimbatore, Tamilnadu, India
| | - Gurmeet Singh Chhabra
- Department Pharmaceutical Chemistry, Indore Institute of Pharmacy, Opposite Indian Institute of Management Rau, Pithampur Road, Indore, Madhya Pradesh, India
| | - Mohammed Sayeed
- Department of Pharmacology, School of Pharmacy, Anurag University, Venkatapur, Ghatkesar, Hyderabad, Telangana, India
| | - Mohammed Ali Alshehri
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Sharuk L Khan
- Department of Pharmaceutical Chemistry, N.B.S. Institute of Pharmacy, Ausa, 413520, Maharashtra, India
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, 1207, Bangladesh.
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh.
| |
Collapse
|
58
|
Xia Y, Li X, Huang F, Wu Y, Liu J, Liu J. Design and advances in antioxidant hydrogels for ROS-induced oxidative disease. Acta Biomater 2025; 194:80-97. [PMID: 39900274 DOI: 10.1016/j.actbio.2025.01.057] [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: 09/30/2024] [Revised: 01/14/2025] [Accepted: 01/29/2025] [Indexed: 02/05/2025]
Abstract
Reactive oxygen species (ROS) play a crucial role in human physiological processes, but oxidative stress caused by excessive ROS may lead to a variety of acute and chronic diseases. Despite the development of various strategies and biomaterials, an efficiently and broadly applied method for treatment of ROS-induced oxidative disease remains a bottleneck. Aiming to improve the local oxidative stress environment, numerous bioactive hydrogels with antioxidant properties have emerged and are proven to quickly and continuously eliminate excessive ROS. To deeply understand the design principles and applications of antioxidant hydrogels is highly beneficial for designing antioxidant hydrogels for treatment of oxidative disease. This review provides a detailed summary of recent advances in design and applications of antioxidant hydrogels for various ROS-induced oxidative diseases. In this review, the kinds of antioxidant components in antioxidant hydrogels are outlined in detail. Additionally, the crosslinking methods and the biomedical applications of antioxidant hydrogels are widely summarized and discussed, especially focusing on their usage in different types of diseases and the attention given to the treatment of diseases such as skin wounds, myocardial infarction, and osteoarthritis. Finally, the future development direction of antioxidant hydrogel is further proposed. STATEMENT OF SIGNIFICANCE: Oxidative stress is a pivotal biochemical process that plays a critical role in cellular homeostasis. Excessive cellular oxidative stress triggers an inflammatory response, which is implicated in a spectrum of associated diseases. Given the critical need for managing oxidative stress, antioxidant therapies have become a vital focus in medical research. Hydrogels have garnered substantial interest among biomaterial scientists due to their hydrophilic nature and biocompatibility. The review delves into the realm of antioxidant hydrogels, encompassing the classification of antioxidant components, the synthesis and fabrication of hydrogels, and a comprehensive overview of the biological applications and challenges of these antioxidant hydrogels. Aiming to provide new perspectives for researchers in developing cutting-edge therapeutic approaches that leverage antioxidant hydrogels.
Collapse
Affiliation(s)
- Yi Xia
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Xinyi Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Fan Huang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Yuanhao Wu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| | - Jinjian Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| | - Jianfeng Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| |
Collapse
|
59
|
Liu R, Zhang J, Chen S, Xiao Y, Hu J, Zhou Z, Xie L. Intestinal mucosal immunity and type 1 diabetes: Non-negligible communication between gut and pancreas. Diabetes Obes Metab 2025; 27:1045-1064. [PMID: 39618164 PMCID: PMC11802406 DOI: 10.1111/dom.16101] [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: 07/28/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 02/08/2025]
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease characterized by T cell-mediated pancreatic β cell loss, resulting in lifelong absolute insulin deficiency and hyperglycaemia. Environmental factors are recognized as a key contributor to the development of T1D, with the gut serving as a primary interface for environmental stimuli. Recent studies have revealed that the alterations in the intestinal microenvironment profoundly affect host immune responses, contributing to the aetiology and pathogenesis of T1D. However, the dominant intestinal immune cells and the underlying mechanisms remain incompletely elucidated. In this review, we provide an overview of the possible mechanisms of the intestinal mucosal system that underpin the pathogenesis of T1D, shedding light on the roles of both non-classical and classical immune cells in T1D. Our goal is to gain insights into how modulating these immune components may hold potential implications for T1D prevention and provide novel perspectives for immune-mediated therapy.
Collapse
Affiliation(s)
- Ruonan Liu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and EndocrinologyThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Jing Zhang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and EndocrinologyThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Si Chen
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life ScienceHunan Normal UniversityChangshaChina
| | - Yang Xiao
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and EndocrinologyThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Jingyi Hu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and EndocrinologyThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and EndocrinologyThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Lingxiang Xie
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and EndocrinologyThe Second Xiangya Hospital of Central South UniversityChangshaChina
| |
Collapse
|
60
|
Pan Y, Zhao H, Huang W, Liu S, Qi Y, Huang Y. Metal-Protein Hybrid Materials: Unlocking New Frontiers in Biomedical Applications. Adv Healthc Mater 2025; 14:e2404405. [PMID: 39778029 DOI: 10.1002/adhm.202404405] [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: 11/07/2024] [Revised: 12/24/2024] [Indexed: 01/11/2025]
Abstract
Metal-protein hybrid materials represent a novel class of functional materials that exhibit exceptional physicochemical properties and tunable structures, rendering them remarkable applications in diverse fields, including materials engineering, biocatalysis, biosensing, and biomedicine. The design and development of multifunctional and biocompatible metal-protein hybrid materials have been the subject of extensive research and a key aspiration for practical applications in clinical settings. This review provides a comprehensive analysis of the design strategies, intrinsic properties, and biomedical applications of these hybrid materials, with a specific emphasis on their potential in cancer therapy, drug and vaccine delivery, antibacterial treatments, and tissue regeneration. Through rational design, stable metal-protein hybrid materials can be synthesized using straightforward methods, enabling them with therapeutic, delivery, immunomodulatory, and other desired functionalities. Finally, the review outlines the existing limitations and challenges associated with metal-protein hybrid materials and evaluates their potential for clinical translation, providing insights into their practical implementation within biomedical applications.
Collapse
Affiliation(s)
- Yong Pan
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Han Zhao
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Wenyong Huang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Siyang Liu
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Yanxin Qi
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| | - Yubin Huang
- Faculty of Chemistry, Northeast Normal University, Changchun, 130024, P.R. China
| |
Collapse
|
61
|
Wang R, Azad AK, Sheikh AM, Tabassum S, Zhang Y, Zhou X, Bhuiya J, Binte Abdullah F, Yano S, Ikeue T, Nagai A. Carboxylated Zn-phthalocyanine attenuates brain Aβ in AD model mouse. Brain Res 2025; 1850:149422. [PMID: 39722311 DOI: 10.1016/j.brainres.2024.149422] [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: 08/27/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
The deposition of aggregated amyloid β (Aβ) is considered as a key factor for Alzheimer's Disease (AD). Previously, we demonstrated that a carboxylated Zn-phthalocyanine (ZnPc) inhibits Aβ fibril formation, consequently protects neurons in culture. This study evaluated the effects of ZnPc on pathological changes in an AD mouse model (J20). Nine-month-old J20 mice received weekly intraperitoneal injection of ZnPc (2 and 4 mg/kg) for 12 weeks. Cognitive performance was assessed using Y-maze and open field tests. ZnPc levels in the tissues were evaluated using near-infrared microscopy and spectroscopy. ZnPc accumulated primarily in the liver and kidney. A considerable amount was also detected in brain tissue, where it co-localized with neurons, microglia, and extracellularly deposited Aβ. ZnPc treatment (2 mg/kg) significantly improved cognitive functions of J20 mice. Immunostaining results showed that Aβ was positive intracellularly in neurons, and extracellularly around the vessels and parenchyma in the cortex and hippocampus of PBS-treated J20 mice, which was significantly decreased in ZnPc-treated J20 mice in a dose-dependent manner. Nissl staining demonstrated that neuronal numbers were increased both in the cortex and hippocampus. GFAP-positive astrocytes and Iba-1 positive microglia were decreased by ZnPc treatment. Also, vessel numbers were increased in ZnPc-treated groups. In PBS-treated group, aquaporin 4 immunopositive area extended beyond STL-positive vessels into the parenchyma, which was confined primarily around the vessels in the ZnPc-treated group. Claudin 5 levels were increased in ZnPc-treated group. Therefore, ZnPc can decrease brain Aβ deposition in J20 mice, suggesting it as a potential therapeutic agent for AD.
Collapse
Affiliation(s)
- Ruochen Wang
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya-Cho, Izumo 693-8501, Japan; Department of Neurology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Abul Kalam Azad
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya-Cho, Izumo 693-8501, Japan
| | - Abdullah Md Sheikh
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan
| | - Shatera Tabassum
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan
| | - Yuchi Zhang
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya-Cho, Izumo 693-8501, Japan; Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Xiaojing Zhou
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya-Cho, Izumo 693-8501, Japan; Department of Neurology, Zhoushan Hospital, Zhoushan 316004, China
| | - Jubo Bhuiya
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya-Cho, Izumo 693-8501, Japan
| | - Fatema Binte Abdullah
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya-Cho, Izumo 693-8501, Japan
| | - Shozo Yano
- Department of Laboratory Medicine, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan
| | - Takahisa Ikeue
- Department of Chemistry, Graduate School of Science and Engineering, Shimane University, 1060 Nishikawatsu, Matsue 690-8504, Japan
| | - Atsushi Nagai
- Department of Neurology, Faculty of Medicine, Shimane University, 89-1 Enya-Cho, Izumo 693-8501, Japan; Department of Laboratory Medicine, Faculty of Medicine, Shimane University, Izumo 693-8501, Japan.
| |
Collapse
|
62
|
Sun R, Li S, Chen Z, Zheng K, Li W, Sun X, Yue W. Oral Antioxidant-Engineered Probiotics for the Treatment of Radiation-Induced Colitis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:10316-10327. [PMID: 39928516 DOI: 10.1021/acsami.4c17651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2025]
Abstract
Radiation colitis is an inflammatory response induced by damage to colonic tissue from radiation therapy that primarily affects patients undergoing abdominal or pelvic radiation treatments. In this study, we designed a selenium-loaded probiotic (EcN-Se@SA) encapsulated in sodium alginate gel for the oral treatment of radiation colitis. Selenium nanoparticles (Se NPs) exhibit antioxidant, anti-inflammatory, ferroptosis, and tissue repair-promoting properties. To enhance the therapeutic efficacy of Se NPs for radiation-induced colitis, we combined Se NPs with Escherichia coli Nissle 1917 and encapsulated them in sodium alginate hydrogel. The resulting EcN-Se@SA formulation not only withstands gastric acid and intestinal fluids but also remains in the colon and cecum for extended periods. Oral administration of EcN-Se@SA attenuates X-ray-induced colitis in mice through its antioxidant and anti-inflammatory effects, macrophage reprogramming, and inhibition of ferroptosis in histiocytes. This work introduces a promising therapeutic candidate for the treatment of radiation colitis.
Collapse
Affiliation(s)
- Rong Sun
- Department of Radiation Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 21000, People's Republic of China
| | - Shuaiwen Li
- Department of Chemistry, Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing 21000, People's Republic of China
| | - Zihui Chen
- Department of Chemistry, Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing 21000, People's Republic of China
| | - Kuo Zheng
- Department of Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 21000, People's Republic of China
| | - Weiqin Li
- Department of Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 21000, People's Republic of China
| | - Xiangdong Sun
- Department of Radiation Oncology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 21000, People's Republic of China
| | - Wanqing Yue
- Department of Chemistry, Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing 21000, People's Republic of China
- Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing 21000, People's Republic of China
| |
Collapse
|
63
|
Saleem W, Aslam A, Tariq M, Nauwynck H. Intestinal mucus: the unsung hero in the battle against viral gastroenteritis. Gut Pathog 2025; 17:11. [PMID: 39972475 PMCID: PMC11841282 DOI: 10.1186/s13099-025-00684-6] [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: 01/03/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025] Open
Abstract
Intestinal mucus plays a crucial role in defending against enteric infections by protecting the vulnerable intestinal epithelial cells both physically and through its various constituents. Despite this, numerous gastroenteritis-causing viruses, such as rotavirus, coronavirus, adenovirus, astrovirus, calicivirus, and enterovirus, continue to pose significant threats to humans and animals. While several studies have examined the interactions between these viruses and intestinal mucus, significant gaps remain in understanding the full protective potential of intestinal mucus against these pathogens. This review aims to elucidate the protective role of intestinal mucus in viral gastroenteritis. It begins with a comprehensive literature overview of (i) intestinal mucus, (ii) enteric viruses of medical and veterinary importance, and (iii) the known interactions between various enteric viruses and intestinal mucus. Following this, a case study is presented to highlight the age-dependent blocking effect of porcine intestinal mucus against transmissible gastroenteritis virus, a porcine coronavirus. Finally, the review discusses future investigation directions to further explore the potential of intestinal mucus as a defense mechanism against viral gastroenteritis to stimulate further research in this dynamic and critical area.
Collapse
Affiliation(s)
- Waqar Saleem
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Merelbeke, 9820, Belgium.
| | - Ateeqa Aslam
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Merelbeke, 9820, Belgium
| | - Mehlayl Tariq
- Laboratory of Biomedical Chemistry, Hirszfeld Institute of Immunology and Experimental Therapy, Wroclaw, 53-114, Poland
| | - Hans Nauwynck
- Laboratory of Virology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Merelbeke, 9820, Belgium
| |
Collapse
|
64
|
Hua D, Yang Q, Li X, Zhou X, Kang Y, Zhao Y, Wu D, Zhang Z, Li B, Wang X, Qi X, Chen Z, Cui G, Hong W. The combination of Clostridium butyricum and Akkermansia muciniphila mitigates DSS-induced colitis and attenuates colitis-associated tumorigenesis by modulating gut microbiota and reducing CD8 + T cells in mice. mSystems 2025; 10:e0156724. [PMID: 39840995 PMCID: PMC11834468 DOI: 10.1128/msystems.01567-24] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 12/23/2024] [Indexed: 01/23/2025] Open
Abstract
The gut microbiota is closely associated with inflammatory bowel disease (IBD) and colorectal cancer (CRC). Probiotics such as Clostridium butyricum (CB) or Akkermansia muciniphila (AKK) have the potential to treat inflammatory bowel disease (IBD) or colorectal cancer (CRC). However, research on the combined therapeutic effects and immunomodulatory mechanisms of CB and AKK in treating IBD or CRC has never been studied. This study evaluates the potential of co-administration of CB and AKK in treating DSS/AOM-induced IBD and colitis-associated CRC. Our results indicate that compared to mono-administration, the co-administration of CB and AKK not only significantly alleviates symptoms such as weight loss, colon shortening, and increased Disease Activity Index in IBD mice but also regulates the gut microbiota composition and effectively suppresses colonic inflammatory responses. In the colitis-associated CRC mice model, a combination of CB and AKK significantly alleviates weight loss and markedly reduces inflammatory infiltration of macrophages and cytotoxic T lymphocytes (CTLs) in the colon, thereby regulating anti-tumor immunity and inhibiting the occurrence of inflammation-induced CRC. In addition, we found that the combined probiotic therapy of CB and AKK can enhance the sensitivity of colitis-associated CRC mice to the immune checkpoint inhibitor anti-mouse PD-L1 (aPD-L1), significantly improving the anti-tumor efficacy of immunotherapy and the survival rate of colitis-associated CRC mice. Furthermore, fecal microbiota transplantation therapy showed that transplanting feces from CRC mice treated with the co-administration of CB and AKK into other CRC mice alleviated the tumor loads in the colon and significantly extended their survival rate. Our study suggests that the combined use of two probiotics, CB and AKK, can not only alleviate chronic intestinal inflammation but also inhibit the progression to CRC. This may be a natural and relatively safe method to support the gut microbiota and enhance the host's immunity against cancer. IMPORTANCE Our study suggests that the combined administration of CB and AKK probiotics, as opposed to a single probiotic strain, holds considerable promise in preventing the advancement of IBD to CRC. This synergistic effect is attributed to the ability of this probiotic combination to more effectively modulate the gut microbiota, curb inflammatory reactions, bolster the efficacy of immunotherapeutic approaches, and optimize treatment results via fecal microbiota transplantation.
Collapse
Affiliation(s)
- Dengxiong Hua
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & School/Hospital of Stomatology Guizhou Medical University, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Qin Yang
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Xiaowei Li
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Xuexue Zhou
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Yingqian Kang
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Yan Zhao
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & School/Hospital of Stomatology Guizhou Medical University, Guiyang, Guizhou, China
| | - Daoyan Wu
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Zhengrong Zhang
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Boyan Li
- School of Public Health, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xinxin Wang
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & School/Hospital of Stomatology Guizhou Medical University, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guiyang, Guizhou, China
| | - Zhenghong Chen
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & School/Hospital of Stomatology Guizhou Medical University, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guiyang, Guizhou, China
| | - Guzhen Cui
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & School/Hospital of Stomatology Guizhou Medical University, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guiyang, Guizhou, China
| | - Wei Hong
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & School/Hospital of Stomatology Guizhou Medical University, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guiyang, Guizhou, China
| |
Collapse
|
65
|
Huang C, Liu H, Luo J, Xu Z, Li J, Tian X. A prospective, single-blinded, non-inferiority, randomized controlled study comparing the effectiveness and safety of oral lactulose combined with carbohydrate-containing clear liquids versus 3-L polyethylene glycol electrolyte for colonoscopy bowel preparation. Eur J Med Res 2025; 30:105. [PMID: 39955567 PMCID: PMC11829565 DOI: 10.1186/s40001-025-02365-1] [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: 09/30/2024] [Accepted: 02/07/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Polyethylene glycol electrolyte solution (PEG-ELS) is the standard for bowel preparation but often suffers from poor patient compliance and tolerability due to its high-volume requirement. This prospective, single-blinded, non-inferiority, randomized control trial aims to investigate the efficacy and safety of a lactulose-based regimen as an alternative for bowel preparation. METHODS Two hundred nine patients were randomly allocated to receive either a combination regimen consisting of 133.4 g lactulose in 200 mL, 800 mL carbohydrate-containing clear liquid, 2L additional water, and 5 g simethicone (n = 104) or 3L PEG-ELS with 5 g simethicone (n = 105), both administered in a split-dose format. The primary outcome was the rate of adequate bowel preparation, measured by the Boston bowel preparation score (BBPS). Adequate bowel preparation was defined as a BBPS score of 2 or 3 in all colon segments. Secondary outcomes included the percentage of high-quality bowel preparation (defining as a total BBPS score of 8 or 9), polyp detection rate (defining as the percentage of procedures where at least one polyp was detected), willingness to repeat the bowel preparation, adverse events, and changes in blood glucose and electrolyte levels. RESULTS The rate of adequate bowel preparation (96.2% vs. 97.1%, p = 0.691), the percentage of high-quality preparation (62.5% vs. 66.7%, p = 0.529), average total BBPS scores (p = 0.607), polyp detection rates (66.3% vs. 77.1%, p = 0.083), and tolerability and acceptability outcomes, including satisfaction (p = 0.729) and willingness to repeat preparation (p = 0.744), were not statistically different between the two arms. Adverse events and changes in blood glucose and electrolytes showed no significant differences (all p > 0.05). CONCLUSION The combination of oral lactulose and carbohydrate-containing clear fluids was non-inferior to 3L PEG-ELS for bowel preparation adequacy and polyp detection, without statistically significant differences in terms of tolerability and safety.
Collapse
Affiliation(s)
- Chen Huang
- Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing, 400016, China
- Department of Clinical Nutrition, Chongqing General Hospital, Chongqing University, Chongqing, 400799, China
- Nutrition Innovation Platform Approved Jointly by Sichuan and Chongqing, Chongqing General Hospital, Chongqing University, Chongqing, 400799, China
| | - Hongli Liu
- Department of Gastroenterology, Chongqing General Hospital, Chongqing, 400799, China
| | - Jing Luo
- Department of Clinical Nutrition, Chongqing General Hospital, Chongqing University, Chongqing, 400799, China
| | - Zhe Xu
- Department of Nutrition, The Chenggong Hospital Affiliated to Xiamen University, Xiamen, 361005, China
| | - Jibin Li
- Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
| | - Xu Tian
- Chongqing Center for Evidence-Based Traditional Chinese Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400020, China.
| |
Collapse
|
66
|
Yao K, Zheng L, Chen W, Xie Y, Liao C, Zhou T. Characteristics, pathogenic and therapeutic role of gut microbiota in immunoglobulin A nephropathy. Front Immunol 2025; 16:1438683. [PMID: 39981255 PMCID: PMC11839611 DOI: 10.3389/fimmu.2025.1438683] [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: 05/26/2024] [Accepted: 01/21/2025] [Indexed: 02/22/2025] Open
Abstract
Immunoglobulin A nephropathy (IgAN) is the most prevalent glomerulonephritis in the world, and it is one of the leading causes of end-stage kidney disease. It is now believed that the pathogenesis of IgAN is the mesangial deposition of immune complex containing galactose-deficient IgA1, resulting in glomerular injury. Current treatments for IgAN include supportive care and immunosuppressive therapy. A growing number of studies found that the gut microbiota in IgAN was dysregulated. Gut microbiota may be involved in the development and progression of IgAN through three main aspects: destruction of intestinal barrier, changes in metabolites and abnormal mucosal immunity. Interestingly, therapies by modulating the gut microbiota, such as fecal microbiota transplantation, antibiotic treatment, probiotic treatment, Chinese herbal medicine Zhen Wu Tang treatment, gluten-free diet, and hydroxychloroquine treatment, can improve IgAN. In this review, the alteration of gut microbiota in IgAN, potential pathogenic roles of gut microbiota on IgAN and potential approaches to treat IgAN by modulating the gut microbiota are summarized.
Collapse
Affiliation(s)
| | | | | | | | | | - Tianbiao Zhou
- Department of Nephrology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
67
|
Wang Z, Lin Z, Mei X, Cai L, Lin KC, Rodríguez JF, Ye Z, Parraguez XS, Guajardo EM, García Luna PC, Zhang JYJ, Zhang YS. Engineered Living Systems Based on Gelatin: Design, Manufacturing, and Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2416260. [PMID: 39910847 DOI: 10.1002/adma.202416260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/26/2024] [Indexed: 02/07/2025]
Abstract
Engineered living systems (ELSs) represent purpose-driven assemblies of living components, encompassing cells, biomaterials, and active agents, intricately designed to fulfill diverse biomedical applications. Gelatin and its derivatives have been used extensively in ELSs owing to their mature translational pathways, favorable biological properties, and adjustable physicochemical characteristics. This review explores the intersection of gelatin and its derivatives with fabrication techniques, offering a comprehensive examination of their synergistic potential in creating ELSs for various applications in biomedicine. It offers a deep dive into gelatin, including its structures and production, sources, processing, and properties. Additionally, the review explores various fabrication techniques employing gelatin and its derivatives, including generic fabrication techniques, microfluidics, and various 3D printing methods. Furthermore, it discusses the applications of ELSs based on gelatin in regenerative engineering as well as in cell therapies, bioadhesives, biorobots, and biosensors. Future directions and challenges in gelatin fabrication are also examined, highlighting emerging trends and potential areas for improvements and innovations. In summary, this comprehensive review underscores the significance of gelatin-based ELSs in advancing biomedical engineering and lays the groundwork for guiding future research and developments within the field.
Collapse
Affiliation(s)
- Zhenwu Wang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Zeng Lin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Xuan Mei
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Ling Cai
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Ko-Chih Lin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Jimena Flores Rodríguez
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Zixin Ye
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Ximena Salazar Parraguez
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Emilio Mireles Guajardo
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Pedro Cortés García Luna
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Jun Yi Joey Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| |
Collapse
|
68
|
Oliyaei N, Zekri S, Iraji A, Oliyaei A, Tanideh R, Mussin NM, Tamadon A, Tanideh N. Health benefits of algae and marine-derived bioactive metabolites for modulating ulcerative colitis symptoms. J Funct Foods 2025; 125:106690. [DOI: 10.1016/j.jff.2025.106690] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
|
69
|
Edo GI, Mafe AN, Ali ABM, Akpoghelie PO, Yousif E, Apameio JI, Isoje EF, Igbuku UA, Garba Y, Essaghah AEA, Ahmed DS, Umar H, Ozsahin DU. Chitosan and its derivatives: A novel approach to gut microbiota modulation and immune system enhancement. Int J Biol Macromol 2025; 289:138633. [PMID: 39675606 DOI: 10.1016/j.ijbiomac.2024.138633] [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: 10/08/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024]
Abstract
Chitosan, a biopolymer derived from the deacetylation of chitin found in crustacean shells and certain fungi, has attracted considerable attention for its promising health benefits, particularly in gut microbiota maintenance and immune system modulation. This review critically examines chitosan's multifaceted role in supporting gut health and enhancing immunity, beginning with a comprehensive overview of its sources, chemical structure, and its dual function as a dietary supplement and biomaterial. Chitosan's prebiotic effects are highlighted, with a focus on its ability to selectively stimulate beneficial gut bacteria, such as Bifidobacteria and Lactobacillus, while enhancing gut barrier integrity and inhibiting the growth of pathogenic microorganisms. The review delves deeply into chitosan's immunomodulatory mechanisms, including its impact on antigen-presenting cells, cytokine profiles, and systemic immune responses. A detailed comparative analysis assesses chitosan's efficacy relative to other prebiotics and immunomodulatory agents, examining challenges related to bioavailability and metabolic activity. Beyond its role in gut health, this review explores chitosan's potential as a dual-action agent that not only supports gut microbiota but also fortifies immune resilience. It introduces emerging research on novel chitosan derivatives, such as chitooligosaccharides, and evaluates their enhanced bioactivity for functional food applications. Special attention is given to sustainability, with an exploration of alternative, plant-based sources of chitosan and their implications for both health and environmental stewardship. Also, the review identifies new research avenues, such as the growing interest in chitosan's role in the gut-brain axis and its potential mental health benefits through microbial interactions. By addressing these innovative areas, the review aims to shift the focus from basic health effects to chitosan's broader impact on public health. The findings encourage further exploration, particularly through human trials, and emphasize chitosan's untapped potential in revolutionizing health and disease management.
Collapse
Affiliation(s)
- Great Iruoghene Edo
- Department of Chemistry, Faculty of Science, Delta State University of Science and Technology, Ozoro, Nigeria; Department of Chemistry, College of Sciences, Al-Nahrain University, Baghdad, Iraq.
| | - Alice Njolke Mafe
- Department of Biological Sciences, Faculty of Science, Taraba State University Jalingo, Taraba State, Nigeria
| | - Ali B M Ali
- Department of Air Conditioning Engineering, Faculty of Engineering, Warith Al-Anbiyaa University, Karbala, Iraq
| | - Patrick Othuke Akpoghelie
- Department of Food Science and Technology, Faculty of Science, Delta State University of Science and Technology, Ozoro, Delta State, Nigeria
| | - Emad Yousif
- Department of Chemistry, College of Sciences, Al-Nahrain University, Baghdad, Iraq
| | - Jesse Innocent Apameio
- Department of Biological Sciences, Faculty of Science, Taraba State University Jalingo, Taraba State, Nigeria
| | - Endurance Fegor Isoje
- Department of Science Laboratory Technology (Biochemistry Option), Faculty of Science, Delta State University of Science and Technology, Ozoro, Nigeria
| | - Ufuoma Augustina Igbuku
- Department of Chemistry, Faculty of Science, Delta State University of Science and Technology, Ozoro, Nigeria
| | - Yasal Garba
- Department of Information Engineering, College of Information Engineering, Al-Nahrain University, Baghdad, Iraq
| | - Arthur Efeoghene Athan Essaghah
- Department of Urban and Regional Planning, Faculty of Environmental Sciences, Delta State University of Science and Technology, Ozoro, Nigeria
| | - Dina S Ahmed
- Department of Chemical Industries, Institute of Technology-Baghdad, Middle Technical University, Baghdad, Iraq
| | - Huzaifa Umar
- Operational Research Centre in Healthcare, Near East University, Nicosia, Cyprus
| | - Dilber Uzun Ozsahin
- Operational Research Centre in Healthcare, Near East University, Nicosia, Cyprus; Department of Medical Diagnostic Imaging, College of Health Sciences, University of Sharjah, P.O. Box 27272, United Arab Emirates; Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, United Arab Emirates
| |
Collapse
|
70
|
Bakshi J, Mishra KP. Sodium butyrate prevents lipopolysaccharide induced inflammation and restores the expression of tight junction protein in human epithelial Caco-2 cells. Cell Immunol 2025; 408:104912. [PMID: 39729961 DOI: 10.1016/j.cellimm.2024.104912] [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: 09/17/2024] [Revised: 12/13/2024] [Accepted: 12/20/2024] [Indexed: 12/29/2024]
Abstract
The gastrointestinal (GI) tract is susceptible to damage under high altitude hypoxic conditions, leading to gastrointestinal discomfort and intestinal barrier injury. Sodium butyrate, a short-chain fatty acid present as a metabolite in the gut, has emerged as a promising therapeutic agent due to its ability to act as an immunomodulatory agent and restore intestinal barrier integrity. This study aimed to explore the mechanism by which sodium butyrate exhibits anti inflammatory effect on intestinal epithelial cells. In vitro, Caco-2 epithelial cells and RAW 264.7 macrophages were used to investigate the protective role of sodium butyrate on Lipopolysaccharide (LPS) induced inflammation. Cell viability assays demonstrated that 1 mM (110.86 μg/mL) of sodium butyrate did not exhibit cytotoxicity on cells in vitro. Treatment with sodium butyrate suppressed reactive oxygen species levels and TNF-α production in LPS-stimulated macrophages, indicating its efficacy in mitigating inflammatory responses. Western blot analysis revealed that sodium butyrate attenuated the expression of iNOS in RAW 264.7 macrophage cells. Moreover, sodium butyrate also reversed the LPS induced over expression of HIF-1α, NLRP3, IL-1β as well as NF-kB in Caco-2 epithelial cells and also had a suppressive effect on IL-8 secretion after LPS stimulation. Immunocytochemistry demonstrated that sodium butyrate enhanced tight junction protein occludin expression in Caco-2 cells while also restoring the decreased permeability of the Caco-2 monolayer due to LPS. These results indicate that sodium butyrate may influence immune responses by suppressing inflammatory mediators and improving the integrity of the epithelial barrier. Understanding the intricate interactions between gut metabolites and host immune responses may help in the development of innovative therapeutic strategies to alleviate intestinal inflammation in high altitude environments.
Collapse
Affiliation(s)
- Jyotsana Bakshi
- Defence Institute of Physiology and Allied Sciences, Delhi 110054, India
| | - K P Mishra
- Defence Institute of Physiology and Allied Sciences, Delhi 110054, India.
| |
Collapse
|
71
|
Chen A, Gong Y, Wu S, Du Y, Liu Z, Jiang Y, Li J, Miao YB. Navigating a challenging path: precision disease treatment with tailored oral nano-armor-probiotics. J Nanobiotechnology 2025; 23:72. [PMID: 39893419 PMCID: PMC11786591 DOI: 10.1186/s12951-025-03141-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/19/2025] [Indexed: 02/04/2025] Open
Abstract
Oral probiotics have significant potential for preventing and treating many diseases. Yet, their efficacy is often hindered by challenges related to survival and colonization within the gastrointestinal tract. Nanoparticles emerge as a transformative solution, offering robust protection and enhancing the stability and bioavailability of these probiotics. This review explores the innovative application of nanoparticle-armored engineered probiotics for precise disease treatment, specifically addressing the physiological barriers associated with oral administration. A comprehensive evaluation of various nano-armor probiotics and encapsulation methods is provided, carefully analyzing their respective merits and limitations, alongside strategies to enhance probiotic survival and achieve targeted delivery and colonization within the gastrointestinal tract. Furthermore, the review explores the potential clinical applications of nano-armored probiotics in precision therapeutics, critically addressing safety and regulatory considerations, and proposing the innovative concept of 'probiotic intestinal colonization with nano armor' for brain-targeted therapies. Ultimately, this review aspires to guide the advancement of nano-armored probiotic therapies, driving progress in precision medicine and paving the way for groundbreaking treatment modalities.
Collapse
Affiliation(s)
- Anmei Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Ying Gong
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Shaoquan Wu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Ye Du
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Zhijun Liu
- Urology Institute of Shenzhen University, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, 518000, China
| | - Yuhong Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China.
| | - Jiahong Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China.
| | - Yang-Bao Miao
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China.
| |
Collapse
|
72
|
Lopes SA, Cardoso VMB, Roque-Borda CA, Chorilli M, Meneguin AB. Dual-action microparticles for ulcerative colitis: Cellulose nanofibers-enhanced delivery of 5-ASA and probiotics. Int J Biol Macromol 2025; 291:139060. [PMID: 39710030 DOI: 10.1016/j.ijbiomac.2024.139060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/10/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease initially treated with mesalazine (5-ASA). However, its effectiveness is limited by rapid absorption, low colonic concentration, and exacerbation of dysbiosis. Probiotics can mitigate dysbiosis if they survive the acidic conditions of the stomach. In this study, colon-specific microparticles (MPs) based on RS/P and reinforced with cellulose nanofibers (CNF) were used to co-encapsulate 5-ASA and L. rhamnosus. MPs prepared by spray-drying demonstrated a spherical shape, with sizes ranging from 1 to 10 μm, high encapsulation efficiency (up to 81.5 %), and maintenance of L. rhamnosus viability (5.74 log CFU/g of sample) even after 30 days of storage at 4 °C. Differential scanning calorimetry indicated a reduction in the melting peak of 5-ASA after microencapsulation, suggesting a decrease in its crystallinity. The samples also exhibited high mucoadhesivity, with the presence of CNF significantly increasing the speed of establishing interactions with mucin. In vitro release profiles showed lower release rates in acidic media, resulting in the majority of 5-ASA being released in intestinal and colonic media. These MPs represent a promising strategy for promoting specific release in the colon, minimizing side effects associated with conventional treatment, and potentially improving therapeutic efficacy in the context of UC.
Collapse
Affiliation(s)
- Sílvio André Lopes
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil.
| | - Vinicius Martinho Borges Cardoso
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil.
| | | | - Marlus Chorilli
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil.
| | - Andréia Bagliotti Meneguin
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil.
| |
Collapse
|
73
|
Abdel-Nasser A, Fathy HM, Badr AN, Barakat OS, Hathout AS. Chitosan nanoparticles loaded with Lactobacillus rhamnosus bioactive metabolites: Preparation, characterization, and antifungal activity. Heliyon 2025; 11:e41875. [PMID: 39885877 PMCID: PMC11780941 DOI: 10.1016/j.heliyon.2025.e41875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/29/2024] [Accepted: 01/09/2025] [Indexed: 02/01/2025] Open
Abstract
Aspergillus flavus is a severe danger to worldwide maize (Zea mays) cultivation, due to its extreme toxicity of aflatoxins produced by the fungi, and its ability to cause economic losses while also posing a health concern to humans and animals. Among the measures that may be considered for A. flavus control, applying coatings based on natural ingredients appears to be the most promising. The current work examines the antagonistic ability of Lactobacillus rhamnosus bioactive metabolites added to chitosan nanoparticles against A. flavus on maize kernels. The chitosan nanoparticles loaded with L. rhamnosus bioactive metabolites were characterized using the transmission electron microscope (TEM), zeta potential, size distribution, polydiversity index (PDI), pH, encapsulation efficiency and Fourier transform infrared spectroscopy (FTIR). The TEM revealed that the chitosan nanoparticles loaded with bioactive metabolites were spherical and smooth on the surface, and by increasing the concentration of bioactive metabolites added to the chitosan nanoparticles, the diameter of the chitosan nanoparticle grew. The zeta potential and size distribution values increased as the quantity of L. rhamnosus bioactive metabolites increased in the chitosan nanoparticles. The FTIR analysis indicated the presence of several functional groups, including alkynes, alkene, aliphatic primary amines, and other functional groups. The chitosan nanoparticles loaded with L. rhamnosus bioactive metabolites at a concentration of 7 mg/mL showed significant antifungal activity against A. flavus, reducing their growth in maize kernels by 89.42 % after 10 days of storage. They also reduced the percentage of germination inhibition rate and viability percentage. It could be concluded that adding L. rhamnosus bioactive metabolites to chitosan nanoparticles might have significant implications for food safety by using it in the industry to reduce the fungal contamination of grains.
Collapse
Affiliation(s)
- Aya Abdel-Nasser
- Food Toxicology and Contaminants Department, National Research Centre, Egypt
| | - Hayam M. Fathy
- Agricultural Microbiology Department, Faculty of Agriculture, Cairo University, Egypt
| | - Ahmed N. Badr
- Food Toxicology and Contaminants Department, National Research Centre, Egypt
| | - Olfat S. Barakat
- Agricultural Microbiology Department, Faculty of Agriculture, Cairo University, Egypt
| | - Amal S. Hathout
- Food Toxicology and Contaminants Department, National Research Centre, Egypt
| |
Collapse
|
74
|
Feng J, Wang Z, Li X, Bao C, Xiao Y. Facile Formulation of a Resveratrol-Mediated Multibond Network Hydrogel with Efficient Sustainable Antibacterial, Reactive Oxygen Species Scavenging, Pro-Angiogenesis, and Immunomodulation Activities for Accelerating Infected Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2025; 17:6144-6160. [PMID: 39814592 DOI: 10.1021/acsami.4c21260] [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: 01/18/2025]
Abstract
The management of chronic infected wounds remains a significant clinical challenge, largely due to the deficiency of optimal wound dressings with adequate mechanical strength, appropriate adhesiveness, and efficient sustainable antibacterial, reactive oxygen species (ROS) scavenging, pro-angiogenesis, and immunomodulation properties. To address such a dilemma, we employed a simple and facile strategy to utilize resveratrol (RSV) as a functional component to mediate hydrogel gelation in this study. The structure of this obtained hydrogel was supported by a multibond network, which not only endowed the resultant product with superior mechanical strength and moderate adhesiveness but also effectively prolonged the bioavailability of RSV. This strategy successfully integrated the entire system with sustainable antibacterial, ROS scavenging, pro-angiogenesis, and immunomodulation properties. Subsequent in vivo evidence has verified that this material was capable to accelerate the healing of chronic infected wounds. The underlying mechanism can be explained that this hydrogel is capable of propelling macrophage polarization from the M1 to M2 phenotype through modulating the PI3K/AKT signaling pathway to activate the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling as well as maintaining the mitochondrial membrane potential level in the normal state under excessive inflammatory and oxidative stimulus. In summary, this multifunctional hydrogel wound dressing provides a feasible way to promote the bioavailability of RSV, which is conducive for preparing a promising candidate for chronic infected wound healing. What is more important, it is also beneficial to reveal the correlative mechanisms to establish advanced therapeutic platform for targeting other complex infection microenvironment.
Collapse
Affiliation(s)
- Jing Feng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zifei Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiyu Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yu Xiao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
75
|
Farmakioti I, Stylianopoulou E, Siskos N, Karagianni E, Kandylas D, Vasileiou AR, Fragkiskatou F, Somalou P, Tsaroucha A, Ypsilantis P, Panas P, Kourkoutas Y, Skavdis G, Grigoriou ME. Enhancing Gut Microbiome and Metabolic Health in Mice Through Administration of Presumptive Probiotic Strain Lactiplantibacillus pentosus PE11. Nutrients 2025; 17:442. [PMID: 39940300 PMCID: PMC11820638 DOI: 10.3390/nu17030442] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/18/2025] [Accepted: 01/22/2025] [Indexed: 02/14/2025] Open
Abstract
Background: Over the past decade, probiotics have gained increasing recognition for their health benefits to the host. While most research has focused on the therapeutic effects of probiotics in the treatment of various diseases, recent years have seen a shift towards exploring their role in enhancing and supporting overall health. Methods: In this work, we have studied the effects of a novel potential probiotic strain, Lactiplantibacillus pentosus PE11, in healthy mice following a six-week dietary intervention. The assessment included monitoring the general health of the animals, biochemical analyses, profiling of the gut and fecal microbial communities, and gene expression analysis. Results: Our results showed that the administration of Lactiplantibacillus pentosus PE11 led to changes in the composition of the fecal microbiome, specifically an increase in the Firmicutes/Bacteroidetes ratio and in the relative abundance of the Lachnospiraceae, Ruminococcaceae, and Rikenellaceae families. Reduced Tnf expression and elevated Zo1 expression were also observed in the cecum, pointing to anti-inflammatory properties and improved intestinal barrier integrity. Additionally, a significant reduction in triglycerides and alanine aminotransferase levels-within physiological ranges-was observed, along with a trend toward decreased total cholesterol levels. Conclusions: These findings suggest that in healthy mice, Lactiplantibacillus pentosus PE11 has the potential to positively influence gut microbiome structure and metabolism, thereby supporting improved overall health.
Collapse
Affiliation(s)
- Ioanna Farmakioti
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - Electra Stylianopoulou
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - Nikistratos Siskos
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - Evangelia Karagianni
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - Dionysios Kandylas
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - Andreas Rafail Vasileiou
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - Fragkiski Fragkiskatou
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - Paraskevi Somalou
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - Alexandra Tsaroucha
- Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (A.T.); (P.Y.)
| | - Petros Ypsilantis
- Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (A.T.); (P.Y.)
| | | | - Yiannis Kourkoutas
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - George Skavdis
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| | - Maria E. Grigoriou
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (I.F.); (E.S.); (N.S.); (E.K.); (D.K.); (A.R.V.); (F.F.); (P.S.); (Y.K.); (G.S.)
| |
Collapse
|
76
|
Chen J, Liu M, Chen S, Chou CP, Liu H, Wu D, Liu Y. Engineered Therapeutic Bacteria with High-Yield Membrane Vesicle Production Inspired by Eukaryotic Membrane Curvature for Treating Inflammatory Bowel Disease. ACS NANO 2025; 19:2405-2418. [PMID: 39772458 DOI: 10.1021/acsnano.4c13069] [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: 01/11/2025]
Abstract
Bacterial membrane vesicles (BMVs) are emerging as powerful natural nanoparticles with transformative potential in medicine and industry. Despite their promise, scaling up BMV production and ensuring stable isolation and storage remain formidable challenges that limit their broader application. Inspired by eukaryotic mechanisms of membrane curvature, we engineered Escherichia coli DH5α to serve as a high-efficiency BMV factory. By fusing the ethanolamine utilization microcompartment shell protein EutS with the outer membrane via the ompA signal peptide, we induced dramatic membrane curvatures that drove enhanced vesiculation. Simultaneously, overexpression of fatty acyl reductase led to the production of amphiphilic fatty alcohols, further amplifying the BMV yield. Dynamic modulation of peptidoglycan hydrolase (PGase) expression facilitated efficient BMV release, resulting in a striking 149.11-fold increase in vesicle production. Notably, the high-yield BMVs from our engineered strain, without the need for purification, significantly bolstered innate immune responses and demonstrated therapeutic efficacy in treating inflammatory bowel disease (IBD). This study presents a strategy to overcome BMV production barriers, showcasing the therapeutic potential of engineered bacteria and BMVs for IBD treatment, while highlighting their potential applications in diverse biomedical fields.
Collapse
Affiliation(s)
- Jinjin Chen
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Mingkang Liu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Shiyi Chen
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - C Perry Chou
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Hongmei Liu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Decheng Wu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yilan Liu
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| |
Collapse
|
77
|
Cao MX, Qian ZY, Liang YJ, Liu QY, Wang HP, Meng Y, Wang YS, Wang Y. Layer-by-layer coated probiotics with tannic acid-Ca 2+ and casein phosphopeptide complexes for caries prevention and enamel remineralization. iScience 2025; 28:111579. [PMID: 39811639 PMCID: PMC11732097 DOI: 10.1016/j.isci.2024.111579] [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: 10/20/2024] [Revised: 11/05/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
Dental caries is a common disease resulting from tooth demineralization caused by bacterial plaque. Probiotics have shown great potential against caries by regulating the balance of oral flora. However, obstacles such as poor colonization and lysozyme sensitivity in oral cavity hinder their further application. In this study, an efficient layer-by-layer surface coating of tannic acid (TA)-Ca2+ and casein phosphopeptide (CPP) was applied to the probiotic Bifidobacterium infantis (BI) with potential anti-caries activity. Multi-functionalized probiotics thus prepared (called BI@TA-Ca2+@CPP) exhibited similar growth pattern, resistance to lysozyme and enhanced colonization potential. The ability of BI@TA-Ca2+@CPP in inhibiting cariogenic biofilm formation was improved. Moreover, BI@TA-Ca2+@CPP showed excellent remineralization efficacy. In a caries-induced rat model, BI@TA-Ca2+@CPP significantly prevented the occurrence and progression of tooth decay, meanwhile showing good biocompatibility. In summary, this study underlines the importance of probiotics coating in antibiofilm and remineralization activity as a promising strategy against dental caries.
Collapse
Affiliation(s)
- Ming-Xin Cao
- Department of Orthodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, P.R. China
- Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, P.R. China
| | - Zhan-Yin Qian
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin 300070, P.R. China
| | - Yan-Jie Liang
- Department of Orthodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, P.R. China
- Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, P.R. China
| | - Qi-Yao Liu
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin 300070, P.R. China
| | - Han-Ping Wang
- Department of Orthodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, P.R. China
- Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, P.R. China
| | - Yang Meng
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin 300070, P.R. China
| | - Yin-Song Wang
- Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, P.R. China
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, No.22 Qixiangtai Road, Heping District, Tianjin 300070, P.R. China
| | - Yue Wang
- Department of Orthodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, P.R. China
- Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, P.R. China
| |
Collapse
|
78
|
Xia R, Wu B, Jian Y, Li X, Zhang W, Zeng X, Chen S. Cordycepin improves liver fibrosis and the intestinal flora disturbance induced by 3,5-diethoxycarbonyl-1,4-dihydroxylidine in mice. Eur J Pharmacol 2025; 987:177172. [PMID: 39681281 DOI: 10.1016/j.ejphar.2024.177172] [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: 08/14/2024] [Revised: 11/22/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024]
Abstract
BACKGROUND AND AIMS Studies have shown that improving the intestinal flora can alleviate the progression of liver fibrosis. Cordycepin has shown potential anti-inflammatory and anti-fibrosis effects. In this study, we aimed to investigate the effects of cordycepin on liver fibrosis and how it affects the intestinal flora composition to determine a potentially effective therapeutic approach for liver fibrosis. EXPERIMENTAL PROCEDURE C57BL/6 mice were fed a special diet containing 3,5-diethoxycarbonyl-1,4-dihydroxylidine (DDC) to induce liver fibrosis. The histopathological changes in liver tissue and intestinal mucosa were determining via immunohistochemical staining. Serum transaminase levels were determined using biochemical test kits. Faecalibaculum samples were sequenced via 16S rRNA sequencing. RESULTS Cordycepin reduced DDC-induced liver collagen deposition, improved serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels, and reduced the levels of endothelial dysfunction markers vascular cell adhesion molecule 1 (VCAM) and thrombomodulin (TM). Our analysis of the intestinal flora composition showed that Dubosiella, Faecalibaculum, and Bifidobacterium were significantly increased in the cordycepin-treated group (P < 0.05). The Dubosiella level was significantly negatively correlated with TM and VCAM levels, and serum levels of ALT and AST (P < 0.05). After treatment with cordycepin, the microvilli length in the intestinal mucosa, the density of goblet cells, and the expressions of occludin and zonula occludens protein 1 (ZO-1) were significantly increased (P < 0.05). CONCLUSION We discovered that cordycepin improves liver fibrosis in vivo. We found that Dubosiella levels were considerably increased in the cordycepin-treated group and were significantly negatively correlated with liver sinusoidal endothelial damage.
Collapse
Affiliation(s)
- Ruiqi Xia
- Department of Gastroenterology, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China; Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bing Wu
- Department of Gastroenterology, Minhang Hospital, Fudan University, Shanghai, China
| | - Yourong Jian
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiangting Li
- Department of Traditional Chinese Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wen Zhang
- Department of Traditional Chinese Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoqing Zeng
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China; Evidence-based Medicine Centre, Fudan University, Shanghai, China.
| | - Shiyao Chen
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China; Evidence-based Medicine Centre, Fudan University, Shanghai, China.
| |
Collapse
|
79
|
Parvin N, Joo SW, Mandal TK. Biodegradable and Stimuli-Responsive Nanomaterials for Targeted Drug Delivery in Autoimmune Diseases. J Funct Biomater 2025; 16:24. [PMID: 39852580 PMCID: PMC11766201 DOI: 10.3390/jfb16010024] [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: 12/06/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/26/2025] Open
Abstract
Autoimmune diseases present complex therapeutic challenges due to their chronic nature, systemic impact, and requirement for precise immunomodulation to avoid adverse side effects. Recent advancements in biodegradable and stimuli-responsive nanomaterials have opened new avenues for targeted drug delivery systems capable of addressing these challenges. This review provides a comprehensive analysis of state-of-the-art biodegradable nanocarriers such as polymeric nanoparticles, liposomes, and hydrogels engineered for targeted delivery in autoimmune therapies. These nanomaterials are designed to degrade safely in the body while releasing therapeutic agents in response to specific stimuli, including pH, temperature, redox conditions, and enzymatic activity. By achieving localized and controlled release of anti-inflammatory and immunosuppressive agents, these systems minimize systemic toxicity and enhance therapeutic efficacy. We discuss the underlying mechanisms of stimuli-responsive nanomaterials, recent applications in treating diseases such as rheumatoid arthritis, multiple sclerosis, and inflammatory bowel disease, and the design considerations essential for clinical translation. Additionally, we address current challenges, including biocompatibility, scalability, and regulatory hurdles, as well as future directions for integrating advanced nanotechnology with personalized medicine in autoimmune treatment. This review highlights the transformative potential of biodegradable and stimuli-responsive nanomaterials, presenting them as a promising strategy to advance precision medicine and improve patient outcomes in autoimmune disease management.
Collapse
Affiliation(s)
| | - Sang Woo Joo
- School of Mechanical Engineering, School of Basic Science, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Tapas K. Mandal
- School of Mechanical Engineering, School of Basic Science, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| |
Collapse
|
80
|
Saban Güler M, Arslan S, Ağagündüz D, Cerqua I, Pagano E, Berni Canani R, Capasso R. Butyrate: A potential mediator of obesity and microbiome via different mechanisms of actions. Food Res Int 2025; 199:115420. [PMID: 39658184 DOI: 10.1016/j.foodres.2024.115420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 11/08/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024]
Abstract
Butyrate, a short-chain fatty acid, is a crucial product of gut microbial fermentation with significant implications for various metabolic and physiological processes. Dietary sources of butyrate are limited, primarily derived from the fermentation of dietary fibers by butyrate-producing gut bacteria. Butyrate exerts its effects primarily as a histone deacetylase (HDAC) inhibitor and through signaling pathways involving G protein-coupled receptors (GPCRs). Its diverse benefits include promoting gut health, enhancing energy metabolism, and potentially alleviating complications associated with obesity. However, the exact role of butyrate in obesity is still under investigation, with a limited number of human trials necessitating further research to determine its efficacy and safety profile. Moreover, butyrate impact on the gut-brain axis and its modulation of microbiome effect on behavior highlight its broader importance in regulating host physiology. A thorough understanding of the metabolic pathways and mechanisms of butyrate is essential for developing targeted interventions for metabolic disorders. Continued research is crucial to fully realize its therapeutic potential and optimize its clinical applications in human health. In summary, this review illuminates the multifaceted role of butyrate as a potential mediator of obesity and related metabolic changes.
Collapse
Affiliation(s)
- Meryem Saban Güler
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490 Ankara, Turkey
| | - Sabriye Arslan
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490 Ankara, Turkey
| | - Duygu Ağagündüz
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490 Ankara, Turkey.
| | - Ida Cerqua
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Ester Pagano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Roberto Berni Canani
- Department of Translational Medical Science and ImmunoNutritionLab at CEINGE Biotechnologies Research Center and Task Force for Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Raffaele Capasso
- Department of Agricultural Sciences, University of Naples Federico II, Portici, 80055 Naples, Italy.
| |
Collapse
|
81
|
Kollaparampil Kishanchand D, K A AK, Chandrababu K, Philips CA, Sivan U, Pulikaparambil Sasidharan BC. The Intricate Interplay: Microbial Metabolites and the Gut-Liver-Brain Axis in Parkinson's Disease. J Neurosci Res 2025; 103:e70016. [PMID: 39754366 DOI: 10.1002/jnr.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 11/21/2024] [Accepted: 12/23/2024] [Indexed: 01/06/2025]
Abstract
Parkinson's Disease (PD) is a neurodegenerative disorder marked by the depletion of dopaminergic neurons. Recent studies highlight the gut-liver-brain (GLB) axis and its role in PD pathogenesis. The GLB axis forms a dynamic network facilitating bidirectional communication between the gastrointestinal tract, liver, and central nervous system. Dysregulation within this axis, encompassing gut dysbiosis and microbial metabolites, is emerging as a critical factor influencing PD progression. Our understanding of PD was traditionally centered on neurodegenerative processes within the brain. However, examining PD through the lens of the GLB axis provides new insights. This review provides a comprehensive analysis of microbial metabolites, such as short-chain fatty acids (SCFAs), trimethylamine-N-oxide (TMAO), kynurenine, serotonin, bile acids, indoles, and dopamine, which are integral to PD pathogenesis by modulation of the GLB axis. Our extensive research included a comprehensive literature review and database searches utilizing resources such as gutMGene and gutMDisorder. These databases have been instrumental in identifying specific microbes and their metabolites, shedding light on the intricate relationship between the GLB axis and PD. This review consolidates existing knowledge and underscores the potential for targeted therapeutic interventions based on the GLB axis and its components, which offer new avenues for future PD research and treatment strategies. While the GLB axis is not a novel concept, this review is the first to focus specifically on its role in PD, highlighting the importance of integrating the liver and microbial metabolites as central players in the PD puzzle.
Collapse
Affiliation(s)
| | - Athira Krishnan K A
- Centre for Neuroscience, Department of Biotechnology, Cochin University of Science and Technology, Kochi, Kerala, India
| | - Krishnapriya Chandrababu
- Centre for Neuroscience, Department of Biotechnology, Cochin University of Science and Technology, Kochi, Kerala, India
| | - Cyriac Abby Philips
- Clinical and Translational Hepatology, The Liver Institute, Centre of Excellence in Gastrointestinal Sciences, Rajagiri Hospital, Aluva, Kerala, India
| | - Unnikrishnan Sivan
- Department of FSQA, FFE, Kerala University of Fisheries and Ocean Studies, Kochi, Kerala, India
| | - Baby Chakrapani Pulikaparambil Sasidharan
- Centre for Neuroscience, Department of Biotechnology, Cochin University of Science and Technology, Kochi, Kerala, India
- Centre for Excellence in Neurodegeneration and Brain Health, Kochi, Kerala, India
| |
Collapse
|
82
|
Wang Q, Jin L, Yang H, Yu L, Cao X, Mao Z. Bacteria/Nanozyme Composites: New Therapeutics for Disease Treatment. SMALL METHODS 2025; 9:e2400610. [PMID: 38923867 DOI: 10.1002/smtd.202400610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/11/2024] [Indexed: 06/28/2024]
Abstract
Bacterial therapy is recognized as a cost-effective treatment for several diseases. However, its development is hindered by limited functionality, weak inherent therapeutic effects, and vulnerability to harsh microenvironmental conditions, leading to suboptimal treatment activity. Enhancing bacterial activity and therapeutic outcomes emerges as a pivotal challenge. Nanozymes have garnered significant attention due to their enzyme-mimic activities and high stability. They enable bacteria to mimic the functions of gene-edited bacteria expressing the same functional enzymes, thereby improving bacterial activity and therapeutic efficacy. This review delineates the therapeutic mechanisms of bacteria and nanozymes, followed by a summary of strategies for preparing bacteria/nanozyme composites. Additionally, the synergistic effects of such composites in biomedical applications such as gastrointestinal diseases and tumors are highlighted. Finally, the challenges of bacteria/nanozyme composites are discussed and propose potential solutions. This study aims to provide valuable insights to offer theoretical guidance for the advancement of nanomaterial-assisted bacterial therapy.
Collapse
Affiliation(s)
- Qirui Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Lulu Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Huang Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Lisha Yu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Xinran Cao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
- State Key Laboratory of Transvascular Implantation Devices, Zhejiang, Hangzhou, 310009, China
| |
Collapse
|
83
|
An R, Zhou X, Zhang J, Lyu C, Wang D. Responses of intestinal microbiota to inulin was initial microbiota context dependent and affected by the supplementation dosage. Food Res Int 2025; 200:115498. [PMID: 39779139 DOI: 10.1016/j.foodres.2024.115498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 11/15/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025]
Abstract
Intestinal microbiota could respond to dietary fibres that are fermented by the gut microbiota, like prebiotics. Nevertheless, the dynamics of intestinal microbial community longitudinally after prebiotics intake, are still largely unknown. The current study unrevealed the successional process of intestinal microbial community after inulin supplementation, and the effect of supplementation dosage thereof, based on analysis of 16S rRNA gene sequences in C57BL/6 mice. We found that independent of supplementation dosage, intake of inulin could affect the intestinal microbial community within a day. Thereafter, the intestinal microbial community kept evolving until the last day of the supplementation (day 14) as a successional process, which was represented by the succession between intermediate and sluggish inulin responders. Remarkably, the successional process was initial microbial community context dependent and affected by the supplementation dosage. Specifically, the supplementation dosage affected the successional speed and the composition of the intermediate and sluggish inulin responders. Decreasing the relative abundance of previously identified intermediate responders, altered the successional process during inulin supplementation. Collectively, independent of supplementation dosage, the response of intestinal microbial community was rapid and the inulin induced temporal dynamics was represented by the succession between the intermediate and sluggish inulin responders. Nevertheless, the inulin induced successional process was initial microbial community context dependent and affected by the supplementation dosage. Findings of the current study would aid in the understanding of intestinal microbes' assembly during inulin supplementation and provide valuable support for dietary recommendations regarding to the use of prebiotics from the intestinal microbiota point of view.
Collapse
Affiliation(s)
- Ran An
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Xilong Zhou
- State Key Laboratory of Dairy Biotechnology, Dairy Research Institute, Bright Dairy and Food, Shanghai, China
| | - Jing Zhang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Chenang Lyu
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Dapeng Wang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
84
|
Wang J, Hou Y, Mu L, Yang M, Ai X. Gut microbiota contributes to the intestinal and extraintestinal immune homeostasis by balancing Th17/Treg cells. Int Immunopharmacol 2024; 143:113570. [PMID: 39547012 DOI: 10.1016/j.intimp.2024.113570] [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: 08/09/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024]
Abstract
Gut microbiota is generally considered to play an important role in host health due to its extensive immunomodulatory activities. Th17 and Treg cells are two important CD4+ T cell subsets involved in immune regulation, and their imbalance is closely tied to many immune diseases. Recently, abundant researches have highlighted the importance of gut microbiota in supporting intestinal and extraintestinal immunity through the balance of Th17 and Treg cells. Here, we presented a comprehensive review of these findings. This review first provided an overview of gut microbiota, along with Th17/Treg cell differentiation and cytokine production. Subsequently, the review summarized the regulatory effects of gut microbiota (in terms of species, components, and metabolites) on the Th17/Treg cell balance in the local intestines and extraintestinal organs, such as lung, liver, brain, kidney, and bone. Specifically, the Th17 and Treg cells that can be modulated by gut microbiota originate not only from the gut and extraintestinal organs, but also from peripheral blood and spleen. Then, the microbial therapeutics, including probiotics, prebiotics, postbiotics, and fecal microbiota transplantation (FMT), were also reviewed because of their therapeutic potentials in addressing intestinal and extraintestinal diseases via the Th17/Treg axis. Finally, the review discussed the clinical applications and future study prospects of microbial therapeutics by targeting the Th17/Treg cell balance. In conclusion, this review focused on elucidating the regulatory effects of gut microbiota in balancing Th17/Treg cells to maintain intestinal and extraintestinal immune homeostasis, contributing to the further development and promotion of microbial therapeutics.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Yaqin Hou
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Lifeng Mu
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Ming Yang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China.
| | - Xiaopeng Ai
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China.
| |
Collapse
|
85
|
Ciernikova S, Sevcikova A, Novisedlakova M, Mego M. Insights into the Relationship Between the Gut Microbiome and Immune Checkpoint Inhibitors in Solid Tumors. Cancers (Basel) 2024; 16:4271. [PMID: 39766170 PMCID: PMC11674129 DOI: 10.3390/cancers16244271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Immunotherapy with immune checkpoint inhibitors represents a revolutionary approach to the treatment of solid tumors, including malignant melanoma, lung cancer, and gastrointestinal malignancies. Anti-CTLA-4 and anti-PD-1/PDL-1 therapies provide prolonged survival for cancer patients, but their efficacy and safety are highly variable. This review focuses on the crucial role of the gut microbiome in modulating the efficacy and toxicity of immune checkpoint blockade. Studies suggest that the composition of the gut microbiome may influence the response to immunotherapy, with specific bacterial strains able to promote an anti-tumor immune response. On the other hand, dysbiosis may increase the risk of adverse effects, such as immune-mediated colitis. Interventions aimed at modulating the microbiome, including the use of probiotics, prebiotics, fecal microbial transplantation, or dietary modifications, represent promising strategies to increase treatment efficacy and reduce toxicity. The combination of immunotherapy with the microbiome-based strategy opens up new possibilities for personalized treatment. In addition, factors such as physical activity and nutritional supplementation may indirectly influence the gut ecosystem and consequently improve treatment outcomes in refractory patients, leading to enhanced patient responses and prolonged survival.
Collapse
Affiliation(s)
- Sona Ciernikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia;
| | - Aneta Sevcikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska Cesta 9, 845 05 Bratislava, Slovakia;
| | - Maria Novisedlakova
- Department of Oncology, Hospital Bory, Ivana Bukovčana 6118, 841 08 Bratislava, Slovakia;
| | - Michal Mego
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, Bratislava and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovakia;
| |
Collapse
|
86
|
Yi L, Han Y, Shen P, Du H, Guo X, Zhou Z, Xiao H. Dietary Porphyra tenera ameliorated dextran sodium sulfate-induced colitis in mice via modulating gut microbiota dysbiosis. Food Chem 2024; 461:140832. [PMID: 39181047 DOI: 10.1016/j.foodchem.2024.140832] [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: 05/27/2024] [Revised: 07/31/2024] [Accepted: 08/09/2024] [Indexed: 08/27/2024]
Abstract
Bioactive components from Porphyra tenera (PT) have been reported to confer various health benefits. The role of PT in inflammatory bowel disease (IBD) has not been fully investigated. This study aimed to explore the anti-inflammatory properties of PT on dextran sodium sulfate (DSS)-treated mice. PT supplementation attenuated the severity of colitis in DSS-treated mice, evidenced by the reduction of disease activity index (DAI), restoration of colonic histological damage and suppression of abnormal inflammatory response. Sequencing analysis indicated that intake of PT alleviated DSS-induced gut microbiota dysbiosis, accompanied by reversing the generation of short-chain fatty acids (SCFAs) and bile acids (BAs). Overall, our findings demonstrated that supplementation of PT attenuated the severity of intestinal inflammation and ameliorated gut microbiota dysbiosis in a murine colitis model, which provided a rationale for further application of edible seaweeds for preventing inflammation-related disorders in humans.
Collapse
Affiliation(s)
- Lingxiao Yi
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Yanhui Han
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Peiyi Shen
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Hengjun Du
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Xiaojing Guo
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Zhihao Zhou
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA.
| |
Collapse
|
87
|
Park JE, Park HY, Kim YS, Park M. The Role of Diet, Additives, and Antibiotics in Metabolic Endotoxemia and Chronic Diseases. Metabolites 2024; 14:704. [PMID: 39728485 DOI: 10.3390/metabo14120704] [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: 11/11/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Background/Objectives: Dietary patterns, including high-fat and high-carbohydrate diets (HFDs and HCDs), as well as non-dietary factors such as food additives and antibiotics, are strongly linked to metabolic endotoxemia, a critical driver of low-grade chronic inflammation. This review explores the mechanisms through which these factors impair intestinal permeability, disrupt gut microbial balance, and facilitate lipopolysaccharide (LPS) translocation into the bloodstream, contributing to metabolic disorders such as obesity, type 2 diabetes mellitus, and inflammatory bowel disease. Methods: The analysis integrates findings from recent studies on the effects of dietary components and gut microbiota interactions on intestinal barrier function and systemic inflammation. Focus is given to experimental designs assessing gut permeability using biochemical and histological methods, alongside microbiota profiling in both human and animal models. Results: HFDs and HCDs were shown to increase intestinal permeability and systemic LPS levels, inducing gut dysbiosis and compromising barrier integrity. The resulting endotoxemia promoted a state of chronic inflammation, disrupting metabolic regulation and contributing to the pathogenesis of various metabolic diseases. Food additives and antibiotics further exacerbated these effects by altering microbial composition and increasing gut permeability. Conclusions: Diet-induced alterations in gut microbiota and barrier dysfunction emerge as key mediators of metabolic endotoxemia and related disorders. Addressing dietary patterns and their impact on gut health is crucial for developing targeted interventions. Further research is warranted to standardize methodologies and elucidate mechanisms for translating these findings into clinical applications.
Collapse
Affiliation(s)
- Ji-Eun Park
- Food Functionality Research Division, Korea Food Research Institute, Jeonju 55365, Republic of Korea
- Department of Food Science and Technology, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Ho-Young Park
- Food Functionality Research Division, Korea Food Research Institute, Jeonju 55365, Republic of Korea
- Department of Food Biotechnology, Korea National University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Young-Soo Kim
- Department of Food Science and Technology, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Miri Park
- Food Functionality Research Division, Korea Food Research Institute, Jeonju 55365, Republic of Korea
| |
Collapse
|
88
|
He X, Liang Z, Huang Z, Wu Y, Liu J, Fu M, Liu J, Pi J, Zhang H. Effects of dietary paddy rice on the growth, serum biochemistry, intestinal development, microbiota, and metabolism of young laying ducks in a rice-duck-crayfish farming system. Poult Sci 2024; 103:104369. [PMID: 39427418 PMCID: PMC11536002 DOI: 10.1016/j.psj.2024.104369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 09/22/2024] [Accepted: 09/24/2024] [Indexed: 10/22/2024] Open
Abstract
This study investigated the effects of feeding paddy rice on the physiology, metabolism, and gut microbiota of ducks in a rice-duck-crayfish (RDC) system. A total of 540 ducks (20-days-old) were randomly divided into 3 groups with 3 replicates and 60 ducks per replicate. The 40-d experiment involved 3 diet treatments: a complete diet (CD), 50% paddy rice + 50% complete diet (RCD), and 100% paddy rice diet (RD). Results show that feeding paddy rice did not significantly affect duck growth, the final weight in the RD group was reduced by 5%, and the feed-to-gain ratio increased by 7% compared to the CD group. Additionally, compared with the CD group, the keel length, gizzard and proventriculus indices, and serum high-density lipoprotein levels increased (P < 0.05), and duodenal villus height and ileal crypt depth (P < 0.05) reduced in the RD group and ileal villus height decreased in the RCD group (P < 0.05). Compared with the CD group, the cecal abundance of Bacteroidota, Prevotellaceae_Ga6A1_group, and Prevotellaceae_NK3B31_group decreased in the RD group (P < 0.05) while the abundance of Firmicutes, Megamonas and Faecalibacterium increased (P < 0.05). Metabolome analysis revealed that serum citric acid increased (P < 0.05) in the RCD and RD groups, whereas cytidine, cytosine, and 4-aminobutyric acid decreased (P < 0.05) in the RD group. In conclusion, these preliminary results suggest that paddy rice supplementation under an RDC system had no significant effect on duck growth, but it did cause changes in intestinal morphology, microbiota, and serum metabolic profiles. However, it is important to note that the limited overall number of replicates in this study contributed to a certain degree of high variance. While the growth differences among groups were not statistically significant, the full replacement of paddy rice still poses potential performance losses. In practical applications, this finding provides a reference for the RDC system, but further validation through larger-scale trials is required.
Collapse
Affiliation(s)
- Xiaolong He
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China
| | - Zhenhua Liang
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Zeheng Huang
- College of Animal Science and Technology, Yangtze University, Jingzhou 434025, China
| | - Yan Wu
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Jia Liu
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China
| | - Ming Fu
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Jingbo Liu
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang 621010, China
| | - Jinsong Pi
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Hao Zhang
- Institute of Animal Husbandry and Veterinary Science, Hubei Academy of Agricultural Sciences, Wuhan 430064, China.
| |
Collapse
|
89
|
Liu Y, Huang K, Zhang Y, Li S, Song H, Guan X. Oat anthranilamides regulates high-fat diet-induced intestinal inflammation by the TLR4/NF-κb signalling pathway and gut microbiota. Int J Food Sci Nutr 2024; 75:786-799. [PMID: 39285614 DOI: 10.1080/09637486.2024.2401130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 08/22/2024] [Accepted: 09/01/2024] [Indexed: 11/26/2024]
Abstract
Oat anthranilamides have demonstrated antioxidant and anti-inflammatory effects; however, the precise mechanism of action remains unclear. This study investigated the impact of oat anthranilamide B (AVN B) on high-fat diet (HFD)-induced intestinal inflammation in mice and its underlying mechanisms. The results indicated that AVN B supplementation mitigated weight gain and reduced inflammatory and oxidative stress markers in serum, liver, and intestines. It improved intestinal barrier dysfunction by upregulating the expression levels of Occludin and MUC2 while simultaneously reducing intestinal inflammation by inhibiting the TLR4/NF-κB signalling pathway. Additionally, AVN B treatment improved gut microbiota composition. It increased the abundance of beneficial flora and the production of short-chain fatty acids (SCFAs), especially propionate and butyrate, associated with reduced production of pro-inflammatory factors and enhanced intestinal protection. The findings provide scientific evidence for the potential of AVN B as an anti-inflammatory agent.
Collapse
Affiliation(s)
- Yongyong Liu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, P.R. China
| | - Kai Huang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, P.R. China
| | - Yu Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, P.R. China
| | - Sen Li
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, P.R. China
| | - Hongdong Song
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, P.R. China
| | - Xiao Guan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, P.R. China
| |
Collapse
|
90
|
Ban SY, Yun DY, Yum SJ, Jeong HG, Park JT. Development of Saccharomyces cerevisiae accumulating excessive amount of glycogen and its effects on gut microbiota in a mouse model. Int J Biol Macromol 2024; 283:137589. [PMID: 39557260 DOI: 10.1016/j.ijbiomac.2024.137589] [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: 05/20/2024] [Revised: 11/01/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024]
Abstract
Saccharomyces cerevisiae accumulates glycogen, a hyperbranched glucose polymer with multiple bio-functionalities. In this study, mutants of S. cerevisiae that accumulate excessive amounts of glycogen were developed through UV mutagenesis. From over 30,000 mutants, the mutant strain CEY1, which exhibited the highest glycogen production, was selected using iodine vapor screening. The glycogen structures of wild type (WT) and CEY1 were analyzed and found to be relatively similar in molecular weight, hydrodynamic diameter, and side-chain distribution. The glycogen from CEY1 contained long branches (DP >12) 23.6 % greater than those in Escherichia coli TBP38. In addition, WT and CEY1 glycogen showed 32 %-34 % digestibility, which is significantly lower than E. coli glycogen. The glycogen content in dried CEY1 cells was increased to 21.7 % during laboratory-scale fed-batch fermentation. Glycogen with a homogeneous structure was accumulated to 17.5 % (w/w dried cell), and the total glucan content was increased by 33.2 % during large-scale fed-batch fermentation. In a mouse model, a diet containing 30 % CEY1 increased the production of butyrate and populations of beneficial bacteria, including Bacteroides and Parabacteroides. Therefore, glycogen from CEY1 exhibits a distinct structure from other polysaccharides, with notably slow and low digestibility, thereby indicating its potential application as a dietary supplement.
Collapse
Affiliation(s)
- So Young Ban
- Department of Food Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea; CARBOEXPERT Inc., Daejeon 34134, Republic of Korea
| | - Da-Young Yun
- Department of Food Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Su-Jin Yum
- Department of Food Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Hee-Gon Jeong
- Department of Food Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea.
| | - Jong-Tae Park
- Department of Food Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea; CARBOEXPERT Inc., Daejeon 34134, Republic of Korea.
| |
Collapse
|
91
|
Cao T, Tian D, Wang S, Pan Y, Xia Z, Chen W, Yang S, Zeng Q, Zhao Y, Zheng L, Li N, Lai Z, Luo Y, Shen Z. Microglial DBP Signaling Mediates Behavioral Abnormality Induced by Chronic Periodontitis in Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406269. [PMID: 39429161 PMCID: PMC11633467 DOI: 10.1002/advs.202406269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/10/2024] [Indexed: 10/22/2024]
Abstract
Several lines of evidence implicate that chronic periodontitis (CP) increases the risk of mental illnesses, such as anxiety and depression, yet, the associated molecular mechanism for this remains poorly defined. Here, it is reported that mice subjected to CP exhibited depression-like behaviors and hippocampal memory deficits, accompanied by synapse loss and neurogenesis impairment in the hippocampus. RNA microarray analysis disclosed that albumin D-site-binding protein (DBP) is identified as the most prominently upregulated target gene following CP, and in vivo and in vitro immunofluorescence methods showed that DBP is preferentially expressed in microglia but not neurons or astrocytes in the hippocampus. Interestingly, it is found that the expression of DBP is significantly increased in microglia after CP, and knockdown of microglial DBP ameliorated the behavioral abnormality, as well as reversed the synapse loss and hippocampal neurogenesis damage induced by CP. Furthermore, DBP knockdown improved the CP-induced hippocampal inflammation and microglial polarization. Collectively, these results indicate a critical role of DBP in orchestrating chronic periodontitis-related behavioral abnormality, hippocampal synapse loss and neurogenesis deficits, in which the microglial activation may be indispensably involved.
Collapse
Affiliation(s)
- Ting Cao
- Department of Children's StomatologyStomatological Hospital of Xiamen Medical College, Xiamen Key Laboratory of Stomatological Disease Diagnosis and TreatmentXiamen361003China
| | - Dan Tian
- Department of PharmacologySchool of PharmacyFujian Medical UniversityFuzhou350122China
| | - Si‐Ying Wang
- Department of PharmacologySchool of PharmacyFujian Medical UniversityFuzhou350122China
| | - Yue Pan
- Department of PharmacologySchool of PharmacyFujian Medical UniversityFuzhou350122China
| | - Zhi‐Xuan Xia
- Department of Pharmacology, School of Basic Medicine and Life ScienceHainan Medical UniversityHaikou571199China
| | - Wei‐Kai Chen
- Department of PharmacologySchool of PharmacyFujian Medical UniversityFuzhou350122China
| | - Shao‐Wei Yang
- Department of PharmacologySchool of PharmacyFujian Medical UniversityFuzhou350122China
| | - Qing‐Quan Zeng
- Department of PharmacologySchool of PharmacyFujian Medical UniversityFuzhou350122China
| | - Yue‐Ling Zhao
- Department of PharmacologySchool of PharmacyFujian Medical UniversityFuzhou350122China
| | - Ling Zheng
- Department of PharmacologySchool of PharmacyFujian Medical UniversityFuzhou350122China
| | - Ning Li
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of PharmacyFujian Medical UniversityFuzhou350122China
| | - Zhong‐Meng Lai
- Department of AnesthesiologyUnion Hospital, Fujian Medical UniversityFuzhou350001China
| | - Yi‐Xiao Luo
- Hunan Province People's HospitalThe First‐affiliated Hospital of Hunan Normal UniversityChangsha410002China
| | - Zu‐Cheng Shen
- Department of PharmacologySchool of PharmacyFujian Medical UniversityFuzhou350122China
| |
Collapse
|
92
|
Chen WJ, Chen YT, Ko JL, Chen JY, Zheng JY, Liao JW, Ou CC. Butyrate modulates gut microbiota and anti-inflammatory response in attenuating cisplatin-induced kidney injury. Biomed Pharmacother 2024; 181:117689. [PMID: 39581143 DOI: 10.1016/j.biopha.2024.117689] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024] Open
Abstract
In our previous research, we reported that administering probiotics Lactobacillus reuteri and Clostridium butyricum (LCs) before cisplatin treatment effectively modifies structures of the gut microbiota and restore ecological balance and significantly increases butyrate levels, a process closely associated with reducing cisplatin-induced nephrotoxicity. This study aims to investigate further whether the elevation of metabolite butyrate in the gut, promoted by probiotics LCs, can effectively mitigate the nephrotoxic effects of cisplatin and the progression of renal senescence in rats. Results show that butyrate administration significantly improved kidney function and decreased renal fibrosis in a dose-dependent manner compared to the cisplatin group. Its effects were associated with reductions in inflammatory responses, evidenced by decreased levels of key inflammatory markers, including KIM-1, MPO, NOX2, F4/80, and TGF-β1, alongside increased production of the anti-inflammatory cytokine IL-10. Furthermore, the butyrate intervention ameliorated cisplatin-induced gut microbiota dysbiosis, preserving the structure and diversity of healthy microbial communities. Specifically, we observed a decrease in the abundance of Escherichia_Shigella and Blautia, alongside an increase in the abundance of the butyrate-producing genus Roseburia. Notably, Escherichia_Shigella exhibited a positive correlation with the pro-inflammatory factor MPO, while displaying a negative correlation with the anti-inflammatory cytokine IL-10. Butyrate also attenuated the cisplatin-induced expression of senescence markers p21 and p16 in kidney tissue. It alleviated the cisplatin-increased senescence-associated beta-galactosidase activity and reactive oxygen species production in SV40 MES-13 cells. These results indicate that butyrate, derived from the gut microbiota, may exert a protective effect against cisplatin-induced kidney damage by regulating microbiota balance and anti-inflammatory effects.
Collapse
Affiliation(s)
- Wen-Jung Chen
- Department of Urology, Chung Shan Medical University Hospital, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yng-Tay Chen
- Graduate Institute of Food Safety, College of Agriculture and Natural Resources, National Chung Hsing University, Taichung, Taiwan; Department of Food Science and Biotechnology, College of Agriculture and Natural Resources, National Chung Hsing University, Taichung, Taiwan; Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Jiunn-Liang Ko
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Oncology and Chest Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Jian-Yuan Chen
- Department of Food Science and Biotechnology, College of Agriculture and Natural Resources, National Chung Hsing University, Taichung, Taiwan
| | - Jun-Yao Zheng
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Jiunn-Wang Liao
- Department of Food Science and Biotechnology, College of Agriculture and Natural Resources, National Chung Hsing University, Taichung, Taiwan; Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, Taichung, Taiwan.
| | - Chu-Chyn Ou
- Department of Nutrition, Chung Shan Medical University Hospital, Taichung, Taiwan; Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan.
| |
Collapse
|
93
|
Li L, Lin Y, Liu K, Huang R, Wen W, Huang Y, Liu M, Zhou C, Ding S, Luo B. Multiple-Effect Combined Hydrogels: "Temporal Regulation" Treatment of Osteosarcoma-Associated Bone Defects with Switchable Hyperthermia and Bioactive Agents. Adv Healthc Mater 2024; 13:e2402505. [PMID: 39233538 DOI: 10.1002/adhm.202402505] [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: 07/08/2024] [Revised: 08/25/2024] [Indexed: 09/06/2024]
Abstract
Achieving the clinically staged treatment of osteosarcoma-associated bone defects encounters the multiple challenges of promptly removing postoperative residual tumor cells and bacterial infection, followed by bone reconstruction. Herein, a core/shell hydrogel with multiple-effect combination is designed to first exert antitumor and antibacterial activities and then promote osteogenesis. Specifically, doxorubicin (DOX) is loaded by magnesium-iron-based layered double hydroxide (LDH) to prepare LDOX, which is introduced into a thermo-sensitive hydrogel to serve as an outer shell of the core/shell hydrogel, meanwhile, LDH-contained liquid crystal hydrogel, abbreviated as LCgel-L, is served as an inner core. At the early stage of treatment, the dissociation of the outer shell triggered by moderate hyperthermia led to the thermo-sensitive release of LDOX, which can be targeted for the release of DOX within tumor cells, thereby promptly removing postoperative residual tumor cells based on the synergistic effect of photothermal therapy (PTT) and DOX, and postoperative bacterial infection can also be effectively prevented by PTT simultaneously. More importantly, the dissociation of the outer shell prompted the full exposure of the inner core, which will exert osteogenic activity based on the synergy of liquid crystal hydrogel as well as LDH-induced mild hyperthermia and ion effects, thereby enabling "temporal regulation" treatment of osteosarcoma-associated bone defects. This study provides a valuable insight for the development of osteosarcoma-associated bone repair materials.
Collapse
Affiliation(s)
- Lin Li
- Biomaterial research laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, P. R. China
| | - Yating Lin
- Biomaterial research laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, P. R. China
| | - Kun Liu
- Biomaterial research laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, P. R. China
| | - Runshan Huang
- Biomaterial research laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, P. R. China
| | - Wei Wen
- Biomaterial research laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou, 510632, P. R. China
| | - Yadong Huang
- Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Guangzhou, 510632, P. R. China
| | - Mingxian Liu
- Biomaterial research laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou, 510632, P. R. China
| | - Changren Zhou
- Biomaterial research laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou, 510632, P. R. China
| | - Shan Ding
- Biomaterial research laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou, 510632, P. R. China
| | - Binghong Luo
- Biomaterial research laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou, 510632, P. R. China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou, 510632, P. R. China
| |
Collapse
|
94
|
Jing J, Li X, Liu S, Yu J, Wang K, Li Y, Wang J, Wan X. Molecular patterns of microbial and metabolic interactions in septic patients with persistent lymphopenia. Microb Pathog 2024; 197:107093. [PMID: 39486555 DOI: 10.1016/j.micpath.2024.107093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 09/30/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Persistent lymphopenia can be regarded as an important index of acquired immune dysfunction in sepsis. Whether the specific immune factor changes in septic patients with lymphopenia and the correlation to gut microbiota and metabolites remain unclear. METHODS This single-center prospective observation conducted lymphocyte subgroup analysis of blood samples and 16S rRNA gene amplicons sequencing and untargeted metabolomics analysis of fecal samples from 36 subjects with the persistent (≥3d) (n = 21) and non-persistent lymphopenia (<3d) (n = 15). RESULTS The persistent lymphopenia showed higher the 28d mortality and 90d mortality, while significantly lower CD3+T/LY, CD3+T cells, CD3+CD4+T cells, CD3+CD8+T cells, Th1 cells, Th2 cells, CD45RA + Treg cells. The 16S rRNA results showed that Staphylococcus, Peptostreptococcus, Bulleidia, Leuconostoc were significant enriched in the persistent lymphopenia. The metabolomics analysis showed that α-Ketoisovaleric acid was increased and 7-DHCA, α-MCA, β-MCA, HCA, LCA-3S, CA, UCA and Citramalic acid were decreased in the persistent lymphopenia. CONCLUSION In the process of interaction between host receptors and gut microbiota in patients with persistent lymphopenia sepsis, with a significant reduction in gut microbiota diversity and bile acid metabolites. That can affect various inflammatory pathways of gut immune cells, causing immune dysfunction in the body, which may be one of the main causes of death.
Collapse
Affiliation(s)
- Juanjuan Jing
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Liaoning, 116011, Dalian, China.
| | - Xiaonan Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Liaoning, 116011, Dalian, China.
| | - Shanshan Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Liaoning, 116011, Dalian, China.
| | - Jiawen Yu
- Department of Hematology, The First Affiliated Hospital of Dalian Medical University, Liaoning, 116011, Dalian, China.
| | - Kaixuan Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Liaoning, 116011, Dalian, China.
| | - Yi Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Liaoning, 116011, Dalian, China.
| | - Jia Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Liaoning, 116011, Dalian, China.
| | - Xianyao Wan
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Liaoning, 116011, Dalian, China.
| |
Collapse
|
95
|
Zhu M, Yang L, Kong S, Bai Y, Zhao B. Lacticaseibacillus rhamnosus LRa05 alleviates cyclophosphamide-induced immunosuppression and intestinal microbiota disorder in mice. J Food Sci 2024; 89:10003-10017. [PMID: 39592250 DOI: 10.1111/1750-3841.17538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/12/2024] [Accepted: 10/26/2024] [Indexed: 11/28/2024]
Abstract
Probiotics play a crucial role in regulating the gut microbiota and enhancing immune response. Oral administration of probiotics modulates intestinal microbiota composition and immune homeostasis. In this study, we investigated the immunoregulatory effect of Lacticaseibacillus rhamnosus LRa05 on cyclophosphamide (CTX)-induced immunosuppressive mice. The results showed that oral administration of LRa05 reduced weight loss, restored immune organ indices, and maintained the structural integrity of the intestinal tissue in CTX-treated mice. Moreover, oral administration of LRa05 exhibited immune-modulating properties by promoting the secretion of cytokines (tumor necrosis factor-α, interleukin-1β, interleukin-10, and secretory immunoglobulin A) in serum. Moreover, the analysis of 16S rRNA amplicon sequencing revealed that LRa05 increased gut microbiota diversity and regulated its composition. In detail, LRa05 intervention restored the Firmicutes/Bacteroidota ratio and significantly increased the relative abundance of Lachnospiraceae_NK4A136_group, Oscillibacter, Alloprevotella, Parasutterella, and Roseburia in immunocompromised mice. Conversely, the abundances of Helicobacter, Bacteroides, and unclassified_Desulfovibrionaceae were significantly decreased after administration of LRa05. Based on these findings, orally administered LRa05 could effectively maintain intestinal microbiota homeostasis and regulate immunity, suggesting the potential of L. rhamnosus LRa05 as a candidate probiotic strain in the application of dietary supplement. PRACTICAL APPLICATION: Supplement with L. rhamnosus LRa05 can improve immunity, regulate gut microbiota and promote body health.
Collapse
Affiliation(s)
- Mingming Zhu
- Wuhan Wecare Probiotic Research Institute, Wuhan, China
| | - Lvzhu Yang
- Wuhan Wecare Probiotic Research Institute, Wuhan, China
| | - Sufen Kong
- Wuhan Wecare Probiotic Research Institute, Wuhan, China
| | - Yuyuan Bai
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Bin Zhao
- Wuhan Wecare Probiotic Research Institute, Wuhan, China
- National Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
96
|
Chandwaskar R, Dalal R, Gupta S, Sharma A, Parashar D, Kashyap VK, Sohal JS, Tripathi SK. Dysregulation of T cell response in the pathogenesis of inflammatory bowel disease. Scand J Immunol 2024; 100:e13412. [PMID: 39394898 DOI: 10.1111/sji.13412] [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: 11/20/2023] [Revised: 08/26/2024] [Accepted: 09/17/2024] [Indexed: 10/14/2024]
Abstract
Inflammatory bowel disease (IBD), comprised of Crohn's disease (CD) and ulcerative colitis (UC), are gut inflammatory diseases that were earlier prevalent in the Western Hemisphere but now are on the rise in the East, with India standing second highest in the incidence rate in the world. Inflammation in IBD is a cause of dysregulated immune response, wherein helper T (Th) cell subsets and their cytokines play a major role in the pathogenesis of IBD. In addition, gut microbiota, environmental factors such as dietary factors and host genetics influence the outcome and severity of IBD. Dysregulation between effector and regulatory T cells drives gut inflammation, as effector T cells like Th1, Th17 and Th9 subsets Th cell lineages were found to be increased in IBD patients. In this review, we attempted to discuss the role of different Th cell subsets together with other T cells like CD8+ T cells, NKT and γδT cells in the outcome of gut inflammation in IBD. We also highlighted the potential therapeutic candidates for IBD.
Collapse
Affiliation(s)
- Rucha Chandwaskar
- Amity Institute of Microbial Technology (AIMT), Amity University Jaipur, Rajasthan, India
| | - Rajdeep Dalal
- Infection and Immunology Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, India
| | - Saurabh Gupta
- Centre for Vaccines and Diagnostic Research, GLA University, Mathura, Uttar Pradesh, India
| | - Aishwarya Sharma
- Sri Siddhartha Medical College and Research Center, Tumkur, Karnataka, India
| | - Deepak Parashar
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Vivek K Kashyap
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
| | - Jagdip Singh Sohal
- Centre for Vaccines and Diagnostic Research, GLA University, Mathura, Uttar Pradesh, India
| | - Subhash K Tripathi
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, Washington, USA
| |
Collapse
|
97
|
Panchal L, Arora S, Pramanik J, Batta K, Kumar A, Prajapati B. Probiotics: a promising intervention for osteoporosis prevention and management. Z NATURFORSCH C 2024; 79:405-411. [PMID: 38965037 DOI: 10.1515/znc-2024-0063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/20/2024] [Indexed: 07/06/2024]
Abstract
Osteoporosis (OP) is a systemic skeletal disease that is characterized by low bone mass and increased fracture risk. This article explores the potential of probiotics as an adjunctive approach for the prevention and management of OP. It has been well established that the gut microbiota (GM), a complex community of microbes, plays an important role in bone health. The gut dysbiosis is linked with a higher risk of OP. However, the consumption of probiotics in adequate amounts restores gut health thus improving bone health. Probiotics may influence bone metabolism through enhanced calcium absorption, reduced inflammation, and increased bone formation. The animal and human studies demonstrate the positive effects of probiotics on bone health parameters like reduced osteoclastogenesis, bone resorption markers, osteoblast, osteocyte apoptosis, and increased bone mineral density and expression of osteoprotegerin. The current evidence suggests that probiotics can be used as an adjunctive approach along with the existing therapies for the prevention and management of OP.
Collapse
Affiliation(s)
- Lakshay Panchal
- M.M Institute of Physiotherapy and Rehabilitation, Maharishi Markandeshwar University, Mullana, India
| | - Shivam Arora
- M.M Institute of Physiotherapy and Rehabilitation, Maharishi Markandeshwar University, Mullana, India
| | - Jhilam Pramanik
- Department of Food Technology, William Carrey University, Shillong, India
| | - Kajol Batta
- Department of Food Technology, ITM University, Gwalior, India
| | - Akash Kumar
- Department of Food Technology, SRM University, Delhi-NCR, Sonepat, India
- MMICT&BM (HM), Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, India
| | - Bhupendra Prajapati
- 79233 Shree S.K. Patel College of Pharmaceutical Education and Research, Ganpat University , Mehsana, India
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| |
Collapse
|
98
|
Rivera K, Gonzalez L, Bravo L, Manjarres L, Andia ME. The Gut-Heart Axis: Molecular Perspectives and Implications for Myocardial Infarction. Int J Mol Sci 2024; 25:12465. [PMID: 39596530 PMCID: PMC11595032 DOI: 10.3390/ijms252212465] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 11/28/2024] Open
Abstract
Myocardial infarction (MI) remains the leading cause of death globally, imposing a significant burden on healthcare systems and patients. The gut-heart axis, a bidirectional network connecting gut health to cardiovascular outcomes, has recently emerged as a critical factor in MI pathophysiology. Disruptions in this axis, including gut dysbiosis and compromised intestinal barrier integrity, lead to systemic inflammation driven by gut-derived metabolites like lipopolysaccharides (LPSs) and trimethylamine N-oxide (TMAO), both of which exacerbate MI progression. In contrast, metabolites such as short-chain fatty acids (SCFAs) from a balanced microbiota exhibit protective effects against cardiac damage. This review examines the molecular mediators of the gut-heart axis, considering the role of factors like sex-specific hormones, aging, diet, physical activity, and alcohol consumption on gut health and MI outcomes. Additionally, we highlight therapeutic approaches, including dietary interventions, personalized probiotics, and exercise regimens. Addressing the gut-heart axis holds promise for reducing MI risk and improving recovery, positioning it as a novel target in cardiovascular therapy.
Collapse
Affiliation(s)
- Katherine Rivera
- Doctoral Program in Medical Sciences, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago de Chile 8331010, Chile;
- Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile, Santiago de Chile 7820436, Chile
- Millennium Institute for Intelligent Healthcare Engineering iHEALTH, Santiago de Chile 7820436, Chile
| | - Leticia Gonzalez
- Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile, Santiago de Chile 7820436, Chile
- Millennium Institute for Intelligent Healthcare Engineering iHEALTH, Santiago de Chile 7820436, Chile
| | - Liena Bravo
- Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile, Santiago de Chile 7820436, Chile
- Millennium Institute for Intelligent Healthcare Engineering iHEALTH, Santiago de Chile 7820436, Chile
| | - Laura Manjarres
- Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile, Santiago de Chile 7820436, Chile
- Millennium Institute for Intelligent Healthcare Engineering iHEALTH, Santiago de Chile 7820436, Chile
| | - Marcelo E. Andia
- Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile, Santiago de Chile 7820436, Chile
- Millennium Institute for Intelligent Healthcare Engineering iHEALTH, Santiago de Chile 7820436, Chile
| |
Collapse
|
99
|
Zhu F, Wen Q, Hu Y, Gong J, Zhang X, Huang C, Zhou H, Chen L, Yu L. Chondroitin sulfate sponge scaffold for slow-release Mg 2+/Cu 2+ in diabetic wound management: Hemostasis, effusion absorption, and healing. Int J Biol Macromol 2024; 282:137561. [PMID: 39537068 DOI: 10.1016/j.ijbiomac.2024.137561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/03/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
The management of diabetic wounds presents significant challenges due to persistent inflammation, microenvironmental disruptions, and impaired angiogenesis. To address these issues, this study developed a multifunctional chondroitin sulfate sponge (CSP@Cu-Mg) with anti-inflammatory properties, hemostatic effects, effusion absorption, and enhanced healing promotion. Through ion crosslinking, MgO and CuO were incorporated into the interpenetrating network structure of chondroitin sulfate and acellular dermal matrix, resulting in a sponge with impressive liquid absorption capacity (3450 %) and porosity (83 %). This sponge enabled sustained release of Mg2+/Cu2+ ions, with approximately 40 % cumulative release over 7 days. This release helped reduce inflammation, promote the proliferation and migration of skin repair-related cells, and stimulate angiogenesis. In vivo studies demonstrated that the CSP@Cu-Mg sponge significantly improved diabetic wound healing by modulating inflammation and accelerating collagen deposition, angiogenesis, and re-epithelialization. This extracellular matrix sponge, which synergistically releases Mg2+/Cu2+, presents a promising strategy for comprehensive diabetic wound management with substantial clinical implications.
Collapse
Affiliation(s)
- Fengyi Zhu
- YunFu People's Hospital, Central Laboratory of YunFu People's Hospital, No. 120 Huanshi East Road, Yuncheng District, Yunfu City 527399, PR China; School of Basic Medicine, Jinzhou Medical University, No.40, Section 4, Road Songpo, Jinzhou, Liaoning 121001, PR China
| | - Qiulan Wen
- Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangdong 510515, PR China
| | - Yuting Hu
- Department of Anesthesiology, Shenzhen Maternal and Child Health Hospital, 2004 Hongli Road, Futian District, Shenzhen City, Guangdong 518031, PR China
| | - Jun Gong
- YunFu People's Hospital, Central Laboratory of YunFu People's Hospital, No. 120 Huanshi East Road, Yuncheng District, Yunfu City 527399, PR China
| | - Xibing Zhang
- YunFu People's Hospital, Central Laboratory of YunFu People's Hospital, No. 120 Huanshi East Road, Yuncheng District, Yunfu City 527399, PR China
| | - Chaoyang Huang
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangdong 510515, PR China.
| | - Hai Zhou
- YunFu People's Hospital, Central Laboratory of YunFu People's Hospital, No. 120 Huanshi East Road, Yuncheng District, Yunfu City 527399, PR China.
| | - Lianglong Chen
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangdong 510515, PR China.
| | - Li Yu
- School of Basic Medicine, Jinzhou Medical University, No.40, Section 4, Road Songpo, Jinzhou, Liaoning 121001, PR China.
| |
Collapse
|
100
|
González-Velázquez G, Aguirre-Garrido JF, Oros-Pantoja R, Salinas-Velarde ID, Contreras I, Estrada JA, Soto-Piña AE. Supplementation with inulin reverses cognitive flexibility alterations and modulates the gut microbiota in high-fat-fed mice. Front Behav Neurosci 2024; 18:1445154. [PMID: 39568732 PMCID: PMC11577567 DOI: 10.3389/fnbeh.2024.1445154] [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: 06/06/2024] [Accepted: 10/16/2024] [Indexed: 11/22/2024] Open
Abstract
Introduction Alterations in cognitive performance are associated with inadequate nutritional states and diet composition. Prebiotics, such as inulin, are substances that can modulate the gut microbiome and, consequently, brain function by producing metabolites such as short-chain fatty acids (SCFAs). This study aimed to evaluate the effect of supplementation with inulin on cognitive flexibility, body composition, and gut microbiota in a murine model exposed to a high-fat (HF) diet. Methods CD1 mice were divided into five groups: control fed a standard diet (C), high-fat diet (HF), inulin (I), high-fat diet with inulin (HFI), and manipulation control (M). Dietary supplementation was administered for 6 weeks. Cognitive flexibility was assessed using the Attentional Set-Shifting Test (AST). In addition, body composition was measured via electrical bioimpedance and adipose tissue compartments of each mouse were removed and weighed. Finally, gut microbiota metataxonomic was analyzed through metataxonomic bacterial 16S rRNA sequencing. Results We observed that HF group required more AST trials than the C, HFI, and I groups in the compound discrimination (CD) and extra-dimensional (ED) stages. Notably, the HFI group required fewer trials than the HF group in the ED stage (p = 0.0187). No significant differences in overall body composition were observed between the groups. However, the percentage of gonadal and peritoneal adipose tissue was significantly higher in the HF and I groups compared to the C group. Statistically significant differences in alpha diversity for gut microbiota were observed using the Shannon, Simpson, and Chao1 indices. The I group showed a decrease in bacterial diversity compared to the HF group. While no differences were observed between groups in the phyla Bacillota and Bacteroidotes, Clostridium bacteria represented a lower proportion of sequences in the I group compared to the C group. Additionally, Lactobacillus represented a lower proportion of sequences in the HF group compared to the C and I groups. Discussion These findings suggest that supplementation with inulin could be a useful approach to mitigate the negative effects of an HF diet on cognitive flexibility and modulate gut microbiota composition.
Collapse
Affiliation(s)
| | - José Félix Aguirre-Garrido
- Departamento de Ciencias Ambientales, Universidad Autónoma Metropolitana-Lerma, Lerma, Estado de México, Mexico
| | - Rigoberto Oros-Pantoja
- Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico
| | | | - Irazú Contreras
- Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico
| | - José Antonio Estrada
- Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Estado de México, Mexico
| | | |
Collapse
|