1
|
Hu X, Wang R, Kille P, Maret W, Hogstrand C. Zinc amino acid chelate and the Aryl Hydrocarbon Receptor (AHR) cooperate in improving the barrier function of a Caco-2 cell intestinal epithelium. J Nutr Biochem 2025; 141:109909. [PMID: 40154643 DOI: 10.1016/j.jnutbio.2025.109909] [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: 08/12/2024] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
Zinc and several physiologically relevant ligands of the aryl hydrocarbon receptor (AHR) are nutrients that promote intestinal barrier function. We have identified that AHR activation upregulates the expression of zinc importers in the intestinal epithelium to increase intracellular zinc concentrations, which leads to improved epithelial barrier function. Here, we investigated if an amino acid chelate of zinc, in cooperation with AHR activation, can improve the barrier function of a differentiated Caco-2 cell epithelium. Functional assays of the Caco-2 cell epithelium demonstrate that both ZnSO4 and a lysine and glutamic acid chelate of Zn, in combination with the physiological AHR agonist 6-formylindolo[3,2-b]carbazole (FICZ), increase expression of tight junction proteins at the mRNA and protein levels. FICZ increases uptake of zinc into the epithelium in the presence of ZnSO4 or the amino acid Zn chelate in the medium to equal extents. We conclude that the lysine and glutamic acid chelate of Zn is as efficacious as ZnSO4 in reducing permeability of the Caco-2 cell epithelium in the presence of FICZ. The results suggest that dietary supplementation with bioavailable forms of zinc together with nutritional AHR agonists may be beneficial in improving gut barrier function and help prevent inflammatory bowel disease (IBD).
Collapse
Affiliation(s)
- Xiuchuan Hu
- Department of Nutritional Sciences, School of Life Course and Population Sciences, King's College London, London, UK
| | - Rui Wang
- Department of Nutritional Sciences, School of Life Course and Population Sciences, King's College London, London, UK
| | - Peter Kille
- School of Biosciences, Cardiff University, Cardiff, UK
| | - Wolfgang Maret
- Department of Nutritional Sciences, School of Life Course and Population Sciences, King's College London, London, UK
| | - Christer Hogstrand
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK.
| |
Collapse
|
2
|
Zhou R, Cui Y, Wang W, Qu Y, Zhang G, Wang X, Sun Y, Li J. The deubiquitinase USP2 preserves intestinal barrier through the TRAF6/NF-κB/MLCK/MLC signaling in ulcerative colitis. Int Immunopharmacol 2025; 162:115099. [PMID: 40540897 DOI: 10.1016/j.intimp.2025.115099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 06/12/2025] [Accepted: 06/12/2025] [Indexed: 06/22/2025]
Abstract
Disruption of the physical barrier of intestinal epithelial cells is a hallmark of ulcerative colitis (UC). Deubiquitinating enzymes (DUBs), which modify the stability, localization, or activity of substrates by removing ubiquitin chains, have been involved in the development of UC. However, the biological role of DUBs in intestinal epithelial cells (IECs) has yet to be extensively investigated. In this work, we revealed that USP2 as the most differentially expressed DUB in the colon of mice with dextran sodium sulfate (DSS)-induced colitis and was downregulated mainly in the intestinal epithelium. Notably, the expression of USP2 was correlated with the levels of tight junction (TJ) proteins. Furthermore we showed that USP2 expression was reduced in the intestine of mice with acute colitis induced by DSS, as well as in IECs stimulated with LPS. Overexpression of USP2 restored the LPS-induced the reduction of TJPs. Mechanistically, USP2 attenuates the activation of NF-κB and the phosphorylation of the myosin light chain, possibly by reducing the K63-linked polyubiquitin chain on the tumour necrosis factor receptor-associated factor 6 (TRAF6). In vivo, colonic-specific overexpression of USP2 in mice ameliorated DSS-induced barrier damage. Together, our study proposed USP2 as a new deubiquitinating enzyme molecule involved in the course of UC regulating the function of IECs during the progression of UC.
Collapse
Affiliation(s)
- Ruochen Zhou
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuanyuan Cui
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Weihao Wang
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China; School of Pharmacy, Shandong Second Medical University, Weifang 261053, Shandong, China
| | - Yidan Qu
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China; Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong 264000, China
| | - Gaojie Zhang
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China; College of Pharmacy, Henan University, Kaifeng 475004, China
| | - Xue Wang
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| | - Yili Sun
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Jia Li
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Pharmacy, Shandong Second Medical University, Weifang 261053, Shandong, China.
| |
Collapse
|
3
|
Li Y, Yang D, Zhao H, Dou L, Chen Q, Cheng Y, Hu B, Tang Y, Duan Y, Guo C, Sakandar A, Li D. The pasteurized Weissella cibaria alleviates sepsis-induced acute lung injury by modulation of intestinal mucus barrier and gut microbiota. J Transl Med 2025; 23:661. [PMID: 40528192 DOI: 10.1186/s12967-025-06674-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 05/30/2025] [Indexed: 06/20/2025] Open
Abstract
BACKGROUND Dysbiosis of intestinal microecology caused by sepsis plays a crucial role in the onset and progression of sepsis-induced acute lung injury (SALI). As a postbiotic type, inactivated probiotic bacteria can regulate the gut microbiome. Pasteurized bacteria are considered safer than live bacteria in immune dysregulation disorders. Weissella cibaria (W. cibaria) is considered a candidate probiotic with certain beneficial functions. However, whether inactivated W. cibaria can alleviate SALI and the underlying mechanisms remain unclear. This study aimed to investigate whether inactivated W. cibaria can regulate intestinal mucosal barrier function and gut microbiota, thereby improving SALI. METHODS Following gavage of pasteurized W. cibaria in septic mice, lung tissue damage and inflammation levels were assessed. Circulating LPS levels and inflammatory cytokine concentrations in the blood were measured. Additionally, colonic tissue inflammation, intestinal mucosal barrier integrity, and alterations in the gut microbiota were evaluated. RESULT Pasteurized W. cibaria increases survival rates in SALI mice and improves pathological damage and cell apoptosis in lung tissue. Pasteurized W. cibaria also reduces the lung inflammatory response in septic mice by lowering pro-inflammatory cytokine levels and increasing anti-inflammatory cytokine levels. Pasteurized W. cibaria appears to exert its effects by improving the intestinal mucosal barrier and reversing gut microbiota dysbiosis caused by sepsis. Specifically, pasteurized W. cibaria alleviates intestinal barrier damage and inflammation in SALI mice, enhancing the integrity of the intestinal mucosal barrier. Additionally, pasteurized W. cibaria increases the abundance of anti-inflammatory bacteria such as Muribaculaceae. Pasteurized W. cibaria also decreases the levels of LPS-producing bacteria, including Escherichia-Shigella and Helicobacter, leading to significant attenuation in metabolic endotoxemia, which in turn alleviates excessive lung inflammation in septic mice. CONCLUSIONS Pasteurized W. cibaria has the potential to act as a postbiotic agent, improving sepsis-induced gut microbiota dysbiosis and acute lung injury, and providing a novel strategy for treating SALI.
Collapse
Affiliation(s)
- Yuanzhe Li
- Department of Pediatrics, The First Affiliated Hospital of Dalian Medical University, Dalian116011, China
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Debin Yang
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Huan Zhao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Liping Dou
- Department of Pediatrics, The First Affiliated Hospital of Dalian Medical University, Dalian116011, China
| | - Qian Chen
- Department of Pediatrics, The First Affiliated Hospital of Dalian Medical University, Dalian116011, China
| | - Yibing Cheng
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Bo Hu
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Yu Tang
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Yongtao Duan
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Caili Guo
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Abbas Sakandar
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Dong Li
- Department of Pediatrics, The First Affiliated Hospital of Dalian Medical University, Dalian116011, China.
| |
Collapse
|
4
|
He J, Tang Q, Liu YC, Wang LJ, Chai YF. Impact of Diquat on the Intestinal Health and the Composition and Function of the Gut Microbiome. Antioxidants (Basel) 2025; 14:721. [PMID: 40563352 PMCID: PMC12189094 DOI: 10.3390/antiox14060721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 06/07/2025] [Accepted: 06/10/2025] [Indexed: 06/28/2025] Open
Abstract
Diquat (DQ) is extensively utilized as a herbicide in farming, and its intake can result in serious systemic toxicity due to its induction of oxidative stress (OS) and disruption of intestinal homeostasis. The gastrointestinal tract is one of the first systems exposed to DQ, and damage to this system can influence the general health of the host. Our review summarizes the toxic effects of DQ on the intestinal barrier integrity, gut microbiome, and microbial metabolites (e.g., short-chain fatty acids [SCFAs], bile acids). By elucidating the mechanisms linking DQ-induced OS to gut dysbiosis, mitochondrial dysfunction, and inflammation, our work provides critical insights into novel therapeutic strategies, including probiotics, antioxidants (e.g., hydroxytyrosol, curcumin), and selenium nanoparticles. These findings address a pressing gap in understanding environmental toxin-related gut pathology and offer potential interventions to mitigate systemic oxidative damage.
Collapse
Affiliation(s)
| | | | | | - Li-Jun Wang
- Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin 300052, China; (J.H.); (Q.T.); (Y.-C.L.)
| | - Yan-Fen Chai
- Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin 300052, China; (J.H.); (Q.T.); (Y.-C.L.)
| |
Collapse
|
5
|
Zhu K, Jiang M, Yan M, Huang Y, Yang T, Zhu C. Characteristics and Functions of Different Intestinal Segments in Juvenile Greater Amberjack ( Seriola dumerili). Animals (Basel) 2025; 15:1672. [PMID: 40509138 PMCID: PMC12153913 DOI: 10.3390/ani15111672] [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: 04/12/2025] [Revised: 05/25/2025] [Accepted: 05/29/2025] [Indexed: 06/16/2025] Open
Abstract
The greater amberjack (Seriola dumerili), a key species in marine aquaculture, relies heavily on its intestine for nutrient absorption and immune function. However, the structural and functional specialization of its intestinal segments remains poorly understood. In this study, we divided the intestine of S. dumerili into foregut, midgut, and hindgut, and conducted a multi-omics analysis integrating histological staining (H&E/AB-PAS), digestive enzyme assays, transcriptome sequencing, and 16S rRNA microbiota profiling to characterize structural, functional, molecular, and microbial differences across intestinal segments. Histological examinations revealed that brush border microvillus length, muscle layer thickness, and folding height were significantly greater in the foregut and hindgut compared to the midgut, while mucus and goblet cell density was higher in the foregut and midgut. Digestive enzyme assays showed that lipase activity peaked in the foregut, α-amylase in the midgut, and protease in the midgut and hindgut. Alkaline phosphatase (AKP) and acid phosphatase (ACP) activities were highest in the foregut and midgut. Immune-related enzyme activities (SOD (Superoxide dismutase), GSH-Px (Glutathione peroxidase), T-AOC (Total Antioxidant Capacity)) were elevated and MDA levels were lower in the midgut, indicating its role as the primary immune site. Transcriptome analysis identified segment-specific expression of nutrient transporters, such as slc6a19b (hindgut, protein), apoa1b (foregut, lipid), and slc37a4 (midgut, carbohydrate). Microbiome analysis revealed Ruminococcus dominance in the foregut (lipid digestion) and Prevotella, Bifidobacterium, and Lactobacillus enrichment in the midgut (carbohydrate metabolism and immunity). These findings highlight functional zonation in S. dumerili: the foregut specializes in lipid digestion, the midgut in carbohydrate metabolism and immunity, and the hindgut in protein digestion. This study provides foundational insights for optimizing aquaculture practices and advancing research in nutrition, immunology, and disease modeling in S. dumerili.
Collapse
Affiliation(s)
- Kunfeng Zhu
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (K.Z.); (M.J.); (M.Y.); (Y.H.); (T.Y.)
| | - Mouyan Jiang
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (K.Z.); (M.J.); (M.Y.); (Y.H.); (T.Y.)
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Ocean University, Zhanjiang 524088, China
- Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong Ocean University, Zhanjiang 524088, China
| | - Mengyao Yan
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (K.Z.); (M.J.); (M.Y.); (Y.H.); (T.Y.)
| | - Yang Huang
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (K.Z.); (M.J.); (M.Y.); (Y.H.); (T.Y.)
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Ocean University, Zhanjiang 524088, China
- Development and Research Center for Biological Marine Resources, Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
| | - Tonglin Yang
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (K.Z.); (M.J.); (M.Y.); (Y.H.); (T.Y.)
- Development and Research Center for Biological Marine Resources, Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
| | - Chunhua Zhu
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (K.Z.); (M.J.); (M.Y.); (Y.H.); (T.Y.)
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Ocean University, Zhanjiang 524088, China
- Development and Research Center for Biological Marine Resources, Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
- Agro-Tech Extension Center of Guangdong Province, Guangzhou 510520, China
| |
Collapse
|
6
|
Yan R, Zhang L, Chen Y, Zheng Y, Xu P, Xu Z. Therapeutic potential of gut microbiota modulation in epilepsy: A focus on short-chain fatty acids. Neurobiol Dis 2025; 209:106880. [PMID: 40118219 DOI: 10.1016/j.nbd.2025.106880] [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: 11/17/2024] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025] Open
Abstract
According to the criteria established by the International League Against Epilepsy (ILAE), epilepsy is defined as a disorder characterized by at least two unprovoked seizures occurring more than 24 h apart. Its pathogenesis is closely related to various physiological and pathological factors. Advances in high-throughput metagenomic sequencing have increasingly highlighted the role of gut microbiota dysbiosis in epilepsy. Short-chain fatty acids (SCFAs), the major metabolites of the gut microbiota and key regulators of the gut-brain axis, support physiological homeostasis through multiple mechanisms. Recent studies have indicated that SCFAs not only regulate seizures by maintaining intestinal barrier integrity and modulating intestinal immune responses, but also affect the structure and function of the blood-brain barrier (BBB) and regulate neuroinflammation. This review, based on current literatures, explores the relationship between SCFAs and epilepsy, emphasizing how SCFAs affect epilepsy by modulating the intestinal barrier and BBB. In-depth studies on SCFAs may reveal their therapeutic potential and inform the development of gut microbiota-targeted epilepsy treatments.
Collapse
Affiliation(s)
- Rong Yan
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Linhai Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ya Chen
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yongsu Zheng
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Ping Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| | - Zucai Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, China; Key Laboratory of Brain Function and Brain Disease Prevention and Treatment of Guizhou Province, Zunyi, China.
| |
Collapse
|
7
|
Guo W, Tang X, Zhang Q, Xiong F, Yan Y, Zhao J, Mao B, Zhang H, Cui S. Lacticaseibacillus paracasei CCFM1222 Ameliorated the Intestinal Barrier and Regulated Gut Microbiota in Mice with Dextran Sulfate Sodium-Induced Colitis. Probiotics Antimicrob Proteins 2025; 17:1001-1013. [PMID: 38376820 DOI: 10.1007/s12602-024-10236-0] [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] [Accepted: 02/13/2024] [Indexed: 02/21/2024]
Abstract
Lacticaseibacillus paracasei has been regarded as a probiotic bacterium because of its role in anti-inflammatory properties and maintenance of intestinal barrier permeability. Here, we explored the anticolitic effects and mechanism of L. paracasei CCFM1222. The results showed that L. paracasei CCFM1222 supplementation could suppress the disease activity index (DAI) and colon length shortening in colitis mice, accompanied by a moderate increase in colonic tight junction proteins (ZO-1, occludin and claudin-1). L. paracasei CCFM1222 intervention significantly suppressed the levels of inflammatory cytokines (TNF-α, IL-1β, and IL-6) and significantly elevated the activities of antioxidant enzymes (including SOD, GSH-Px, and CAT) in the colon by regulating the TLR4/MyD88/NF-κB and Nrf2 signaling pathways in colitis mice. In addition, L. paracasei CCFM1222 significantly shifted the gut microbiota, including elevating the abundance of Catabacter, Ruminiclostridium 9, Alistipes, and Faecalibaculum, as well as reducing the abundance of Mucispirillum, Escherichia-Shigella, and Salmonella, which was associated with the improvement of colonic barrier damage. Overall, these results suggest that L. paracasei CCFM1222 is a good candidate for probiotic of improving colonic barrier damage and associated diseases.
Collapse
Affiliation(s)
- Weiling Guo
- State Key Laboratory of Food Science and Resources, Jiangnan University, Lihu Avenue 1800, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Xin Tang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Lihu Avenue 1800, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Qiuxiang Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Lihu Avenue 1800, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Feifei Xiong
- Ningbo Yuyi Biotechnology Co., Ltd, Ningbo, 315153, China
| | - Yongqiu Yan
- Ningbo Yuyi Biotechnology Co., Ltd, Ningbo, 315153, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Lihu Avenue 1800, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Bingyong Mao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Lihu Avenue 1800, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.
- Ningbo Yuyi Biotechnology Co., Ltd, Ningbo, 315153, China.
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Lihu Avenue 1800, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, 214122, China
| | - Shumao Cui
- State Key Laboratory of Food Science and Resources, Jiangnan University, Lihu Avenue 1800, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- Ningbo Yuyi Biotechnology Co., Ltd, Ningbo, 315153, China
| |
Collapse
|
8
|
Neurath MF, Artis D, Becker C. The intestinal barrier: a pivotal role in health, inflammation, and cancer. Lancet Gastroenterol Hepatol 2025; 10:573-592. [PMID: 40086468 DOI: 10.1016/s2468-1253(24)00390-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/08/2024] [Accepted: 11/15/2024] [Indexed: 03/16/2025]
Abstract
The intestinal barrier serves as a boundary between the mucosal immune system in the lamina propria and the external environment of the intestinal lumen, which contains a diverse array of microorganisms and ingested environmental factors, including pathogens, food antigens, toxins, and other foreign substances. This barrier has a central role in regulating the controlled interaction between luminal factors and the intestinal immune system. Disruptions of intestinal epithelial cells, which serve as a physical barrier, or the antimicrobial peptides and mucins they produce, which act as a chemical barrier, can lead to a leaky gut. In this state, the intestinal wall is unable to efficiently separate the intestinal flora and luminal contents from the intestinal immune system. The subsequent activation of the immune system has an important role in the pathogenesis of inflammatory bowel disease, as well as in metabolic dysfunction-associated steatohepatitis, primary sclerosing cholangitis, and colorectal cancer. Dysregulated intestinal barrier integrity has also been described in patients with chronic inflammatory diseases outside the gastrointestinal tract, including rheumatoid arthritis and neurodegenerative disorders. Mechanistic studies of barrier dysfunction have revealed that the subsequent local activation and systemic circulation of activated immune cells and the cytokines they secrete, as well as extracellular vesicles, promote proinflammatory processes within and outside the gastrointestinal tract. In this Review, we summarise these findings and highlight several new therapeutic concepts currently being developed that attempt to control inflammatory processes via direct or indirect modulation of intestinal barrier function.
Collapse
Affiliation(s)
- Markus F Neurath
- Medical Clinic 1, Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA; Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA; Joan and Sanford I Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Allen Discovery Center for Neuroimmune Interactions, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Christoph Becker
- Medical Clinic 1, Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
9
|
Li Z, Liu L, Sun Y, Liu X, Zhang P, Wang Y, Ding G. Mesenchymal stem/stromal cells-derived exosomes: possible therapeutic mechanism in inflammatory bowel disease. Hum Cell 2025; 38:111. [PMID: 40434563 DOI: 10.1007/s13577-025-01243-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder of the gastrointestinal tract caused by dysfunction of the immune system in genetically susceptible individuals. As current pharmacologic and surgical treatments remain suboptimal, increasing attention has been directed toward exosomes derived from mesenchymal stem/stromal cells (MSCs) as alternative therapeutic approaches. MSCs are multipotent stromal cells that can be isolated from various human tissues such as bone marrow, adipose, umbilical cord and periodontal ligament. Exosomes are cell-derived membrane-bound vesicles enclosing RNAs, proteins, growth factors, and cytokines. Previous studies indicate that the anti-inflammatory, immunomodulatory, and regenerative effects of MSCs are largely mediated by MSC-derived exosomes (MSC-Exos). Therefore, this review outlines current insights into the molecular mechanisms of MSC-Exos in IBD treatment to support the future development of MSC-Exos as a therapeutic strategy, thus providing novel observations into the clinical applications of MSC-Exos in IBD management.
Collapse
Affiliation(s)
- Zekun Li
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China
| | - Luyun Liu
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China
| | - Yuhui Sun
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China
| | - Xinjuan Liu
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China
| | - Ping Zhang
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China
| | - Yue Wang
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China
| | - Gang Ding
- School of Stomatology, Shandong Second Medical University, Baotong West Street No. 7166, Weifang, 261053, Shandong Province, China.
| |
Collapse
|
10
|
Quan T, Li R, Chen Y, Gao T. Regulatory Mechanism of Intestinal Stem Cells Based on Hippo Pathway and Signaling Crosstalk in Chicken. Int J Mol Sci 2025; 26:5067. [PMID: 40507877 PMCID: PMC12155279 DOI: 10.3390/ijms26115067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2025] [Revised: 05/16/2025] [Accepted: 05/21/2025] [Indexed: 06/16/2025] Open
Abstract
Recently, there has been a gradual increase in the demand for chicken and eggs. The gut, as the vital place of nutrient digestion and absorption, is highly associated with the development of livestock and poultry and the quality of meat, eggs, and milk. Intestinal stem cells, as an important source of intestinal cell proliferation and renewal, exert a vital effect on repairing injured intestinal epithelial cells and keeping homeostasis. Intestinal stem cell-regulated intestinal epithelial balance is closely controlled and modulated by interlinked developmental loops that maintain cell proliferation and differentiation processes in balance. Some conservative signaling pathways, including the Wnt, Notch, hedgehog, and bone morphogenetic protein (BMP) loops, have been proved to modulate intestinal health in poultry. Meanwhile, studies have revealed the importance of the Hippo pathway in gastrointestinal tract physiology by regulating intestinal stem cells. Moreover, crosstalk between Hippo and other signaling pathways provides tight, yet versatile, regulation of tissue homeostasis. In this review, we summarize studies on the role of the Hippo pathway in the intestine in these physiological processes and the underlying mechanisms responsible via interacting with these signaling pathways and discuss future research directions and potential therapeutic strategies targeting Hippo signaling in intestinal disease. A comprehensive understanding of how these signaling pathways regulate stem cell proliferation, differentiation, and self-renewal will help to understand the regulation of intestinal homeostasis. In addition, it has the capacity for creative ways to govern intestinal damage, enteritis, and associated disorders induced by different factors.
Collapse
Affiliation(s)
| | | | | | - Ting Gao
- College of Veterinary Medicine, China Agricultural University, Beijing 100083, China; (T.Q.); (R.L.); (Y.C.)
| |
Collapse
|
11
|
Hu K, Ren H, Yuan C, Liu Z, Zhang X, Zhang H, Zhou N, Chen L, Wang N, Zhang G. Comparison of Different Methods of Constructing a Celiac Disease Model in BALB/c Mice. Mol Nutr Food Res 2025:e70118. [PMID: 40395135 DOI: 10.1002/mnfr.70118] [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: 09/10/2024] [Revised: 04/24/2025] [Accepted: 05/02/2025] [Indexed: 05/22/2025]
Abstract
Efficient and stable animal models of celiac disease (CD) are crucial for CD research, dietary supplementation research, and new drug development. This study aimed to establish CD models by administering gliadin to parental and first-generation mice on a gluten-free diet using two intraperitoneal injection and a combination of sensitization by intraperitoneal injection and gavage. Various indicators, including clinical manifestations, characteristic indicators of CD, inflammatory factors, intestinal barriers, immune cells, and other related indicators, were used to compare different modeling methods. The results showed that all four methods induced varying degrees of CD symptoms in the mice. The analysis revealed that intraperitoneal injection in first-generation mice significantly increased specific IgG and total IgE antibody levels; markedly shortened intestinal villus and crypt hyperplasia in the jejunum; significantly increased pro-inflammatory cytokines IFN-γ, IL-17, IL-15, and TNF-α; and significantly decreased the anti-inflammatory cytokine IL-4. Additionally, immune cells in the spleen and intestinal lymph nodes were severely imbalanced. These results suggest that the intraperitoneal injection model in first-generation mice is more efficient, stable, and significant. This study provides a theoretical basis for the efficient construction of CD models.
Collapse
Affiliation(s)
- Kexin Hu
- Zhengzhou Key Laboratory of Nutrition and Health Food, College of Food Science and Technology, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory, Zhengzhou, China
| | - Hongtao Ren
- Zhengzhou Key Laboratory of Nutrition and Health Food, College of Food Science and Technology, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory, Zhengzhou, China
| | - Chong Yuan
- Zhengzhou Key Laboratory of Nutrition and Health Food, College of Food Science and Technology, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory, Zhengzhou, China
| | - Zhigang Liu
- Longhu Laboratory, Zhengzhou, China
- College of Food Science and Engineering, Northwest A&F University, Shaanxi, China
| | - Xing Zhang
- Longhu Laboratory, Zhengzhou, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Huaqiang Zhang
- Longhu Laboratory, Zhengzhou, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Ning Zhou
- Zhengzhou Key Laboratory of Nutrition and Health Food, College of Food Science and Technology, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory, Zhengzhou, China
| | - Linlin Chen
- Zhengzhou Key Laboratory of Nutrition and Health Food, College of Food Science and Technology, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory, Zhengzhou, China
| | - Na Wang
- Zhengzhou Key Laboratory of Nutrition and Health Food, College of Food Science and Technology, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory, Zhengzhou, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Gaiping Zhang
- Longhu Laboratory, Zhengzhou, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- School of Advanced Agricultural Sciences, Peking University, Beijing, China
| |
Collapse
|
12
|
Zhang X, Wang Y, E Q, Naveed M, Wang X, Liu Y, Li M. The biological activity and potential of probiotics-derived extracellular vesicles as postbiotics in modulating microbiota-host communication. J Nanobiotechnology 2025; 23:349. [PMID: 40380331 PMCID: PMC12082936 DOI: 10.1186/s12951-025-03435-6] [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: 01/20/2025] [Accepted: 05/01/2025] [Indexed: 05/19/2025] Open
Abstract
Probiotics such as Lactobacillus and Bifidobacterium spp. have been shown to be critical for maintaining host homeostasis. In recent years, key compounds of postbiotics derived from probiotic metabolism and cellular secretion have been identified for their role in maintaining organ immunity and regulating intestinal inflammation. In particular, probiotic-derived extracellular vesicles (PEVs) can act as postbiotics, maintaining almost the same functional activity as probiotics. They also have strong biocompatibility and loading capacity to carry exogenous or parental active molecules to reach distal organs to play their roles. This provides a new direction for understanding the intrinsic microbiota-host communication mechanism. However, most current studies on PEVs are limited to their functional effects/benefits, and their specific physicochemical properties, composition, intrinsic mechanisms for maintaining host homeostasis, and possible threats remain to be explored. Here, we review and summarize the unique physicochemical properties of PEVs and their bioactivities and mechanisms in mediating microbiota-host communication, and elucidate the limitations of the current research on PEVs and their potential application as postbiotics.
Collapse
Affiliation(s)
- Xiaoming Zhang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Ye Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Qiyu E
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Muhammad Naveed
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Xiuli Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Yinhui Liu
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Ming Li
- College of Basic Medical Science, Dalian Medical University, Dalian, China.
| |
Collapse
|
13
|
He C, Cai G, Jia Y, Jiang R, Wei X, Tao N. Effect of Diquat on gut health: molecular mechanisms, toxic effects, and protective strategies. Front Pharmacol 2025; 16:1562182. [PMID: 40421207 PMCID: PMC12104255 DOI: 10.3389/fphar.2025.1562182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 04/28/2025] [Indexed: 05/28/2025] Open
Abstract
Diquat is a widely used bipyridyl herbicide that is extensively applied in agricultural production and water management due to its high efficacy in weed control. However, its environmental persistence and the toxic effects it induces have raised widespread concern. Studies show that Diquat primarily enters the body through the digestive tract, leading to poisoning. The core mechanism of its toxicity involves reactive oxygen species (ROS)-induced oxidative stress, which not only directly damages the intestinal barrier function but also exacerbates inflammation and systemic toxicity by disrupting the balance of the gut microbiota and the normal production of metabolic products. This review systematically summarizes the physicochemical properties of Diquat, with a focus on analyzing the mechanisms by which it damages the gut tissue structure, barrier function, and microbiota after digestive tract exposure. The aim is to provide theoretical support for a deeper understanding of Diquat's toxic mechanisms and its digestive tract-centered toxic characteristics, laying a scientific foundation for the development of effective interventions and protective strategies against its toxicity.
Collapse
Affiliation(s)
| | | | | | | | - Xiaolan Wei
- Department of Emergency, Suining Central Hospital in Sichuan Province, Suining, Sichuan, China
| | - Ning Tao
- Department of Emergency, Suining Central Hospital in Sichuan Province, Suining, Sichuan, China
| |
Collapse
|
14
|
Zhang X, Jiang A, An S, Guo C, You F, Huang Z, Feng S, Zhang Y, Chang X, Yang G, Meng X. Dietary resistant starch supplementation improves the fish growth, lipid metabolism and intestinal barrier in largemouth bass (Micropterus salmoides) fed high-fat diets. Int J Biol Macromol 2025; 306:141356. [PMID: 39988156 DOI: 10.1016/j.ijbiomac.2025.141356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
Resistant starch (RS) is a novel type of prebiotic that exerts positive effects on lipid metabolism and intestinal flora. In this study, we investigated the effects of dietary RS on lipid metabolism and the intestinal barrier in largemouth bass (Micropterus salmoides). The experimental fish were fed either a control diet (C), a high-fat diet (H), or H diets supplemented with 0.5 %, 1.5 %, and 3 % RS (HRS0.5, HRS1.5, and HRS3.0). Dietary supplementation with 1.5 % and 3.0 % RS increased the final weight and feed utilization. Moreover, the hepatic crude protein content and the expression of genes related to lipid lipolysis were significantly higher in the HRS1.5 group compared to the H group, whereas hepatic crude lipid content and the expression of genes related to lipid synthesis were considerably lower in the HRS1.5 and HRS3.0 groups than in the H group. Additionally, hepatocyte vacuolation was alleviated in the HRS1.5 and HRS3.0 groups, and the number of liver lipid droplets was significantly decreased. Dietary supplementation with 1.5 % and 3.0 % RS downregulated the expression of pro-inflammatory factors while upregulating the expression of anti-inflammatory factors. Furthermore, analysis of gut microbiota composition revealed that RS supplementation increased the population of beneficial bacteria and short-chain fatty acid (SCFA) contents, decreased the abundance of pathogenic bacteria, and enhanced the diversity and richness of the intestinal flora. Non-targeted metabolomics analysis indicated that the levels of L-arginine and betaine were significantly higher in the HRS1.5 group, while levels of L-methionine and taurocholic acid were notably elevated in the HRS3.0 group. In conclusion, dietary supplementation with 1.5-3.0 % RS improved the balance of intestinal flora, promoted the growth of beneficial bacteria, adjusted the metabolites profile, and increased the SCFA levels. These results suggest that dietary supplementation with 1.5-3.0 % RS can restore the intestinal protective barrier, reduce hepatic lipid accumulation, and regulate lipid metabolism in largemouth bass.
Collapse
Affiliation(s)
- Xindang Zhang
- College of Fisheries, Henan Normal University, Xinxiang 453007, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, China
| | - Aixia Jiang
- College of Fisheries, Henan Normal University, Xinxiang 453007, China
| | - Shuxia An
- College of Fisheries, Henan Normal University, Xinxiang 453007, China
| | - Chongchong Guo
- College of Fisheries, Henan Normal University, Xinxiang 453007, China
| | - Fu You
- College of Fisheries, Henan Normal University, Xinxiang 453007, China
| | - Zhenyi Huang
- College of Fisheries, Henan Normal University, Xinxiang 453007, China
| | - Shikun Feng
- College of Fisheries, Henan Normal University, Xinxiang 453007, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, China
| | - Yanmin Zhang
- College of Fisheries, Henan Normal University, Xinxiang 453007, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, China
| | - Xulu Chang
- College of Fisheries, Henan Normal University, Xinxiang 453007, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, China
| | - Guokun Yang
- College of Fisheries, Henan Normal University, Xinxiang 453007, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, China
| | - Xiaolin Meng
- College of Fisheries, Henan Normal University, Xinxiang 453007, China; Engineering Technology Research Center of Henan Province for Aquatic Animal Cultivation, Henan Normal University, Xinxiang 453007, China.
| |
Collapse
|
15
|
Martínez‐Augustin O, Tena‐Garitaonaindia M, Ceacero‐Heras D, Jiménez‐Ortas Á, Enguix‐Huete JJ, Álvarez‐Mercado AI, Ruiz‐Henares G, Aranda CJ, Gámez‐Belmonte R, Sánchez de Medina F. Macronutrients as Regulators of Intestinal Epithelial Permeability: Where Do We Stand? Compr Rev Food Sci Food Saf 2025; 24:e70178. [PMID: 40421830 PMCID: PMC12108046 DOI: 10.1111/1541-4337.70178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/23/2025] [Accepted: 04/04/2025] [Indexed: 05/28/2025]
Abstract
The intestinal barrier function (IBF) is essential for intestinal homeostasis. Its alterations have been linked to intestinal and systemic disease. Regulation of intestinal permeability is key in the maintenance of the IBF, in which the intestinal epithelium and tight junctions, the mucus layer, sIgA, and antimicrobial peptides are important factors. This review addresses the concept of IBF, focusing on permeability, and summarizes state-of-the-art information on how starvation and macronutrients regulate it. Novel mechanisms regulate intestinal permeability, like its induction by the normal process of nutrient absorption, the contribution of starvation-induced autophagy, or the stimulation of sIgA production by high-protein diets in a T-cell-independent fashion. In addition, observations evidence that starvation and protein restriction increase intestinal permeability, compromising mucin, antimicrobial peptides, and/or intestinal sIgA production. Regarding specific macronutrients, substantial evidence indicates that casein (compared to other protein sources), specific protein-derived peptides and glutamine reinforce IBF. Dietary carbohydrates regulate intestinal permeability in a structure- and composition-dependent fashion; fructose, glucose, and sucrose increase it, while nondigestible oligosaccharides (NDOs) decrease it. Among NDOs, human milk oligosaccharides (HMOs) stand as a promising tool. NODs effects are mediated by intestinal microbiota modulation, production of short-chain fatty acids, and direct interactions with intestinal cells. Finally, evidence supports avoiding high-fat diets for their detrimental effects on IBF. Most studies have been carried out in vitro or in animal models. More information is needed from clinical studies to substantiate beneficial effects and the use of macronutrients in the treatment and prevention of IBF-related diseases.
Collapse
Affiliation(s)
- Olga Martínez‐Augustin
- Department of Biochemistry and Molecular Biology II, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Insituto de Nutrición y Tecnología de los alimentos José Mataix and Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Mireia Tena‐Garitaonaindia
- Department of Biochemistry and Molecular Biology II, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Insituto de Nutrición y Tecnología de los alimentos José Mataix and Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Diego Ceacero‐Heras
- Department of Biochemistry and Molecular Biology II, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Insituto de Nutrición y Tecnología de los alimentos José Mataix and Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Ángela Jiménez‐Ortas
- Department of Biochemistry and Molecular Biology II, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Insituto de Nutrición y Tecnología de los alimentos José Mataix and Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Juan J. Enguix‐Huete
- Department of Pharmacology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Ana I. Álvarez‐Mercado
- Department of Pharmacology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Guillermo Ruiz‐Henares
- Department of Pharmacology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Carlos J. Aranda
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina‐ IBIMA Plataforma BIONANDRICORS “Enfermedades inflamatorias”MálagaSpain
| | - Reyes Gámez‐Belmonte
- Department of Pharmacology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
- Department of Medicine 1University of Erlangen‐NurembergErlangenGermany
| | - Fermín Sánchez de Medina
- Department of Pharmacology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| |
Collapse
|
16
|
Hsu SJ, Chung HC, Chang CH, Liao YC, Liu TW, Lin SM, Lee CK. Rapid evaluation of apigenin bioavailability and hypouricemic bioactivity by targeted metabolomics study in enterohepatic microenvironment mimetic cell culture model. Food Res Int 2025; 209:116281. [PMID: 40253145 DOI: 10.1016/j.foodres.2025.116281] [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: 11/30/2024] [Revised: 02/25/2025] [Accepted: 03/12/2025] [Indexed: 04/21/2025]
Abstract
Apigenin was previously identified as a potent hypouricemic agent by comparative metabolomics and in silico analysis. Bioavailability (absorption, distribution, metabolism, and excretion) is the major concern for bioefficacy of phytochemicals administered orally. Therefore, the objective of this study was to establish an enterohepatic microenvironment biomimetic model comprised of intestine epithelial cells and liver cells to evaluate the bioavailability and hypouricemic bioactivity of apigenin. The results indicated that cumulative 26 % of the supplemented apigenin was absorbed by intestinal epithelium over 240 min. With a maximum 24 % (1.37 μg/mL) of the absorbed apigenin, in its parent form, was distributed to the basolateral extra-enterohepatic compartment. Extensive phase I and phase II metabolism occurred in both enterocytes and hepatocytes. Ten and eighteen metabolites were detected in apical and basolateral medium representing intestinal excretion and systemic distribution, respectively. Apigenin-7-sulfate was the predominant metabolite released in intestinal lumen, while apigenin sulfation, acetylation, and taurine-conjugated products were the major metabolites likely distributed systemically. Importantly, apigenin supplementation significantly lowered uric acid level in the basolateral compartment, which demonstrated its hypouricemic bioactivity after the absorption through intestinal epithelium and supported its potential as a nutraceutical for hyperuricemia prevention. This in vitro enterohepatic model provides a valuable tool for rapidly assessing the bioavailability and bioactivity of dietary components.
Collapse
Affiliation(s)
- Su-Jung Hsu
- Institute of Fisheries Science, National Taiwan University, Taipei City, Taiwan; School of Pharmacy, Taipei Medical University, Taipei City, Taiwan
| | - Hui-Chen Chung
- Department of Medical Research, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi City, Taiwan
| | - Chia-Hsuan Chang
- Department of Food Science, National Chiayi University, Chiayi City, Taiwan
| | - Yi-Chi Liao
- Department of Food Science, National Chiayi University, Chiayi City, Taiwan
| | - Ta-Wei Liu
- School of Pharmacy, Taipei Medical University, Taipei City, Taiwan
| | - Shu-Mei Lin
- Department of Food Science, National Chiayi University, Chiayi City, Taiwan.
| | - Ching-Kuo Lee
- School of Pharmacy, Taipei Medical University, Taipei City, Taiwan.
| |
Collapse
|
17
|
Janto NV, Gleizes AR, Sun SJ, Ari G, Rao V, Gracz AD. Tritrichomonas muris sensitizes the intestinal epithelium to doxorubicin-induced apoptosis. Am J Physiol Gastrointest Liver Physiol 2025; 328:G594-G609. [PMID: 40243204 DOI: 10.1152/ajpgi.00242.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/26/2024] [Accepted: 04/03/2025] [Indexed: 04/18/2025]
Abstract
Doxorubicin (DXR) is a widely used chemotherapy drug that can induce severe intestinal mucositis. Although the influence of gut bacteria on DXR-induced damage has been documented, the role of eukaryotic commensals remains unexplored. We discovered Tritrichomonas muris (Tmu) in one of our mouse colonies exhibiting abnormal tuft cell hyperplasia, prompting an investigation into its impact on DXR-induced intestinal injury. Mice from Tmu-colonized and Tmu-excluded facilities were injected with DXR. Tissue morphology and gene expression were evaluated at acute injury (6 h) and regenerative (72 h and 120 h) phases. Changes to crypt and villus morphology were more subtle than previously reported and region-specific, with significantly shorter jejunal villi in Tmu+ mice at 72 h post-DXR compared with Tmu- controls. Most notably, we observed elevated rates of DXR-induced apoptosis, measured by cleaved caspase 3 (CC3) staining, in Tmu+ intestinal crypts at 6 h post-DXR. Tmu+ mice also exhibited reduced expression of active intestinal stem cell (aISC) marker Lgr5 and facultative ISC (fISC) marker Ly6a at 6 h post-DXR compared with Tmu- controls. Tmu, but not DXR, was associated with increased inflammation and expression of type 2 cytokines IL-5 and IL-13. Tmu+ mice also exhibited a decreased fecal abundance of Lactobacillus, which promotes gut barrier integrity, and reduced claudin expression, indicating potential barrier dysfunction that could explain the increase in DXR-induced apoptosis. These findings highlight the significant influence of commensal microbiota, particularly eukaryotic organisms like Tmu, on intestinal biology and response to chemotherapy, underscoring the complexity of gut microbiota interactions in drug-induced mucositis.NEW & NOTEWORTHY Our study found that the eukaryotic commensal Tritrichomonas muris (Tmu) significantly increases DXR-induced intestinal apoptosis in mice. Tmu also reduces Lgr5 expression post-DXR injury and elevates inflammation and type 2 cytokine expression in the absence of injury. 16S sequencing identifies decreased abundance of protective Lactobacillus in Tmu colonized mice, as well as decreased expression of barrier-forming claudins, which may explain increased apoptosis. These findings emphasize the complex role of microbiota in drug-induced intestinal damage.
Collapse
Affiliation(s)
- Nicolas V Janto
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, Georgia, United States
- Graduate Program in Genetics and Molecular Biology, Emory University, Atlanta, Georgia, United States
| | - Antoine R Gleizes
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, Georgia, United States
| | - Siyang J Sun
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, Georgia, United States
| | - Gurel Ari
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, Georgia, United States
| | - Vivek Rao
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, Georgia, United States
| | - Adam D Gracz
- Department of Medicine, Division of Digestive Diseases, Emory University, Atlanta, Georgia, United States
- Graduate Program in Genetics and Molecular Biology, Emory University, Atlanta, Georgia, United States
- Department of Human Genetics, Emory University, Atlanta, Georgia, United States
| |
Collapse
|
18
|
Lin W, Ruishi X, Caijiao X, Haoming L, Xuefeng H, Jiyou Y, Minqiang L, Shuo Z, Ming Z, Dongyang L, Xiaoxue F. Potential applications and mechanisms of natural products in mucosal-related diseases. Front Immunol 2025; 16:1594224. [PMID: 40370438 PMCID: PMC12075308 DOI: 10.3389/fimmu.2025.1594224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Accepted: 04/09/2025] [Indexed: 05/16/2025] Open
Abstract
The mucosal barrier serves as a crucial defense against external pathogens and allergens, being widely distributed across the respiratory, gastrointestinal, urogenital tracts, and oral cavity. Its disruption can lead to various diseases, including inflammatory bowel disease, asthma, urinary tract infections, and oral inflammation. Current mainstream treatments for mucosa-associated diseases primarily involve glucocorticoids and immunosuppressants, but their long-term use may cause adverse effects. Therefore, the development of safer and more effective therapeutic strategies has become a focus of research. Natural products, with their multi-target and multi-system regulatory advantages, offer a promising avenue for the treatment of mucosal diseases. This review summarizes the potential applications of natural products in diseases of mucosal barrier dysfunction through mechanisms such as immune modulation, inflammation inhibition, tight junction protein restoration, and gut microbiota regulation, with the aim of providing insights for the exploration of novel therapeutic strategies.
Collapse
Affiliation(s)
- Wang Lin
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Xie Ruishi
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Xu Caijiao
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Luo Haoming
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Hua Xuefeng
- The First People’s Hospital of Guangzhou, Department of Hepatobiliary and Pancreatic Surgery, Guangzhou, China
| | - Yao Jiyou
- The First People’s Hospital of Guangzhou, Department of Hepatobiliary and Pancreatic Surgery, Guangzhou, China
| | - Lu Minqiang
- The First People’s Hospital of Guangzhou, Department of Hepatobiliary and Pancreatic Surgery, Guangzhou, China
| | - Zhou Shuo
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Zhu Ming
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Li Dongyang
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Fang Xiaoxue
- Changchun University of Chinese Medicine, Changchun, China
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
19
|
Lin X, Xia L, Zhou Y, Xie J, Tuo Q, Lin L, Liao D. Crosstalk Between Bile Acids and Intestinal Epithelium: Multidimensional Roles of Farnesoid X Receptor and Takeda G Protein Receptor 5. Int J Mol Sci 2025; 26:4240. [PMID: 40362481 PMCID: PMC12072030 DOI: 10.3390/ijms26094240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Bile acids and their corresponding intestinal epithelial receptors, the farnesoid X receptor (FXR), the G protein-coupled bile acid receptor (TGR5), play crucial roles in the physiological and pathological processes of intestinal epithelial cells. These acids and receptors are involved in the regulation of intestinal absorption, signal transduction, cellular proliferation and repair, cellular senescence, energy metabolism, and the modulation of gut microbiota. A comprehensive literature search was conducted using PubMed, employing keywords such as bile acid, bile acid receptor, FXR (nr1h4), TGR5 (gpbar1), intestinal epithelial cells, proliferation, differentiation, senescence, energy metabolism, gut microbiota, inflammatory bowel disease (IBD), colorectal cancer (CRC), and irritable bowel syndrome (IBS), with a focus on publications available in English. This review examines the diverse effects of bile acid signaling and bile receptor pathways on the proliferation, differentiation, senescence, and energy metabolism of intestinal epithelial cells. Additionally, it explores the interactions between bile acids, their receptors, and the microbiota, as well as the implications of these interactions for host health, particularly in relation to prevalent intestinal diseases. Finally, the review highlights the importance of developing highly specific ligands for FXR and TGR5 receptors in the context of metabolic and intestinal disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Duanfang Liao
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (X.L.); (L.X.); (Y.Z.); (J.X.); (Q.T.); (L.L.)
| |
Collapse
|
20
|
Zhong C, Du J, Zhu H, Gao J, Xu G, Xu P. Intestinal Microbiota and Gene Expression Alterations in Chinese Mitten Crab ( Eriocheir sinensis) Under Deltamethrin Exposure. Antioxidants (Basel) 2025; 14:510. [PMID: 40427392 PMCID: PMC12108348 DOI: 10.3390/antiox14050510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/15/2025] [Accepted: 03/26/2025] [Indexed: 05/29/2025] Open
Abstract
The intestine is an important immune organ of aquatic animals and it plays an essential role in maintaining body health and anti-oxidative stress. To investigate the toxic effects of deltamethrin in intestinal tissue of Chinese mitten crabs (Eriocheir sinensis), 120 healthy crabs were randomly divided into two experimental groups (blank control group and deltamethrin-treated group), with three replicates in each group. After being treated with deltamethrin for 24 h, 48 h, 72 h, and 96 h, intestinal tissues were collected aseptically to assess the effects of deltamethrin on oxidative stress, immunity, apoptosis-related genes, and the structure of microflora in intestinal tissues. Additionally, correlations between gut microbiota composition and intestinal tissue damage-associated genes were analyzed. The results demonstrated that prolonged exposure to deltamethrin induced oxidative stress damage in intestinal tissue. Compared with the blank control group, the expression of autophagy-related genes B-cell lymphoma/Leukemia-2 (bcl-2), c-Jun N-terminal kinase (jnk), Microtuble-associated protein light chain 3 (lc3c), Cysteine-dependent Aspartate-specific Protease 8 (caspase 8), BECN1(beclin1), oxidative stress damage-related genes MAS1 proto-oncogene (mas), Glutathione Peroxidase (gpx), kelch-like ECH-associated protein 1 (keap1), Sequestosome 1 (p62), Interleukin-6 (il-6), and immune-related genes Lipopolysaccharide-induced TNF-alpha Factor (litaf), Heat shock protein 90 (hsp90) and prophenoloxidase (propo) in the deltamethrin treatment group were significantly up-regulated at 96 h (p < 0.05 or p < 0.01). Additionally, 16S rRNA sequencing showed that the diversity of intestinal flora in the deltamethrin-treated group was significantly higher compared with the blank control group (p < 0.01). Analysis of the differences in the composition of intestinal flora at the genus level showed that the relative abundance of Candidatus Bacilloplasma in the deltamethrin treatment group was significantly lower than that in the blank control group (p < 0.01). In contrast, the relative abundances of Flavobacterium, Lachnospiraceae_NK4A136_group, Acinetobacter, Chryseobacterium, Lacihabitans, Taibaiella, Hydrogenophaga, Acidovorax, and Undibacterium were significantly higher than those in the blank control group (p < 0.05 or p < 0.01). Pearson correlation analysis revealed that Malaciobacter, Shewanella, and Prevotella exhibited significant positive correlations with gene indicators (jnk, gpx, lc3c, litaf, hsp90), while Dysgonomonas, Vibrio, and Flavobacterium demonstrated significant negative correlations with multiple gene indicators (caspase 8, p62, il-16, keap1, jnk, etc). These results demonstrate that deltamethrin significantly impacts the gut microbiota, immune function, and antioxidant capacity of E. sinensis. The changes in gut microbiota have correlations with the biomarkers of intestinal tissue injury genes, indicating that gut microbiota plays a crucial role in deltamethrin-induced intestinal tissue damage. These insights contribute to a better understanding of the ecological risks associated with deltamethrin exposure in aquatic organisms.
Collapse
Affiliation(s)
- Chunyi Zhong
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (C.Z.)
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (H.Z.)
| | - Jinliang Du
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (C.Z.)
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (H.Z.)
- International Joint Research Laboratory for Fish Immunopharmacology, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Haojun Zhu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (H.Z.)
| | - Jiancao Gao
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (H.Z.)
| | - Gangchun Xu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (C.Z.)
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (H.Z.)
| | - Pao Xu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China; (C.Z.)
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (H.Z.)
| |
Collapse
|
21
|
Nieman DC, Sakaguchi CA, Williams JC, Pathmasiri W, Rushing BR, McRitchie S, Sumner SJ. Selective Influence of Hemp Fiber Ingestion on Post-Exercise Gut Permeability: A Metabolomics-Based Analysis. Nutrients 2025; 17:1384. [PMID: 40284247 PMCID: PMC12030204 DOI: 10.3390/nu17081384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/02/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
Objectives: This study investigated the effects of 2-week ingestion of hemp fiber (high and low doses) versus placebo bars on gut permeability and plasma metabolite shifts during recovery from 2.25 h intensive cycling. Hemp hull powder is a rich source of two bioactive compounds, N-trans-caffeoyl tyramine (NCT) and N-trans-feruloyl tyramine (NFT), with potential gut health benefits. Methods: The study participants included 23 male and female cyclists. A three-arm randomized, placebo-controlled, double-blind, crossover design was used with two 2-week supplementation periods and 2-week washout periods. Supplement bars provided 20, 5, or 0 g/d of hemp hull powder. Participants engaged in an intensive 2.25 h cycling bout at the end of each of the three supplementation periods. Five blood samples were collected before and after supplementation (overnight fasted state), and at 0 h-, 1.5 h-, and 3 h-post-exercise. Five-hour urine samples were collected pre-supplementation and post-2.25 h cycling after ingesting a sugar solution containing 5 g of lactulose, 100 mg of 13C mannitol, and 1.9 g of mannitol in 450 mL of water. An increase in the post-exercise lactulose/13C mannitol ratio (L:13CM) was used as the primary indicator of altered gut permeability. Other outcome measures included muscle damage biomarkers (serum creatine kinase, myoglobin), serum cortisol, complete blood cell counts, and shifts in plasma metabolites using untargeted metabolomics. Results: No trial differences were found for L:13CM, cortisol, blood cell counts, and muscle damage biomarkers. Orthogonal partial least-squares discriminant analysis (OPLSDA) showed distinct trial differences when comparing high- and low-dose hemp fiber compared to placebo supplementation (R2Y = 0.987 and 0.995, respectively). Variable Importance in Projection (VIP) scores identified several relevant metabolites, including 3-hydroxy-4-methoxybenzoic acid (VIP = 1.9), serotonin (VIP = 1.5), 5-hydroxytryptophan (VIP = 1.4), and 4-methoxycinnamic acid (VIP = 1.4). Mummichog analysis showed significant effects of hemp fiber intake on multiple metabolic pathways, including alpha-linolenic acid, porphyrin, sphingolipid, arginine and proline, tryptophan, and primary bile acid metabolism. Conclusions: Hemp fiber intake during a 2-week supplementation period did not have a significant effect on post-exercise gut permeability in cyclists (2.25 h cycling bout) using urine sugar data. On the contrary, untargeted metabolomics showed that the combination of consuming nutrient-rich hemp fiber bars and exercising for 135 min increased levels of beneficial metabolites, including those derived from the gut in healthy cyclists.
Collapse
Affiliation(s)
- David C. Nieman
- Human Performance Laboratory, Appalachian State University, North Carolina Research Campus (NCRC), Kannapolis, NC 28081, USA; (C.A.S.); (J.C.W.)
| | - Camila A. Sakaguchi
- Human Performance Laboratory, Appalachian State University, North Carolina Research Campus (NCRC), Kannapolis, NC 28081, USA; (C.A.S.); (J.C.W.)
| | - James C. Williams
- Human Performance Laboratory, Appalachian State University, North Carolina Research Campus (NCRC), Kannapolis, NC 28081, USA; (C.A.S.); (J.C.W.)
| | - Wimal Pathmasiri
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (W.P.); (B.R.R.); (S.J.S.)
- Nutrition Research Institute, University of North Carolina at Chapel Hill, North Carolina Research Campus (NCRC), Kannapolis, NC 28081, USA;
| | - Blake R. Rushing
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (W.P.); (B.R.R.); (S.J.S.)
- Nutrition Research Institute, University of North Carolina at Chapel Hill, North Carolina Research Campus (NCRC), Kannapolis, NC 28081, USA;
| | - Susan McRitchie
- Nutrition Research Institute, University of North Carolina at Chapel Hill, North Carolina Research Campus (NCRC), Kannapolis, NC 28081, USA;
| | - Susan J. Sumner
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (W.P.); (B.R.R.); (S.J.S.)
- Nutrition Research Institute, University of North Carolina at Chapel Hill, North Carolina Research Campus (NCRC), Kannapolis, NC 28081, USA;
| |
Collapse
|
22
|
Atzeni A, Hernández-Cacho A, Khoury N, Babio N, Belzer C, Vioque J, Corella D, Fitó M, Clish C, Vidal J, Konstanti P, Gonzales-Palacios S, Coltell O, Goday A, Moreno Indias I, Carlos Chillerón S, Ruiz-Canela M, Tinahones FJ, Hu FB, Salas-Salvadó J. The link between ultra-processed food consumption, fecal microbiota, and metabolomic profiles in older mediterranean adults at high cardiovascular risk. Nutr J 2025; 24:62. [PMID: 40247349 PMCID: PMC12007308 DOI: 10.1186/s12937-025-01125-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/02/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Ultra-processed food (UPF) consumption has been linked to adverse metabolic outcomes, potentially mediated by alterations in gut microbiota and metabolite production. OBJECTIVE This study aims to explore the cross-sectional and longitudinal associations between NOVA-classified UPF consumption, fecal microbiota, and fecal metabolome in a population of Mediterranean older adults at high cardiovascular risk. METHODS A total of 385 individuals, aged between 55 and 75 years, were included in the study. Dietary and lifestyle information, anthropometric measurements, and stool samples were collected at baseline and after 1-year follow-up. Fecal microbiota and metabolome were assessed using 16 S rRNA sequencing and liquid chromatography-tandem mass spectrometry, respectively. RESULTS At baseline, higher UPF consumption was associated with lower abundance of Ruminococcaceae incertae sedis (β = - 0.275, P = 0.047) and lower concentrations of the metabolites propionylcarnitine (β = - 0.0003, P = 0.013) and pipecolic acid (β = - 0.0003, P = 0.040) in feces. Longitudinally, increased UPF consumption was linked to reduced abundance of Parabacteroides spp. after a 1-year follow-up (β = - 0.278, P = 0.002). CONCLUSIONS High UPF consumption was associated with less favorable gut microbiota and metabolite profiles, suggesting a possible link to reduced short-chain fatty acid (SCFA) production, altered mitochondrial energy metabolism, and impaired amino acid metabolism. These findings support the reduction of UPF consumption and the promotion of dietary patterns rich in fiber for better gut health. Further research is needed to confirm these associations and clarify the underlying mechanisms. TRIAL REGISTRATION ISRCTN89898870 ( https://doi.org/10.1186/ISRCTN89898870 ).
Collapse
Affiliation(s)
- Alessandro Atzeni
- Centro de Investigación Biomédica en Red - Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
- Desenvolupament i Salut Mental (ANUT-DSM) Departament de Bioquímica i Biotecnologia, Unitat de Nutrició Humana, Alimentació, Nutrició, Universitat Rovira i Virgili, Reus, Spain.
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain.
| | - Adrián Hernández-Cacho
- Centro de Investigación Biomédica en Red - Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Desenvolupament i Salut Mental (ANUT-DSM) Departament de Bioquímica i Biotecnologia, Unitat de Nutrició Humana, Alimentació, Nutrició, Universitat Rovira i Virgili, Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Nadine Khoury
- Centro de Investigación Biomédica en Red - Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Desenvolupament i Salut Mental (ANUT-DSM) Departament de Bioquímica i Biotecnologia, Unitat de Nutrició Humana, Alimentació, Nutrició, Universitat Rovira i Virgili, Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Nancy Babio
- Centro de Investigación Biomédica en Red - Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Desenvolupament i Salut Mental (ANUT-DSM) Departament de Bioquímica i Biotecnologia, Unitat de Nutrició Humana, Alimentació, Nutrició, Universitat Rovira i Virgili, Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Clara Belzer
- Laboratory of Microbiology, Wageningen University, Wageningen, Netherlands
| | - Jesús Vioque
- CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante, Universidad Miguel Hernández (ISABIAL-UMH), Alicante, Spain
| | - Dolores Corella
- Centro de Investigación Biomédica en Red - Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Preventive Medicine, University of Valencia, Valencia, Spain
| | - Montserrat Fitó
- Centro de Investigación Biomédica en Red - Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar de Investigaciones Médicas Municipal d'Investigació Médica (IMIM), Barcelona, Spain
| | - Clary Clish
- Metabolomics Platform, The Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Josep Vidal
- CIBER Diabetes y Enfermedades Metabólicas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Endocrinology, Institut d'Investigacions Biomédiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Prokopis Konstanti
- Laboratory of Microbiology, Wageningen University, Wageningen, Netherlands
| | - Sandra Gonzales-Palacios
- CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante, Universidad Miguel Hernández (ISABIAL-UMH), Alicante, Spain
| | - Oscar Coltell
- Centro de Investigación Biomédica en Red - Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Computer Languages and Systems, Universitat Jaume I, Castellón, Spain
| | - Albert Goday
- Centro de Investigación Biomédica en Red - Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar de Investigaciones Médicas Municipal d'Investigació Médica (IMIM), Barcelona, Spain
- Departament de Medicina, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Isabel Moreno Indias
- Centro de Investigación Biomédica en Red - Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Silvia Carlos Chillerón
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain
- Epidemiología y Salud Pública, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Miguel Ruiz-Canela
- Centro de Investigación Biomédica en Red - Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain
- Epidemiología y Salud Pública, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Francisco J Tinahones
- Centro de Investigación Biomédica en Red - Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology and Nutrition, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Frank B Hu
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Channing Division for Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jordi Salas-Salvadó
- Centro de Investigación Biomédica en Red - Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
- Desenvolupament i Salut Mental (ANUT-DSM) Departament de Bioquímica i Biotecnologia, Unitat de Nutrició Humana, Alimentació, Nutrició, Universitat Rovira i Virgili, Reus, Spain.
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain.
| |
Collapse
|
23
|
Klein Cerrejon D, Krupke H, Gao D, Paunović N, Sachs D, Leroux JC. Optimized suction patch design for enhanced transbuccal macromolecular drug delivery. J Control Release 2025; 380:875-891. [PMID: 39938719 DOI: 10.1016/j.jconrel.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/14/2025]
Abstract
Peptides represent a rapidly expanding class of drugs with broad therapeutic potential. However, due to their large molecular weight and susceptibility to degradation in the gastrointestinal tract, most peptide drugs are administered via subcutaneous injections. Despite extensive research, a painless broad delivery platform for these drugs is still lacking. Recently, an octopus-inspired buccal patch has shown promise in addressing this challenge by leveraging a synergistic combination of mechanical stretching and permeation enhancers. In this study, the patch and the loaded formulation were optimized to improve ease of use, scalability, and efficacy. Through assessments of mechanical properties, finite element simulations, and ex vivo experiments, we evaluated the effects of patch design and material, as well as the drug matrix composition and the formulation preparation methods on the delivery performance. A patch with a > 9-fold larger effective surface area, produced via mold casting of medical-grade silicone (shore hardness 50) and loaded with a lyophilized drug matrix, emerged as the most promising system. In beagle dogs, 30-min application of this patch resulted in a 14.6 % bioavailability for teriparatide (4118 g mol-1), while bioavailability of semaglutide (4114 g mol-1) was 9.6 times higher than that of the commercial tablet. This work showcases how systematic optimization of this technology can improve and simplify the buccal administration of macromolecular drugs, facilitating the clinical translation of this non-invasive dosage form.
Collapse
Affiliation(s)
- David Klein Cerrejon
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Hanna Krupke
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Daniel Gao
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Nevena Paunović
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - David Sachs
- Citus AG, Ueberlandstrasse 129, 8600 Dübendorf, Switzerland
| | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
24
|
Heckmann M, Sadova N, Sandner G, Neuhauser C, Blank-Landeshammer B, Schwarzinger B, König A, Liang M, Spitzer M, Weghuber J, Stadlbauer V. Herbal extract fermented with inherent microbiota improves intestinal health by exerting antioxidant and anti-inflammatory effects in vitro and in vivo. J Anim Sci Biotechnol 2025; 16:52. [PMID: 40188119 PMCID: PMC11972464 DOI: 10.1186/s40104-025-01178-w] [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: 11/22/2024] [Accepted: 02/13/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Maintaining intestinal health is crucial for the overall well-being and productivity of livestock, as it impacts nutrient absorption, immune function, and disease resistance. Oxidative stress and inflammation are key threats to intestinal integrity. This study explored the antioxidant, anti-inflammatory, and barrier-strengthening properties of a fermented plant macerate (FPM) derived from 45 local herbs, using a specifically developed fermentation process utilizing the plants' inherent microbiota to enhance bioactivity and sustainability. RESULTS In vitro experiments with IPEC-J2 cells showed that FPM significantly reduced intracellular reactive oxygen species (ROS) levels, improved barrier integrity, and enhanced cell migration under stress. Similar antioxidant effects were observed in THP-1 macrophages, where FPM reduced ROS production and modulated inflammatory responses by decreasing pro-inflammatory cytokines [tumor necrosis factor alpha (TNF-α), monokine induced by gamma interferon (MIG), interferon-inducible T cell alpha chemoattractant (I-TAC), macrophage inflammatory proteins (MIP)-1α and -1β] and increasing anti-inflammatory interleukin (IL)-10 levels. Mechanistic studies with HEK-Blue reporter cell lines revealed that FPM inhibited nuclear factor kappa B (NF-κB) activation via a toll-like receptor (TLR)4-independent pathway. In vivo, FPM significantly reduced ROS levels in Drosophila melanogaster and improved activity and LT50 values in Caenorhabditis elegans under oxidative stress, although it did not affect intestinal barrier integrity in these models. CONCLUSION The findings indicate that FPM shows promising application as a functional feed supplement for improving intestinal health in livestock by mitigating oxidative stress and inflammation. Further studies, including livestock feeding trials, are recommended to validate these results.
Collapse
Affiliation(s)
- Mara Heckmann
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, 4600, Wels, Austria
| | - Nadiia Sadova
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, 4600, Wels, Austria
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, 3430, Tulln, Austria
| | - Georg Sandner
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, 3430, Tulln, Austria
| | - Cathrina Neuhauser
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, 4600, Wels, Austria
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, 3430, Tulln, Austria
| | - Bernhard Blank-Landeshammer
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, 4600, Wels, Austria
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, 3430, Tulln, Austria
| | - Bettina Schwarzinger
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, 4600, Wels, Austria
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, 3430, Tulln, Austria
| | - Alice König
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, 4600, Wels, Austria
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, 3430, Tulln, Austria
| | - Meizhen Liang
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, 3430, Tulln, Austria
| | - Michael Spitzer
- TVA Produktions- & Vertriebs-Gesellschaft m.b.H, Dorf 156, 3343, Hollenstein, Austria
| | - Julian Weghuber
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, 4600, Wels, Austria.
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, 3430, Tulln, Austria.
| | - Verena Stadlbauer
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Stelzhamerstraße 23, 4600, Wels, Austria.
- FFoQSI GmbH-Austrian Competence Centre for Feed and Food Quality, Safety and Innovation, Technopark 1D, 3430, Tulln, Austria.
| |
Collapse
|
25
|
Luo L, Zuo Y, Dai L. Metabolic rewiring and inter-organ crosstalk in diabetic HFpEF. Cardiovasc Diabetol 2025; 24:155. [PMID: 40186193 PMCID: PMC11971867 DOI: 10.1186/s12933-025-02707-7] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) represents a significant and growing clinical challenge. Initially, for an extended period, HFpEF was simply considered as a subset of heart failure, manifesting as haemodynamic disorders such as hypertension, myocardial hypertrophy, and diastolic dysfunction. However, the rising prevalence of obesity and diabetes has reshaped the HFpEF phenotype, with nearly 45% of cases coexisting with diabetes. Currently, it is recognized as a multi-system disorder that involves the heart, liver, kidneys, skeletal muscle, adipose tissue, along with immune and inflammatory signaling pathways. In this review, we summarize the landscape of metabolic rewiring and the crosstalk between the heart and other organs/systems (e.g., adipose, gut, liver and hematopoiesis system) in diabetic HFpEF for the first instance. A diverse array of metabolites and cytokines play pivotal roles in this intricate crosstalk process, with metabolic rewiring, chronic inflammatory responses, immune dysregulation, endothelial dysfunction, and myocardial fibrosis identified as the central mechanisms at the heart of this complex interplay. The liver-heart axis links nonalcoholic steatohepatitis and HFpEF through shared lipid accumulation, inflammation, and fibrosis pathways, while the gut-heart axis involves dysbiosis-driven metabolites (e.g., trimethylamine N-oxide, indole-3-propionic acid and short-chain fatty acids) impacting cardiac function and inflammation. Adipose-heart crosstalk highlights epicardial adipose tissue as a source of local inflammation and mechanical stress, whereas the hematopoietic system contributes via immune cell activation and cytokine release. We contend that, based on the viewpoints expounded in this review, breaking this inter-organ/system vicious cycle is the linchpin of treating diabetic HFpEF.
Collapse
Affiliation(s)
- Lingyun Luo
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan, 430030, Hubei, China
| | - Yuyue Zuo
- Department of Dermatology, Wuhan No. 1 Hospital, Wuhan, 430030, Hubei, China.
| | - Lei Dai
- Department of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan, 430030, Hubei, China.
| |
Collapse
|
26
|
Liu HY, Li S, Ogamune KJ, Yuan P, Shi X, Ennab W, Ahmed AA, Kim IH, Hu P, Cai D. Probiotic Lactobacillus johnsonii Reduces Intestinal Inflammation and Rebalances Splenic Treg/Th17 Responses in Dextran Sulfate Sodium-Induced Colitis. Antioxidants (Basel) 2025; 14:433. [PMID: 40298818 PMCID: PMC12024357 DOI: 10.3390/antiox14040433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/30/2025] Open
Abstract
Inflammatory bowel disease (IBD), a chronic inflammatory disorder of the gastrointestinal tract, is frequently complicated by extraintestinal manifestations such as functional hyposplenism. Increasing evidence highlights its pathogenesis as a multifactorial interplay of gut dysbiosis, intestinal barrier dysfunction, and dysregulated immune responses. While probiotics, particularly Lactobacillus spp., have emerged as potential therapeutics for IBD, restoring intestinal homeostasis, their systemic immunomodulatory effects remain underexplored. Here, we investigated the protective role of Lactobacillus johnsonii N5 in DSS-induced colitis, focusing on inflammation inhibition and splenic T cell regulation. Pretreatment with L. johnsonii N5 significantly attenuated colitis severity, as evidenced by preserved body weight, reduced disease activity index, and prevention of colon shortening. N5 suppressed colonic pro-inflammatory factors such as TNF-α, Il-1b, Il-6, and CXCL1, while elevating anti-inflammatory IL-10 at both mRNA and protein levels. Transcriptomic analysis of the spleen revealed that N5 mediated the downregulation of inflammatory pathways, including the IL-17 and TNF signaling pathways, as well as the HIF-1 signaling pathway, and modulated the metabolic pathway of oxidative phosphorylation. Flow cytometry analysis demonstrated that N5 rebalanced splenic Treg/Th17 responses by expanding the Treg population and reducing the production of IL-17A in Th17 cells. Notably, Th17-associated IL-17A positively correlated with intestinal pro-inflammatory mediators, emphasizing the role of Th17 cells in driving colitis. In contrast, splenic Treg abundance positively correlated with colonic IL-10 levels, suggesting a link between systemic immune regulation and intestinal anti-inflammatory responses. Our study underscores the therapeutic potential of targeting gut-immune crosstalk through probiotics, thereby offering valuable insights for developing live bacterial-based interventions for IBD and other inflammatory disorders.
Collapse
Affiliation(s)
- Hao-Yu Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.); (P.Y.); (X.S.); (W.E.); (P.H.)
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Shicheng Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.); (P.Y.); (X.S.); (W.E.); (P.H.)
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Kennedy Jerry Ogamune
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.); (P.Y.); (X.S.); (W.E.); (P.H.)
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Peng Yuan
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.); (P.Y.); (X.S.); (W.E.); (P.H.)
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Xinyu Shi
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.); (P.Y.); (X.S.); (W.E.); (P.H.)
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Wael Ennab
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.); (P.Y.); (X.S.); (W.E.); (P.H.)
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Abdelkareem A. Ahmed
- Department of Veterinary Biomedical Sciences, Botswana University of Agriculture and Natural Resources, Gaborone P.O. Box 100, Botswana;
| | - In Ho Kim
- Department of Animal Resource and Science, Dankook University, Cheonan 31116, Republic of Korea;
| | - Ping Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.); (P.Y.); (X.S.); (W.E.); (P.H.)
| | - Demin Cai
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.); (P.Y.); (X.S.); (W.E.); (P.H.)
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
27
|
Sun X, Zhuang Y, Wang Y, Zhang Z, An L, Xu Q. Polyethylene terephthalate microplastics affect gut microbiota distribution and intestinal damage in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 294:118119. [PMID: 40164037 DOI: 10.1016/j.ecoenv.2025.118119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Polyethylene terephthalate microplastics (PET-MPs) have been detected in the environment and human metabolites or tissues; however, their potential effects on humans under actual exposure doses remain unclear. Herein, male adult mice were exposed to 10 µm PET-MPs at concentrations of 10, 50, and 250 mg/kg per body weight consecutively for 28 days. Changes in blood biochemistry, inflammatory factors, colonic histopathology, colonic mucus gene mRNA levels, and the gut microflora were monitored to study PET-MPs toxicity. The results showed that PET-MPs exposure increased relative serum alanine aminotransferase (ALT) and glucose (GLU) levels in 50 mg/kg bw PET-MPs exposure group, and altered relative levels of inflammatory factors, thereby inducing the inflammatory response. Moreover, PET-MPs exposure increased mRNA expression levels of colonic mucus secretion related and barrier function related genes, indicating intestinal mucus secretion and barrier integrity dysfunction, which was consistent with the results of histopathological results. In addition, gut microbiota analysis revealed that the diversity and community composition were altered after PET-MPs exposure, suggesting a metabolic disorder. Therefore, our results demonstrated that exposure to PET-MPs led to intestinal injury and changes in the gut microbiome composition in mice. Overall, the study findings provided basic data about the health risks of PET-MPs to humans, highlighting that MPs-induced toxicity warrants more concern in the future.
Collapse
Affiliation(s)
- Xiangying Sun
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environment Science, Beijing 100012, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Kaifeng Center for Disease Control and Prevention, Kaifeng, Henan 475000, China
| | - Yin Zhuang
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yubang Wang
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drug of Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Zhenbo Zhang
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Lihui An
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environment Science, Beijing 100012, China.
| | - Qiujin Xu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environment Science, Beijing 100012, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
28
|
Chen A, Peng X, Shen T, Zheng L, Wu D, Wang S. Discovery, design, and engineering of enzymes based on molecular retrobiosynthesis. MLIFE 2025; 4:107-125. [PMID: 40313979 PMCID: PMC12042125 DOI: 10.1002/mlf2.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 02/06/2025] [Accepted: 02/13/2025] [Indexed: 05/03/2025]
Abstract
Biosynthesis-a process utilizing biological systems to synthesize chemical compounds-has emerged as a revolutionary solution to 21st-century challenges due to its environmental sustainability, scalability, and high stereoselectivity and regioselectivity. Recent advancements in artificial intelligence (AI) are accelerating biosynthesis by enabling intelligent design, construction, and optimization of enzymatic reactions and biological systems. We first introduce the molecular retrosynthesis route planning in biochemical pathway design, including single-step retrosynthesis algorithms and AI-based chemical retrosynthesis route design tools. We highlight the advantages and challenges of large language models in addressing the sparsity of chemical data. Furthermore, we review enzyme discovery methods based on sequence and structure alignment techniques. Breakthroughs in AI-based structural prediction methods are expected to significantly improve the accuracy of enzyme discovery. We also summarize methods for de novo enzyme generation for nonnatural or orphan reactions, focusing on AI-based enzyme functional annotation and enzyme discovery techniques based on reaction or small molecule similarity. Turning to enzyme engineering, we discuss strategies to improve enzyme thermostability, solubility, and activity, as well as the applications of AI in these fields. The shift from traditional experiment-driven models to data-driven and computationally driven intelligent models is already underway. Finally, we present potential challenges and provide a perspective on future research directions. We envision expanded applications of biocatalysis in drug development, green chemistry, and complex molecule synthesis.
Collapse
Affiliation(s)
- Ancheng Chen
- Shanghai Zelixir Biotech Company Ltd.ShanghaiChina
| | - Xiangda Peng
- Shanghai Zelixir Biotech Company Ltd.ShanghaiChina
| | - Tao Shen
- Shanghai Zelixir Biotech Company Ltd.ShanghaiChina
| | | | - Dong Wu
- Shanghai Zelixir Biotech Company Ltd.ShanghaiChina
| | - Sheng Wang
- Shanghai Zelixir Biotech Company Ltd.ShanghaiChina
| |
Collapse
|
29
|
Zhu X, Sylvetsky AC, McCullough ML, Welsh JA, Hartman TJ, Ferranti EP, Um CY. Association of Low-Calorie Sweeteners with Selected Circulating Biomarkers of Intestinal Permeability in the Cancer Prevention Study-3 Diet Assessment Substudy. J Nutr 2025; 155:1226-1235. [PMID: 40032143 DOI: 10.1016/j.tjnut.2025.02.022] [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: 12/11/2024] [Revised: 02/14/2025] [Accepted: 02/25/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Low-calorie sweeteners (LCSs) are popular sugar substitutes and have been shown to alter the gut microbiota, which raises concerns about potential impacts on intestinal permeability. OBJECTIVES This study aimed to examine cross-sectional associations between LCS consumption and circulating biomarkers of intestinal permeability. METHODS We analyzed data from 572 United States adults participating in the Cancer Prevention Study-3 Diet Assessment Substudy who provided ≤2 fasting blood samples, collected 6 mo apart, to measure biomarkers of intestinal permeability including antibodies to flagellin (anti-flagellin), lipopolysaccharide (anti-LPS), and total antibodies; and ≤6 24-h dietary recalls, collected over the course of 12 mo, to estimate average intake of LCS including aspartame, sucralose, acesulfame-potassium, and saccharin. Multivariable linear regression, adjusted for sociodemographic characteristics, lifestyle factors, and medical history, was used to examine associations between LCS consumption and levels of intestinal permeability biomarkers by comparing mean differences in biomarkers among lower (>0 to ≤50th percentile) (n = 158) and higher (>50th percentile) LCS consumers (n = 157) than nonconsumers. A linear trend across nonconsumers and the 2 consumption categories was evaluated using a continuous variable based on the median LCS intake (median = 0, 11.3, and 124.2 mg/d for non-, lower, and higher consumers, respectively). RESULTS Among the 572 study participants, the mean age was 52.5 y, 63.3% were female, 60.7% were on-Hispanic White, and 55.1% reported consuming LCS-containing products. Greater LCS consumption was not associated with anti-flagellin, anti-LPS, or total antibodies. Additionally, no associations between specific types of LCS and intestinal permeability biomarkers were observed. CONCLUSIONS The results of our study did not demonstrate an association between LCS consumption and intestinal permeability biomarkers. Further research with larger sample sizes and randomized controlled trials is needed to confirm our findings.
Collapse
Affiliation(s)
- Xinyu Zhu
- Nutrition and Health Sciences Program, Laney Graduate School, Emory University, Atlanta, GA, United States.
| | - Allison C Sylvetsky
- Department of Exercise and Nutrition Sciences, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| | - Marjorie L McCullough
- Department of Population Science, American Cancer Society, Atlanta, GA, United States
| | - Jean A Welsh
- Nutrition and Health Sciences Program, Laney Graduate School, Emory University, Atlanta, GA, United States; Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, United States; Department of Child Advocacy, Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - Terryl J Hartman
- Nutrition and Health Sciences Program, Laney Graduate School, Emory University, Atlanta, GA, United States; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States; Winship Cancer Institute, Emory University, Atlanta, GA, United States
| | - Erin P Ferranti
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, United States
| | - Caroline Y Um
- Department of Population Science, American Cancer Society, Atlanta, GA, United States.
| |
Collapse
|
30
|
Wei ZX, Jiang SH, Qi XY, Cheng YM, Liu Q, Hou XY, He J. scRNA-seq of the intestine reveals the key role of mast cells in early gut dysfunction associated with acute pancreatitis. World J Gastroenterol 2025; 31:103094. [PMID: 40182603 PMCID: PMC11962851 DOI: 10.3748/wjg.v31.i12.103094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/09/2025] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Intestinal barrier dysfunction is a prevalent and varied manifestation of acute pancreatitis (AP). Molecular mechanisms underlying the early intestinal barrier in AP remain poorly understood. AIM To explore the biological processes and mechanisms of intestinal injury associated with AP, and to find potential targets for early prevention or treatment of intestinal barrier injury. METHODS This study utilized single-cell RNA sequencing of the small intestine, alongside in vitro and in vivo experiments, to examine intestinal barrier function homeostasis during the early stages of AP and explore involved biological processes and potential mechanisms. RESULTS Seventeen major cell types and 33232 cells were identified across all samples, including normal, AP1 (4x caerulein injections, animals sacrificed 2 h after the last injection), and AP2 (8x caerulein injections, animals sacrificed 4 h after the last injection). An average of 980 genes per cell was found in the normal intestine, compared to 927 in the AP1 intestine and 1382 in the AP2 intestine. B cells, dendritic cells, mast cells (MCs), and monocytes in AP1 and AP2 showed reduced numbers compared to the normal intestine. Enterocytes, brush cells, enteroendocrine cells, and goblet cells maintained numbers similar to the normal intestine, while cytotoxic T cells and natural killer (NK) cells increased. Enterocytes in early AP exhibited elevated programmed cell death and intestinal barrier dysfunction but retained absorption capabilities. Cytotoxic T cells and NK cells showed enhanced pathogen-fighting abilities. Activated MCs, secreted chemokine (C-C motif) ligand 5 (CCL5), promoted neutrophil and macrophage infiltration and contributed to barrier dysfunction. CONCLUSION These findings enrich our understanding of biological processes and mechanisms in AP-associated intestinal injury, suggesting that CCL5 from MCs is a potential target for addressing dysfunction.
Collapse
Affiliation(s)
- Zu-Xing Wei
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Shi-He Jiang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Xiao-Yan Qi
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Yi-Miao Cheng
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Qiong Liu
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Xu-Yang Hou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Jun He
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| |
Collapse
|
31
|
Perumal SK, Arumugam MK, Osna NA, Rasineni K, Kharbanda KK. Betaine regulates the gut-liver axis: a therapeutic approach for chronic liver diseases. Front Nutr 2025; 12:1478542. [PMID: 40196019 PMCID: PMC11973089 DOI: 10.3389/fnut.2025.1478542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
Chronic liver disease is defined by persistent harm to the liver that might result in decreased liver function. The two prevalent chronic liver diseases are alcohol-associated liver disease (ALD) and metabolic dysfunction-associated steatotic liver disease (MASLD). There is ample evidence that the pathogenesis of these two chronic liver diseases is closely linked to gastrointestinal dysfunctions that alters the gut-liver crosstalk. These alterations are mediated through the imbalances in the gut microbiota composition/function that combined with disruption in the gut barrier integrity allows for harmful gut microbes and their toxins to enter the portal circulation and reach the liver to elicit an inflammatory response. This leads to further recruitment of systemic inflammatory cells, such as neutrophils, T-cells, and monocytes into the liver, which perpetuate additional inflammation and the development of progressive liver damage. Many therapeutic modalities, currently used to prevent, attenuate, or treat chronic liver diseases are aimed at modulating gut dysbiosis and improving intestinal barrier function. Betaine is a choline-derived metabolite and a methyl group donor with antioxidant, anti-inflammatory and osmoprotectant properties. Studies have shown that low betaine levels are associated with higher levels of organ damage. There have been several publications demonstrating the role of betaine supplementation in preventing the development of ALD and MASLD. This review explores the protective effects of betaine through its role as a methyl donor and its capacity to regulate the protective gut microbiota and maintain intestinal barrier integrity to prevent the development of these chronic liver diseases. Further studies are needed to enhance our understanding of its therapeutic potential that could pave the way for targeted interventions in the management of not only chronic liver diseases, but other inflammatory bowel diseases or systemic inflammatory conditions.
Collapse
Affiliation(s)
- Sathish Kumar Perumal
- Research Service, Department of Veterans Affairs, Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Madan Kumar Arumugam
- Research Service, Department of Veterans Affairs, Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Cancer Biology Lab, Centre for Molecular and Nanomedical Sciences, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Natalia A. Osna
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Karuna Rasineni
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kusum K. Kharbanda
- Research Service, Department of Veterans Affairs, Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
32
|
Liao J, Wang M, Li H, Li T, Deng Z, Li J, Zheng L, Yan Y, Duan S, Zhang B. Human Milk Oligosaccharide LNnT Promotes Intestinal Epithelial Growth and Maturation During the Early Life of Infant Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6678-6690. [PMID: 40048505 DOI: 10.1021/acs.jafc.4c10055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Lacto-N-neotetraose (LNnT) is a prevalent neutral core human milk oligosaccharides (HMOs) recognized for its numerous benefits to infant health. In infant formula, galactooligosaccharide (GOS) are frequently used as substitutes for HMOs. However, the regulatory roles of LNnT and GOS in early intestinal development are not yet fully understood. This study aims to elucidate the effects of LNnT and GOS on intestinal development during early life. Our findings show that administering LNnT or GOS significantly increased the spleen and liver indices of infant mice at postnatal day 21. Immunofluorescence and qPCR analysis showed that feeding LNnT significantly promoted the proliferation and differentiation of intestinal stem cells (ISCs) in the colon of infant mice at postnatal day 21, and increased the expression of differentiation markers of goblet cells, intestinal epithelial cells, Paneth cells, and intestinal endocrine cells. Conversely, feeding GOS had no significant effect on the proliferation and differentiation of ISCs. Furthermore, intestinal microbiota analysis showed that LNnT increased the microbiota associated with intestinal regeneration and ISCs proliferation and differentiation in infant mice at postnatal day 21. In conclusion, LNnT promoted ISCs proliferation and differentiation in the colon and alters the composition and function of the intestinal microbiota to support intestinal development in infant mice.
Collapse
Affiliation(s)
- Jinqiang Liao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
| | - Minghui Wang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
| | - Hongyan Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051 Jiangxi, China
| | - Ting Li
- Yili Maternal and Infant Nutrition Institute (YMINI), Inner Mongolia Yili Industrial Group, Co. Ltd, Beijing 100070, China
- Inner Mongolia Dairy Technology Research Institute Co. Ltd, Hohhot 010110, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051 Jiangxi, China
| | - Jing Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051 Jiangxi, China
| | - Liufeng Zheng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051 Jiangxi, China
| | - Yalu Yan
- Yili Maternal and Infant Nutrition Institute (YMINI), Inner Mongolia Yili Industrial Group, Co. Ltd, Beijing 100070, China
- Inner Mongolia Dairy Technology Research Institute Co. Ltd, Hohhot 010110, China
| | - Sufang Duan
- Yili Maternal and Infant Nutrition Institute (YMINI), Inner Mongolia Yili Industrial Group, Co. Ltd, Beijing 100070, China
- Inner Mongolia Dairy Technology Research Institute Co. Ltd, Hohhot 010110, China
| | - Bing Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047 Jiangxi, China
- International Institute of Food Innovation, Nanchang University, Nanchang 330051 Jiangxi, China
| |
Collapse
|
33
|
Liu J, Dai Y, Yang W, Chen ZY. Role of Mushroom Polysaccharides in Modulation of GI Homeostasis and Protection of GI Barrier. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6416-6441. [PMID: 40063730 PMCID: PMC11926878 DOI: 10.1021/acs.jafc.5c00745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/25/2025] [Accepted: 02/28/2025] [Indexed: 03/20/2025]
Abstract
Edible and medicinal mushroom polysaccharides (EMMPs) have been widely studied for their various biological activities. It has been shown that EMMPs could modulate microbiota in the large intestine and improve intestinal health. However, the role of EMMPs in protecting the gastric barrier, regulating gastric microbiota, and improving gastric health cannot be ignored. Hence, this review will elucidate the effect of EMMPs on gastric and intestinal barriers, with emphasis on the interaction of EMMPs with microbiota in maintaining overall gastrointestinal health. Additionally, this review highlights the gastroprotective effects and underlying mechanisms of EMMPs against gastric mucosa injury, gastritis, gastric ulcer, and gastric cancer. Furthermore, the effects of EMMPs on intestinal diseases, including inflammatory bowel disease, colorectal cancer, and intestinal infection, are also summarized. This review will also discuss the future perspective and challenges in the use of EMMPs as a dietary supplement or a nutraceutical in preventing and treating gastrointestinal diseases.
Collapse
Affiliation(s)
- Jianhui Liu
- Collaborative
Innovation Center for Modern Grain Circulation and Safety, Jiangsu
Province Engineering Research Center of Edible Fungus Preservation
and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
- School
of Life Sciences, The Chinese University
of Hong Kong, Shatin, NT, Hong Kong 999077, China
| | - Yi Dai
- Collaborative
Innovation Center for Modern Grain Circulation and Safety, Jiangsu
Province Engineering Research Center of Edible Fungus Preservation
and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Wenjian Yang
- Collaborative
Innovation Center for Modern Grain Circulation and Safety, Jiangsu
Province Engineering Research Center of Edible Fungus Preservation
and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Zhen-Yu Chen
- School
of Life Sciences, The Chinese University
of Hong Kong, Shatin, NT, Hong Kong 999077, China
| |
Collapse
|
34
|
Mallardi D, Maqoud F, Guido D, Aloisio M, Linsalata M, Russo F. Mapping Research Trends on Intestinal Permeability in Irritable Bowel Syndrome with a Focus on Nutrition: A Bibliometric Analysis. Nutrients 2025; 17:1064. [PMID: 40292517 PMCID: PMC11945834 DOI: 10.3390/nu17061064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/14/2025] [Accepted: 03/16/2025] [Indexed: 04/30/2025] Open
Abstract
Irritable Bowel Syndrome (IBS) is a complex gastrointestinal disorder characterized by chronic abdominal pain and altered bowel habits, often linked to disruptions in intestinal barrier function. Increased intestinal permeability plays a key role in IBS pathogenesis, affecting immune responses, gut microbiota, and inflammation. This study conducts a bibliometric analysis to explore global research trends on intestinal permeability in IBS, focusing on key contributors, collaboration networks, and thematic shifts, particularly the interplay between the intestinal barrier, gut microbiota, and dietary components. A total of 411 articles were retrieved from Scopus, with 232 studies analyzed using Bibliometrix in R. To optimize screening, ASReview, a machine learning tool, was employed, utilizing the Naïve Bayes algorithm combined with Term Frequency-Inverse Document Frequency (TF-IDF) for adaptive ranking of articles by relevance. This approach significantly improved screening step efficacy. The analysis highlights growing research interest, with China and the USA as leading contributors. Key themes include the role of gut microbiota in modulating permeability, the impact of dietary components (fiber, probiotics, bioactive compounds) on tight junction integrity, and the exploration of therapeutic agents. Emerging studies suggest integrating gut barrier modulation with nutritional and microbiome-targeted strategies for IBS management. This study provides a comprehensive overview of research on intestinal permeability in IBS, mapping its evolution and identifying major trends. By highlighting key contributors and thematic areas, it offers insights to guide future investigations into the interplay between gut permeability, diet, and microbiota, advancing understanding of IBS pathophysiology and management.
Collapse
Affiliation(s)
- Domenica Mallardi
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (D.M.); (F.M.); (M.A.); (M.L.)
| | - Fatima Maqoud
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (D.M.); (F.M.); (M.A.); (M.L.)
| | - Davide Guido
- Data Science Unit, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy;
| | - Michelangelo Aloisio
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (D.M.); (F.M.); (M.A.); (M.L.)
| | - Michele Linsalata
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (D.M.); (F.M.); (M.A.); (M.L.)
| | - Francesco Russo
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (D.M.); (F.M.); (M.A.); (M.L.)
| |
Collapse
|
35
|
Gu C, Sha G, Zeng B, Cao H, Cao Y, Tang D. Therapeutic potential of fecal microbiota transplantation in colorectal cancer based on gut microbiota regulation: from pathogenesis to efficacy. Therap Adv Gastroenterol 2025; 18:17562848251327167. [PMID: 40104324 PMCID: PMC11915259 DOI: 10.1177/17562848251327167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related deaths worldwide, with its progression intricately linked to gut microbiota dysbiosis. Disruptions in microbial homeostasis contribute to tumor initiation, immune suppression, and inflammation, establishing the microbiota as a key therapeutic target. Fecal microbiota transplantation (FMT) has emerged as a transformative approach to restore microbial balance, enhance immune responses, and reshape the tumor microenvironment. This review explores the mechanisms underlying FMT's therapeutic potential, evaluates its advantages over other microbiota-based interventions, and addresses challenges such as donor selection, safety concerns, and treatment standardization. Looking forward, the integration of FMT into personalized CRC therapies requires robust clinical trials and the identification of predictive biomarkers to optimize its efficacy and safety.
Collapse
Affiliation(s)
- Chen Gu
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Gengyu Sha
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Binbin Zeng
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Herong Cao
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yibo Cao
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dong Tang
- Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou 225000, China
- The Yangzhou Clinical College of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, 221000, China
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People’s Hospital, Yangzhou University, Yangzhou, 225000, China
- Northern Jiangsu People’s Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Nanjing, 210000, China
| |
Collapse
|
36
|
Li Z, Chu T, Sun X, Zhuang S, Hou D, Zhang Z, Sun J, Liu Y, Li J, Bian Y. Polyphenols-rich Portulaca oleracea L. (purslane) alleviates ulcerative colitis through restiring the intestinal barrier, gut microbiota and metabolites. Food Chem 2025; 468:142391. [PMID: 39675274 DOI: 10.1016/j.foodchem.2024.142391] [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: 11/03/2024] [Accepted: 12/04/2024] [Indexed: 12/17/2024]
Abstract
Ulcerative colitis (UC) is a recurrent intestinal disease caused by a complex of factors, and there are serious adverse effects and tolerance problems associated with the current long-term use of therapeutic drugs. The development of natural food sources and multi-targeted drugs for the treatment of UC is imminent. Portulaca oleracea L. (PO), as a vegetable, has been shown in studies to have an anti-UC effects. However, the relationship between the abundant active ingredients contained in Portulaca oleracea L. and the improvement of intestinal barrier, gut microbiota and metabolites is unclear. In the present study, Portulaca oleracea L. which was found to be rich in phenolic acid-based active ingredients, were effective in alleviating dextran sulfate sodium (DSS)-induced body weight loss, disease activity index (DAI) score and colon length in mice. It also decreased C-reactive protein (CRP) and myeloperoxidase (MPO) responses, reduced the permeation of fluorescein isothiocyanate (FITC)-dextran, lipopolysaccharide (LPS) and evans blue (EB), and improved histopathological scores. Meanwhile, in vitro and in vivo validation revealed the protective effects of purslane on the intestinal barrier indicators ZO-1, Occludin and Claudin-1, and inhibited the expression of inflammation-associated iNOS and NLRP3 proteins through the NF-κB signaling pathway. In addition, purslane increased the diversity of the intestinal flora, enhancing the proportion of the genera Butyricoccus, Dorea and Bifidobacterium and decreasing the percentage of Bacteroides, Turicibacter and Parabacteroides. Serum metabolomics analysis showed that the imbalance of 39 metabolites was significantly reversed after PO deployment. Enrichment analysis showed that Pentose phosphate pathway and Pyruvate metabolism pathway were the key pathways of PO against UC. Overall, purslane effectively improved the intestinal barrier disruption and intestinal inflammation by inhibiting the NF-κB signaling pathway, and adjusted the disorder of gut microbiota and metabolites to exert anti-UC effects.
Collapse
Affiliation(s)
- Zheng Li
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Tianjiao Chu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xin Sun
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Shen Zhuang
- College of Veterinary Medicine & Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Dianbo Hou
- School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zhaohan Zhang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jialu Sun
- School of Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yuhong Liu
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Jing Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Yifei Bian
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| |
Collapse
|
37
|
Hou XW, Meng J, Chen XT, Zhao JX, Shang KM, Wei YJ, Liu R. Bacillus safensis M01 reversed the inflammatory injury of mice jejunum caused by enterotoxigenic Escherichia coli K88. Arch Microbiol 2025; 207:87. [PMID: 40087175 DOI: 10.1007/s00203-025-04287-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/15/2025] [Accepted: 02/24/2025] [Indexed: 03/17/2025]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a major pathogen causing neonatal diarrhea in livestock, with antibiotics commonly used for control. However, antibiotic overuse has led to issues such as residues and bacterial resistance, underscoring the need for alternative prevention strategies. This study investigated the potential of Bacillus safensis (B. safensis) M01, isolated from healthy porcine feces in Shandong, China, to prevent ETEC infections. M01 exhibited over 80% inhibition of ETEC in vitro and was selected for further analysis. Pre-treatment of IPEC-J2 cells with M01 significantly reduced ETEC-induced cellular damage, enhanced cell viability, and inhibited bacterial adhesion. It modulated inflammatory responses by down-regulating IL-1β and TNF-α while up-regulating IL-10. Additionally, M01 promoted the expression of tight junction proteins, including Claudin-1, Occludin, and ZO-1. In the C57BL/6 mouse model, pre-feeding with M01 for 14 days improved jejunal injury caused by ETEC, as indicated by increased villus height/crypt depth ratios. Similar to in vitro findings, M01 reduced IL-1β and TNF-α expression while enhancing tight junction protein levels. These results suggest that B. safensis M01 is a promising probiotic candidate for preventing ETEC infections in livestock, offering an effective alternative to antibiotics.
Collapse
Affiliation(s)
- Xin-Wen Hou
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China
| | - Jinxin Meng
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China
| | - Xiao-Tong Chen
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China
| | - Ji-Xin Zhao
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China
| | - Kai-Meng Shang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China
| | - Yong-Jie Wei
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China
| | - Rui Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, People's Republic of China.
| |
Collapse
|
38
|
Li Y, Zhang Q, Liu X, Wang Y, Yang C, Wu Y, Xiao B, Feng Y, Wu A, Yi J, Wu J, Liang Z, Yuan Z. Citrinin-Induced Intestinal Onset of Pyroptosis via the IP3R1-GRP75-VDAC1 Complex-Mediated Mitochondrial Oxidative Stress. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:5803-5815. [PMID: 40000072 DOI: 10.1021/acs.jafc.4c11218] [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: 02/27/2025]
Abstract
Citrinin (CTN) is commonly found in animal feed and stored grains and poses a serious threat to human and animal health. Formation of the IP3R1-GRP75-VDAC1 complex has been shown to play a key role in intestinal defense against harmful stimuli, but the mechanism of its action in CTN-exposure-induced enterotoxicity is not clear. Therefore, the aim of this study was to investigate the role of the IP3R1-GRP75-VDAC1 complex in CTN-exposure-induced intestinal and IPEC-J2 monolayer cell damage in mice. It was shown that CTN exposure triggered intestinal cell pyroptosis and increased IP3R1-GRP75-VDAC1 complex formation as well as mitochondrial levels of calcium ions and mitochondrial reactive oxygen species (mtROS). And mtROS is considered to be a key factor in cellular pyroptosis. Therefore, the removal of mtROS by using Mito-Tempo was found to attenuate CTN-exposure-induced cellular pyroptosis but failed to attenuate mitochondrial calcium ion overload. However, silencing of GRP75 alleviated CTN-exposure-induced increases in the level of mtROS, mitochondrial calcium ions, and subsequent cellular pyroptosis. Therefore, this study confirms that CTN exposure induces cellular juxtaposition in intestinal tissues and points out that mitochondrial oxidative stress mediated by the IP3R1-GRP75-VDAC1 complex is a key mechanism by which CTN exposure triggers intestinal cellular pyroptosis.
Collapse
Affiliation(s)
- Yuanyuan Li
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - Qike Zhang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - Xiaofang Liu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - Yongkang Wang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - Chenglin Yang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - You Wu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - Bo Xiao
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - Yiya Feng
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - Aoao Wu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - Jine Yi
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - Jing Wu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
- Institute of Yunnan Circular Agricultural Industry, Puer 665000, P. R. China
| | - Zengenni Liang
- Dongting Laboratory, Hunan Provincial Key Laboratory for Fruits and Vegetables Storage Processing and Quality Safety, Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, P. R. China
- Yulushan Laboratory, Changsha 410128, P. R. China
| | - Zhihang Yuan
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
- Institute of Yunnan Circular Agricultural Industry, Puer 665000, P. R. China
| |
Collapse
|
39
|
Cai C, Yang P, Shi Y, Wang X, Chen G, Zhang Q, Cheng G, Kong W, Xu Z. Transcriptomic and metabolomic analysis revealed potential mechanisms of growth and disease resistance dimorphism in male and female common carp (Cyprinus carpio). FISH & SHELLFISH IMMUNOLOGY 2025; 158:110150. [PMID: 39842680 DOI: 10.1016/j.fsi.2025.110150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/16/2025] [Accepted: 01/19/2025] [Indexed: 01/24/2025]
Abstract
Sexual dimorphism is well-documented in aquaculture, particularly regarding growth differences, wherein one sex often grows faster than the other. However, despite the phenomenon being so widely documented, its underlying molecular mechanisms remain poorly understood. As an important digestive and immune organ, the gut plays key roles in the regulation of fish growth. In this study, we conducted RNA-seq and metabolomic analysis on the gut of female and male common carp. We discovered that growth-related pathways, such as "Glycolysis/Gluconeogenesis" and "Riboflavin metabolism" are significantly enriched in the gut of female carp. Conversely, pathways linked to disease resistance, such as "Th17 cell differentiation" and "Autophagy-animal" are predominantly enriched in male carp. Following intraperitoneal injection of spring viraemia of carp virus (SVCV) into both male and female carp, quantitative reverse transcription polymerase chain reaction (RT-qPCR) analysis and histopathological staining confirmed that male carp exhibit greater disease resistance compared to females. This study identified the disease resistance dimorphism in common carp and specific mechanisms underlying growth differences. Our findings offer valuable insights for the application of growth dimorphism and disease-resistant breeding in fish.
Collapse
Affiliation(s)
- Chang Cai
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Peng Yang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Yong Shi
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Xinyou Wang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Guanghui Chen
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Qianqian Zhang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Gaofeng Cheng
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Weiguang Kong
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Zhen Xu
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.
| |
Collapse
|
40
|
Kim H, Girardi G, Pickle A, Kim TS, Seker E. Microfluidic tools to model, monitor, and modulate the gut-brain axis. BIOMICROFLUIDICS 2025; 19:021301. [PMID: 40060273 PMCID: PMC11890156 DOI: 10.1063/5.0253041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 02/12/2025] [Indexed: 03/24/2025]
Abstract
The gut-brain axis (GBA) connects the gastrointestinal tract and the central nervous system (CNS) via the peripheral nervous system and humoral (e.g., circulatory and lymphatic system) routes. The GBA comprises a sophisticated interaction between various mammalian cells, gut microbiota, and systemic factors. This interaction shapes homeostatic and pathophysiological processes and plays an important role in the etiology of many disorders including neuropsychiatric conditions. However, studying the underlying processes of GBA in vivo, where numerous confounding factors exist, is challenging. Furthermore, conventional in vitro models fall short of capturing the GBA anatomy and physiology. Microfluidic platforms with integrated sensors and actuators are uniquely positioned to enhance in vitro models by representing the anatomical layout of cells and allowing to monitor and modulate the biological processes with high spatiotemporal resolution. Here, we first briefly describe microfluidic technologies and their utility in modeling the CNS, vagus nerve, gut epithelial barrier, blood-brain barrier, and their interactions. We then discuss the challenges and opportunities for each model, including the use of induced pluripotent stem cells and incorporation of sensors and actuator modalities to enhance the capabilities of these models. We conclude by envisioning research directions that can help in making the microfluidics-based GBA models better-suited to provide mechanistic insight into pathophysiological processes and screening therapeutics.
Collapse
Affiliation(s)
- Hyehyun Kim
- Department of Biomedical Engineering, University of California—Davis, Davis, California 95616, USA
| | | | - Allison Pickle
- Department of Biomedical Engineering, University of California—Davis, Davis, California 95616, USA
| | - Testaverde S. Kim
- Department of Biomedical Engineering, University of California—Davis, Davis, California 95616, USA
| | - Erkin Seker
- Department of Electrical and Computer Engineering, University of California—Davis, Davis, California 95616, USA
| |
Collapse
|
41
|
Conway J, De Jong EN, White AJ, Dugan B, Rees NP, Parnell SM, Lamberte LE, Sharma‐Oates A, Sullivan J, Mauro C, van Schaik W, Anderson G, Bowdish DME, Duggal NA. Age-related loss of intestinal barrier integrity plays an integral role in thymic involution and T cell ageing. Aging Cell 2025; 24:e14401. [PMID: 39547946 PMCID: PMC11896561 DOI: 10.1111/acel.14401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/29/2024] [Accepted: 10/18/2024] [Indexed: 11/17/2024] Open
Abstract
The intestinal epithelium serves as a physical and functional barrier against harmful substances, preventing their entry into the circulation and subsequent induction of a systemic immune response. Gut barrier dysfunction has recently emerged as a feature of ageing linked to declining health, and increased intestinal membrane permeability has been shown to promote heightened systemic inflammation in aged hosts. Concurrent with age-related changes in the gut microbiome, the thymic microenvironment undergoes a series of morphological, phenotypical and architectural alterations with age, including disorganisation of the corticomedullary junction, increased fibrosis, increased thymic adiposity and the accumulation of senescent cells. However, a direct link between gut barrier dysbiosis and thymic involution leading to features of immune ageing has not been explored thus far. Herein, we reveal strong associations between enhanced microbial translocation and the peripheral accumulation of terminally differentiated, senescent and exhausted T cells and the compensatory expansion of regulatory T cells in older adults. Crucially, we demonstrate that aged germ-free mice are protected from age-related increases in intestinal permeability, highlighting the direct impact of mucosal permeability on thymic ageing. Together, these findings establish a novel mechanism by which gut barrier dysfunction drives systemic activation of the immune system during ageing through thymic involution. This enhances our understanding of drivers of T cell ageing and opens up the possibility for the use of microbiome-based interventions to restore immune homeostasis and promote healthy ageing in older adults.
Collapse
Affiliation(s)
- Jessica Conway
- Institute of Inflammation and AgeingUniversity of BirminghamBirminghamUK
| | | | - Andrea J. White
- Institute for Immunology and ImmunotherapyUniversity of BirminghamBirminghamUK
| | - Ben Dugan
- Institute of Inflammation and AgeingUniversity of BirminghamBirminghamUK
| | - Nia Paddison Rees
- Institute of Inflammation and AgeingUniversity of BirminghamBirminghamUK
| | - Sonia M. Parnell
- Institute for Immunology and ImmunotherapyUniversity of BirminghamBirminghamUK
| | - Lisa E. Lamberte
- Institute of Microbiology and InfectionUniversity of BirminghamUK
| | | | - Jack Sullivan
- Institute of Inflammation and AgeingUniversity of BirminghamBirminghamUK
| | - Claudio Mauro
- Institute of Inflammation and AgeingUniversity of BirminghamBirminghamUK
| | | | - Graham Anderson
- Institute for Immunology and ImmunotherapyUniversity of BirminghamBirminghamUK
| | | | - Niharika A. Duggal
- Institute of Inflammation and AgeingUniversity of BirminghamBirminghamUK
| |
Collapse
|
42
|
Chen J, Lei KW, Li SY, Li DP, Wang YL, Wang X, Bai X, Huang YL. Dose effects of iron on growth, antioxidant potential, intestinal morphology, and intestinal barrier in yellow-feathered broilers. Poult Sci 2025; 104:104865. [PMID: 39919560 PMCID: PMC11851222 DOI: 10.1016/j.psj.2025.104865] [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: 10/22/2024] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 02/09/2025] Open
Abstract
This experiment was conducted to investigate the dose effects of iron on growth performance, antioxidant function, small intestinal histology, and intestinal barrier of 63-day-old yellow-feathered broilers. A total of 720 1-day-old male yellow-feathered broilers were randomly divided into 9 treatments, with 8 cages per treatment and 10 birds per cage. The Fe supplementation was 0, 20, 40, 60, 80, 160, 320, 640, and 1280 mg/kg, respectively, in the form of FeSO4•7H2O. The results showed that the ADG (P = 0.002) and ADFI (P < 0.001) decreased linearly with increased dietary Fe supplementation. Malondialdehyde (MDA) concentration in plasma (P = 0.001), duodenum (P < 0.001), and jejunum (P < 0.001) were increased linearly as dietary Fe increased. As dietary Fe increased, there was a linear decrease in the villus height and the villus height/crypt depth in the duodenum (P = 0.003; P = 0.001) and jejunum (P = 0.001; P < 0.001). Decreased secretory immunoglobulin A (sIgA) concentration in jejunal mucosa (P < 0.001) was observed with increased dietary Fe concentration. Lower jejunal sIgA concentrations were observed in birds consuming more than 160 mg/kg of Fe (P < 0.001). A quadratic response was found for jejunal diamine oxidase (DAO) activity (P = 0.011) as dietary Fe supplementation was increased. The highest response of DAO in jejunal mucosa was observed for broilers supplemented with 160 mg/kg of Fe. Furthermore, the mRNA expressions of ZO-1 (P < 0.001), occludin (P = 0.004), and claudin-1 (P = 0.007) in jejunal mucosa decreased linearly with increased dietary Fe concentration. Data from the study suggests that there is no need to supplement additional Fe to a corn-soybean-based diet for yellow-feathered broilers based on growth performance, antioxidant potential, small intestinal histology, and intestinal barrier. Chronic iron exposure (≥ 160 mg/kg) can damage the intestinal barrier function, and further increase of Fe supplementation can lead to oxidative stress and even cause growth inhibition for yellow-feathered broilers.
Collapse
Affiliation(s)
- J Chen
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - K W Lei
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - S Y Li
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - D P Li
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - Y L Wang
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - X Wang
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - X Bai
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - Y L Huang
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China.
| |
Collapse
|
43
|
Ma G, Chen Z, Xie Z, Liu J, Xiao X. Mechanisms underlying changes in intestinal permeability during pregnancy and their implications for maternal and infant health. J Reprod Immunol 2025; 168:104423. [PMID: 39793281 DOI: 10.1016/j.jri.2025.104423] [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: 08/31/2024] [Revised: 12/01/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
Proper regulation of intestinal permeability is essential for maintaining the integrity of the intestinal mucosal barrier. An abnormal increase in permeability can significantly contribute to the onset and progression of various diseases, including autoimmune disorders, metabolic conditions, allergies, and inflammatory bowel diseases. The potential connection between intestinal permeability and maternal health during pregnancy is increasingly recognized, yet a comprehensive review remains lacking. Pregnancy triggers a series of physiological structural adaptations and significant hormonal fluctuations that collectively contribute to an increase in intestinal permeability. Although an increase in intestinal permeability is typically a normal physiological response during pregnancy, an abnormal rise is associated with immune dysregulation, metabolic disorders, and various pregnancy-related complications, such as recurrent pregnancy loss, gestational diabetes mellitus, overweight and obesity during pregnancy, intrahepatic cholestasis of pregnancy, and preeclampsia. This paper discusses the components of the intestinal mucosal barrier, the concept of intestinal permeability and its measurement methods, and the mechanisms and physiological significance of increased intestinal permeability during pregnancy. It thoroughly explores the association between abnormal intestinal permeability during pregnancy and maternal diseases, aiming to provide evidence for the pathophysiology of disease development in pregnant women. Additionally, the paper examines intervention methods, such as gut microbiota modulation and nutritional interventions, to regulate intestinal permeability during pregnancy, improve immune and metabolic states, and offer feasible strategies for the prevention and adjuvant treatment of clinical pregnancy complications.
Collapse
Affiliation(s)
- Guangyu Ma
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Zhongsheng Chen
- Department of Colorectal Cancer Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Zhuojun Xie
- General Medicine Department, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - JinXiang Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Xiaomin Xiao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China.
| |
Collapse
|
44
|
Luca BGD, Almeida PP, Junior RR, Soares DJS, Frantz EDC, Miranda-Alves L, Stockler-Pinto MB, Machado Dos Santos C, Magliano DC. Environmental contamination by bisphenols: From plastic production to modulation of the intestinal morphophysiology in experimental models. Food Chem Toxicol 2025; 197:115280. [PMID: 39923829 DOI: 10.1016/j.fct.2025.115280] [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: 09/19/2024] [Revised: 01/15/2025] [Accepted: 01/24/2025] [Indexed: 02/11/2025]
Abstract
Bisphenols are frequently found in a range of plastic products and have been associated with the development of diseases such as diabetes mellitus type 2 and obesity. These compounds are known as endocrine disruptors and have led to restrictions on their use due to their presence in the environment and their association with non-communicable chronic diseases. The gastrointestinal tract, being the primary site of food and water absorption, is particularly vulnerable to the effects of bisphenols. For this reason, a review of studies showing associations between bisphenols exposure and adverse effects in the gut microbiota, morphology tissue, gut permeability, and on the enteric nervous system was carried out. We have included perinatal studies and in different adult experimental models. The effects of bisphenol exposure on the gut microbiota are complex and varied. Bisphenol exposure generally leads to a decrease in microbial diversity and may impact the integrity of the intestinal barrier, resulting in elevated levels of inflammation, changes in morphological and metabolic characteristics of the gut, modifications in tight junction expression, and changes in goblet cell expression. In addition, bisphenol exposure in the perinatal phase can lead to important intestinal changes, including increased colonic inflammation and decreased colonic paracellular permeability.
Collapse
Affiliation(s)
- Beatriz Gouvêa de Luca
- Research Center on Morphology and Metabolism, Biomedical Institute, Federal Fluminense University, Niteroi, RJ, Brazil; Laboratory of Teaching and Research in Histology and Comparative Embryology (LEPHEC), Federal Fluminense University, Niterói, RJ, Brazil; Pathology Graduate Program, Federal Fluminense University (UFF), Niteroi, RJ, Brazil
| | - Patricia Pereira Almeida
- Pathology Graduate Program, Federal Fluminense University (UFF), Niteroi, RJ, Brazil; Nutrition Sciences Graduate Program, Federal Fluminense University (UFF), Niteroi, RJ, Brazil
| | - Reinaldo Röpke Junior
- Laboratory of Experimental Endocrinology (LEEx), Institute of Biomedical Science, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Endocrinology Graduate Program, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Brazil
| | - Débora Júlia Silva Soares
- Research Center on Morphology and Metabolism, Biomedical Institute, Federal Fluminense University, Niteroi, RJ, Brazil
| | - Eliete Dalla Corte Frantz
- Research Center on Morphology and Metabolism, Biomedical Institute, Federal Fluminense University, Niteroi, RJ, Brazil; Cardiovascular Sciences Graduate Program, Fluminense Federal University (UFF), Niteroi, RJ, Brazil
| | - Leandro Miranda-Alves
- Laboratory of Experimental Endocrinology (LEEx), Institute of Biomedical Science, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Endocrinology Graduate Program, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Brazil; Pharmacology and Medicinal Chemistry Graduate Program, Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Brazil; Morphological Sciences Graduate Program, Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Brazil
| | - Milena Barcza Stockler-Pinto
- Pathology Graduate Program, Federal Fluminense University (UFF), Niteroi, RJ, Brazil; Nutrition Sciences Graduate Program, Federal Fluminense University (UFF), Niteroi, RJ, Brazil
| | - Clarice Machado Dos Santos
- Laboratory of Teaching and Research in Histology and Comparative Embryology (LEPHEC), Federal Fluminense University, Niterói, RJ, Brazil
| | - D'Angelo Carlo Magliano
- Research Center on Morphology and Metabolism, Biomedical Institute, Federal Fluminense University, Niteroi, RJ, Brazil; Pathology Graduate Program, Federal Fluminense University (UFF), Niteroi, RJ, Brazil; Laboratory of Experimental Endocrinology (LEEx), Institute of Biomedical Science, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Endocrinology Graduate Program, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Brazil.
| |
Collapse
|
45
|
Kerezoudi EN, Zervakis GI, Pletsa V, Kyriacou A, Brummer RJ, Rangel I. Pleurotus eryngii Mushrooms Fermented with Human Fecal Microbiota Protect Intestinal Barrier Integrity: Immune Modulation and Signalling Pathways Counter Deoxycholic Acid-Induced Disruption in Healthy Colonic Tissue. Nutrients 2025; 17:694. [PMID: 40005021 PMCID: PMC11858169 DOI: 10.3390/nu17040694] [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: 01/15/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Background: This study explores the potential of the Pleurotus eryngii mushroom fermentation supernatant (FS-PEWS) as an intervention for mitigating sodium deoxycholate (SDC)-induced intestinal barrier dysfunction and inflammation. Methods: FS-PEWS was assessed for its protective effects against SDC-induced barrier dysfunction and inflammation using an in vitro Caco-2 cell model and ex vivo colonic biopsies from healthy adult donors, where barrier integrity, permeability, immunomodulation and receptor-mediated pathways were evaluated. Results: In Caco-2 cells, SDC exposure downregulated ZO-1, occludin, and claudin-1 expression, with FS-PEWS restoring ZO-1 and claudin-1 levels while maintaining cell viability. In colonic biopsies from healthy adults, FS-PEWS maintained tissue integrity and selectively mitigated transcellular permeability without affecting paracellular permeability when combined with the stressor. Additionally, FS-PEWS exhibited potent anti-inflammatory effects, reducing pro-inflammatory cytokines, e.g., TNF-α, IL-6, and IL-1β and modulating receptor-mediated pathways, i.e., TLR-4, dectin-1. Conclusions: These results demonstrate the potential of FS-PEWS to sustain intestinal barrier function and modulate immune responses under stress, highlighting its therapeutic potential for managing gut barrier dysfunction and inflammation associated with microbial metabolite-induced disruptions.
Collapse
Affiliation(s)
- Evangelia N. Kerezoudi
- Nutrition-Gut-Brain Interactions Research Centre, School of Medical Sciences, Örebro University, 70182 Örebro, Sweden; (R.J.B.); (I.R.)
- Department of Nutrition and Dietetics, Harokopio University, 17676 Athens, Greece;
| | - Georgios I. Zervakis
- Laboratory of General and Agricultural Microbiology, Department of Crop Science, Agricultural University of Athens, 11855 Athens, Greece;
| | - Vasiliki Pletsa
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece;
| | - Adamantini Kyriacou
- Department of Nutrition and Dietetics, Harokopio University, 17676 Athens, Greece;
| | - Robert J. Brummer
- Nutrition-Gut-Brain Interactions Research Centre, School of Medical Sciences, Örebro University, 70182 Örebro, Sweden; (R.J.B.); (I.R.)
| | - Ignacio Rangel
- Nutrition-Gut-Brain Interactions Research Centre, School of Medical Sciences, Örebro University, 70182 Örebro, Sweden; (R.J.B.); (I.R.)
| |
Collapse
|
46
|
Wei X, Feng X. DS0384 Alleviates Necrotizing Enterocolitis: Secretes N-carbamyl glutamic Acid and Participates in Lipid Metabolism and Lipid Peroxidation Processes. J Microbiol Biotechnol 2025; 35:e2410040. [PMID: 39947675 PMCID: PMC11879329 DOI: 10.4014/jmb.2410.10040] [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: 10/25/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 03/06/2025]
Abstract
Necrotizing enterocolitis (NEC) is a life-threatening inflammatory bowel disease linked to gut microbiome dysbiosis. This study evaluates the efficacy of Limosilactobacillus reuteri strain DS0384, which secretes N-carbamyl glutamic acid (NCG), in modulating lipid peroxidation and inflammatory pathways in NEC. After pretreatment with DS0384, NEC mouse model was induced by gavage with bacteria-containing formula. NCG levels in the ileum were measured via CE-TOFMS metabolomic analysis. Additionally, rat small intestinal epithelial IEC-6 cells were exposed to lipopolysaccharide (LPS), treated with DS0384 DNA (D-DNA), and/or transfected to overexpress fatty acid synthase (FASN) and Toll-like receptor 4 (TLR4). Lipid peroxidation, peroxidation and inflammatory factors and NF-κB pathways were analysed. Immunofluorescence was used to measure the expression levels of ZO-1 and TLR4 in the ileum. DS0384 treatment significantly reduced more histological abnormalities, apoptosis, and TLR4 expression in NEC mice, while restoring NCG levels, downregulating FASN and inhibiting lipid peroxidation and inflammation. Pre-treatment with D-DNA maintained cell vitality, reduced apoptosis, and suppressed TLR4/NF-κB-mediated inflammasome activation. Overexpression of FASN or TLR4 reversed these effects. DS0384 is a promising therapeutic against NEC, enhancing gut barrier integrity and modulating inflammatory and oxidative responses, suggesting potential clinical benefits in preventing NEC progression.
Collapse
Affiliation(s)
- Xiaofan Wei
- Department of Paediatrics, Zhongshan Hospital of Xiamen University, No.201-209, Hubin South Road, Siming District, Xiamen 361009, P.R. China
| | - Xiao Feng
- Department of Paediatrics, Zhongshan Hospital of Xiamen University, No.201-209, Hubin South Road, Siming District, Xiamen 361009, P.R. China
| |
Collapse
|
47
|
Liu D, Qu P, Huangfu Y, Wang X, Pan M, Wu Y, Mai K, Zhang W. Supplementation of recombinant human lysozyme into diets affects the growth performance, muscle quality, immunity and intestinal microbiota in large yellow croaker Larimichthyscrocea. FISH & SHELLFISH IMMUNOLOGY 2025; 157:110062. [PMID: 39617307 DOI: 10.1016/j.fsi.2024.110062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/22/2024] [Accepted: 11/29/2024] [Indexed: 12/08/2024]
Abstract
The aim of the present study was to investigate the influence of supplementation of lysozyme (LZM) into diet on the growth performance, muscle quality, immunity, intestinal microbiota in large yellow croaker (Larimichthys crocea) with initial body weight of 194.5 ± 0.27 g. After a 70-day feeding trial, 6 experimental diets with LZM supplementation at 0 (LZM0), 10 (LZM10), 30 (LZM30), 50 (LZM50), 70 (LZM70) and 90 mg/kg (LZM90) were tested. Results showed that the fish in the LZM70 group exhibited the lowest feed conversion ratio and the highest weight gain (WG), along with the highest trypsin and Na+/K+ ATPase activities in intestine (P < 0.05). The LZM activity in serum and intestine was significantly reduced in all dietary LZM supplemented groups compared to the LZM0 group (P < 0.05). Compared with that in the LZM0 group, the gene expressions of claudin 11, bcl-2, nlrp 3, tnf α, il-10 and tgf β in intestine in the LZM90 group were significantly elevated, while bax and caspase3 were significantly downregulated (P < 0.05). Meanwhile, the group supplemented with 90 mg/kg of dietary LZM also increased muscle crude lipid content, springiness and drip loss, along with decreased crude protein content, shear force and hardness compared with other groups (P < 0.05). Furthermore, the results of intestinal microbiota showed that compared to those in the LZM0 group, relative abundances of Fusobacterium in the LZM30 and LZM90 groups were decreased, and the relative abundances of Achromobacter, Mycoplasma and Cetobacterium were increased. In conclusion, appropriate supplementation of LZM in diet promoted the growth performance, improved immunity, adjusted intestinal microbiota and muscle quality of large yellow croaker. Furthermore, the optimal level of dietary LZM supplementation for large yellow croaker was estimated to be 67.14 mg/kg based on the quadratic regression for WG.
Collapse
Affiliation(s)
- Dayin Liu
- The Key Laboratory of Mariculture (Ministry of Education), Shenzhen Institute, Fisheries College, Ocean University of China, Qingdao 266003, China
| | - Peng Qu
- The Key Laboratory of Mariculture (Ministry of Education), Shenzhen Institute, Fisheries College, Ocean University of China, Qingdao 266003, China
| | - Yingxu Huangfu
- The Key Laboratory of Mariculture (Ministry of Education), Shenzhen Institute, Fisheries College, Ocean University of China, Qingdao 266003, China
| | - Xinyu Wang
- The Key Laboratory of Mariculture (Ministry of Education), Shenzhen Institute, Fisheries College, Ocean University of China, Qingdao 266003, China
| | - Mingzhu Pan
- College of Marine and Biology Engineering, Yancheng Institute of Technology, Yancheng, 224051, China
| | - Yang Wu
- The Key Laboratory of Mariculture (Ministry of Education), Shenzhen Institute, Fisheries College, Ocean University of China, Qingdao 266003, China
| | - Kangsen Mai
- The Key Laboratory of Mariculture (Ministry of Education), Shenzhen Institute, Fisheries College, Ocean University of China, Qingdao 266003, China
| | - Wenbing Zhang
- The Key Laboratory of Mariculture (Ministry of Education), Shenzhen Institute, Fisheries College, Ocean University of China, Qingdao 266003, China.
| |
Collapse
|
48
|
Sagada G, Wang L, Xu B, Sun Y, Shao Q. Interactive Effect of Dietary Heat-Killed Lactobacillus Plantarum L-137 and Berberine Supplementation on Intestinal Mucosa and Microbiota of Juvenile Black Sea Bream (Acanthopagrus Schlegelii). Probiotics Antimicrob Proteins 2025; 17:419-431. [PMID: 37740880 DOI: 10.1007/s12602-023-10153-8] [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] [Accepted: 08/23/2023] [Indexed: 09/25/2023]
Abstract
To compare the synergistic impact of dietary heat-killed Lactobacillus plantarum and berberine supplementation on intestinal health of juvenile black sea bream, the test fish (5.67 ± 0.05 g) were fed three diets: a basal control diet designated as Con; basal diet supplemented with 400 mg/kg L. plantarum, labelled LP; and basal diet supplemented with 400 mg/kg L. plantarum + 50 mg/k berberine, labelled LPBB. After 56 days of feeding, the control fish had significantly lower intestinal villus height (VH), villus surface area (VSA), and muscularis mucosae (MS) thickness than the rest of the groups (P < 0.05). The LPBB fish had significantly higher VH than the control fish, and wider MS and VSA than the rest of the groups (P < 0.05). Occludin was significantly upregulated in the LPBB fish, and heat shock protein 90 was upregulated in the control fish (P < 0.05). The abundance of Proteobacteria family was significantly higher in the intestinal microbiome of the control and LP fish, the LPBB fish had higher abundance of Cyanobacteria and Spirochaetes, and the LP group had higher Bacteroidetes abundance (P < 0.05). Potentially beneficial Delftia and Brevinema were the significantly abundant genera in the LP and LPBB fish, respectively; potentially pathogenic Elizabethkingia was abundant in the LP fish; and the control fish had higher abundance of potentially pathogenic Burkholderia-Caballeronia-Paraburkholderia (P < 0.05). According to these results, there is possible synergy between L. plantarum and berberine as dietary supplements in fostering healthy intestine for black sea bream than L. plantarum alone.
Collapse
Affiliation(s)
- Gladstone Sagada
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Lei Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
- School of Ecology and Environment, Anhui Normal University, Wuhu, 241000, China
| | - Bingying Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuxiao Sun
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qingjun Shao
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
- Ocean Academy, Zhejiang University, Zhoushan, 316021, China.
| |
Collapse
|
49
|
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
|
50
|
Liu L, Dai J, Yang Q, Lv L. A comprehensive review on anti-allergic natural bioactive compounds for combating food allergy. Food Res Int 2025; 201:115565. [PMID: 39849714 DOI: 10.1016/j.foodres.2024.115565] [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: 07/20/2024] [Revised: 11/18/2024] [Accepted: 12/28/2024] [Indexed: 01/25/2025]
Abstract
Food allergy poses a great challenge to food safety and public health worldwide. Currently, clinical symptoms are primarily managed with medications, which can lead to drug resistance, adverse effects, and disruptions in gut flora balance. As a result, there has been a focus on researching safe and effective anti-allergic natural ingredients. This paper provides a comprehensive overview of food allergy mechanisms, methods of assessment of anti-food allergy studies, and a classification of natural substances with anti-allergic properties. It also examines the anti-allergic effects of these substances on food allergies and investigates gut microbiota changes induced by these natural bioactives, highlighting their significance to food allergies.Natural actives with anti-food allergic properties may alleviate allergic reactions through multiple targets and pathways. These mechanisms include promoting a shift in the Th1/Th2 balance, reducting IgE synthesis, preventing cellular degranulation and reducing the release of allergic mediator. The gut environment is closely related to food allergy and there is a significant interaction between the two. By targeting the intestinal flora, we can adopt dietary interventions to effectively address and control food allergies. This provides valuable insights for the future development of functional foods targeting the alleviation of food allergies.
Collapse
Affiliation(s)
- Lu Liu
- School of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Jing Dai
- School of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Qingli Yang
- School of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Liangtao Lv
- School of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, PR China.
| |
Collapse
|