1
|
Lee CC, Fan H, Tsopmo A, Regenstein JM, Ashaolu TJ. Plant-based antioxidant peptides: impact on oxidative stress and gut microbiota. Crit Rev Food Sci Nutr 2025:1-24. [PMID: 40219794 DOI: 10.1080/10408398.2025.2490270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2025]
Abstract
Plant-based peptides can be obtained from natural and climate-friendly sources. These peptides show various bioactivities including antioxidant activity. Oxidative stress has an impact on the gut microbiota causing inflammation, insulin resistance, osteoporosis, cancer, and several chronic diseases like type 2 diabetes, arthritis, hypertension, and atherosclerosis. Therefore, antioxidant peptides may significantly affect oxidative stress as a potential alternative to conventional medication. The production of antioxidant peptides from plant-based protein sources through conventional and innovative approaches may provide promising strategies to improve gut microbiota. Recent studies in plant-based antioxidant peptides (PBAP) focus on their advanced identification and characterization techniques, structure-activity relationship, improvement of extraction and purification, cellular and molecular mechanisms, specific health applications in preventing and managing conditions with gut microbiota, and commercial applications in nutraceuticals. Short-chain fatty acids and reactive sulfur species are specific gut-derived metabolites that can improve metabolic function by modulating oxidative stress and the immune system. This review highlights the influence of food oxidants on the gut microbiota and PBAP-induced modulation of gut microbiota. Moreover, the production of PBAP and the challenges in their application will be discussed.
Collapse
Affiliation(s)
- Chi Ching Lee
- Department of Food Engineering, Faculty of Engineering and Natural Sciences, Istanbul Sabahattin Zaim University, Istanbul, Turkey
- Department of Food Technology and Nutrition, Faculty of Technologies, Klaipeda State University of Applied Sciences, Klaipeda, Lithuania
| | - Hongbing Fan
- Department of Animal and Food Sciences, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY, USA
| | - Apollinaire Tsopmo
- Food Science and Nutrition Program, Department of Chemistry, Carleton University, Ottawa, Canada
- Institute of Biochemistry, Carleton University, Ottawa, Canada
| | - Joe M Regenstein
- Department of Food Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA
| | - Tolulope Joshua Ashaolu
- Institute for Global Health Innovations, Duy Tan University, Da Nang, Vietnam
- Faculty of Medicine, Duy Tan University, Da Nang, Vietnam
| |
Collapse
|
2
|
Xu X, Zhang Y, Huang G, Perekatt A, Wang Y, Chen L. Advances and applications of gut organoids: modeling intestinal diseases and therapeutic development. LIFE MEDICINE 2025; 4:lnaf012. [PMID: 40276096 PMCID: PMC12018802 DOI: 10.1093/lifemedi/lnaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 03/04/2025] [Indexed: 04/26/2025]
Abstract
Gut organoids are 3D cellular structures derived from adult or pluripotent stem cells, capable of closely replicating the physiological properties of the gut. These organoids serve as powerful tools for studying gut development and modeling the pathogenesis of intestinal diseases. This review provides an in-depth exploration of technological advancements and applications of gut organoids, with a focus on their construction methods. Additionally, the potential applications of gut organoids in disease modeling, microenvironmental simulation, and personalized medicine are summarized. This review aims to offer perspectives and directions for understanding the mechanisms of intestinal health and disease as well as for developing innovative therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoting Xu
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210031, China
| | - Yuping Zhang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210031, China
| | - Guoxin Huang
- Clinical Research Center, Shantou Key Laboratory of Basic and Translational Research of Malignant Tumor, Shantou Central Hospital, Shantou 515041, China
| | - Ansu Perekatt
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ 07030, United States
| | - Yan Wang
- Center for Translation Medicine Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Lei Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210031, China
- Institute of Microphysiological Systems, Southeast University, Nanjing 211189, China
| |
Collapse
|
3
|
Pitt N, Morrissette M, Gates MF, Bargabos R, Krumpoch M, Hawkins B, Lewis K. Bacterial membrane vesicles restore gut anaerobiosis. NPJ Biofilms Microbiomes 2025; 11:48. [PMID: 40121189 PMCID: PMC11929906 DOI: 10.1038/s41522-025-00676-z] [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: 10/02/2024] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
Inflammation damages the epithelial cell barrier, allowing oxygen to leak into the lumen of the gut. Respiring E. coli and other Enterobacteriaceae produce proinflammatory lipopolysaccharide, exacerbating inflammatory bowel disease. Here we show that respiring membrane vesicles (MV) from E. coli ameliorate symptoms in a mouse model of gut inflammation. Membrane vesicle treatment diminished weight loss and limited shortening of the colon. Notably, oxygenation of the colonic epithelium was significantly decreased in animals receiving wild type MVs, but not MVs from an E. coli mutant lacking cytochromes. Metatranscriptomic analysis of the microbiome shows an increase in anaerobic Lactobacillaceae and a decrease in Enterobacteriaceae, as well as a general shift towards fermentation in MV-treated mice. This is accompanied by a decrease in proinflammatory TNF-α. We report that MVs may lead to the development of a novel type of a therapeutic for dysbiosis, and for treating IBD.
Collapse
Affiliation(s)
- Norman Pitt
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, Boston, MA, USA
| | - Madeleine Morrissette
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, Boston, MA, USA
| | - Michael F Gates
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, Boston, MA, USA
| | - Rachel Bargabos
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, Boston, MA, USA
| | - Megan Krumpoch
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, Boston, MA, USA
| | - Bryson Hawkins
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, Boston, MA, USA
| | - Kim Lewis
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, Boston, MA, USA.
| |
Collapse
|
4
|
Hollinger MK, Grayson EM, Ferreira CM, Sperling AI. Harnessing the Farm Effect: Microbial Products for the Treatment and Prevention of Asthma Throughout Life. Immunol Rev 2025; 330:e70012. [PMID: 40035333 PMCID: PMC11877632 DOI: 10.1111/imr.70012] [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: 09/19/2024] [Accepted: 02/10/2025] [Indexed: 03/05/2025]
Abstract
It has long been appreciated that farm exposure early in life protects individuals from allergic asthma. Understanding what component(s) of this exposure is responsible for this protection is crucial to understanding allergic asthma pathogenesis and developing strategies to prevent or treat allergic asthma. In this review, we introduce the concept of Farm-Friends, or specific microbes associated with both a farm environment and protection from allergic asthma. We review the mechanism(s) by which these Farm-Friends suppress allergic inflammation, with a focus on the molecule(s) produced by these Farm-Friends. Finally, we discuss the relevance of Farm-Friend administration (oral vs. inhaled) for preventing the development and severity of allergic asthma throughout childhood and adulthood. By developing a fuller understanding of which Farm-Friends modulate host immunity, a greater wealth of prophylactic and therapeutic options becomes available to counter the current allergy epidemic.
Collapse
Affiliation(s)
- Maile K. Hollinger
- Beirne B. Carter Center for Immunology ResearchUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Medicine, Pulmonary and Critical CareUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Emily M. Grayson
- Beirne B. Carter Center for Immunology ResearchUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Medicine, Pulmonary and Critical CareUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Microbiology, Immunology, and Cancer BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Caroline M. Ferreira
- Department of Medicine, Pulmonary and Critical CareUniversity of VirginiaCharlottesvilleVirginiaUSA
- Institute of Environmental, Chemistry and Pharmaceutics Sciences, Department of Pharmaceutics SciencesFederal University of São PauloSão PauloBrazil
| | - Anne I. Sperling
- Beirne B. Carter Center for Immunology ResearchUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Medicine, Pulmonary and Critical CareUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Microbiology, Immunology, and Cancer BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| |
Collapse
|
5
|
Zhang ZS, Yang A, Luo X, Zhou HN, Liu YY, Bao DQ, Zhang J, Zang JT, Li QH, Li T, Liu LM. Pericyte-derived extracellular vesicles improve vascular barrier function in sepsis via the Angpt1/PI3K/AKT pathway and pericyte recruitment: an in vivo and in vitro study. Stem Cell Res Ther 2025; 16:70. [PMID: 39940043 PMCID: PMC11823130 DOI: 10.1186/s13287-025-04201-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 01/29/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Extracellular vesicles derived from pericytes (PCEVs) have been shown to improve vascular permeability, with their therapeutic effects attributed to the presence of pro-regenerative molecules. We hypothesized that angiopoietin 1 (Angpt1) carried by PCEVs contributes to their therapeutic effects after sepsis. METHODS A cecal ligation and puncture (CLP)-induced sepsis rat model was used in vivo, and the effects of PCEVs on vascular endothelial cells were studied in vitro. First, proteomic and Gene Ontology enrichment analyses were performed to analyze the therapeutic mechanism of PCEVs, revealing that the angiogenesis-related protein Angpt1 was highly expressed in PCEVs. We then down-regulated Angpt1 in PCEVs. The role of PCEV-carried Angpt1 on intestinal barrier function, PCs recruitment, and inflammatory cytokines was measured by using septic Sprague-Dawley rats and platelet-derived growth factor receptor beta (PDGFR-β)-Cre + mT/mG transgenic mice. RESULTS PCEVs significantly improved vascular permeability, proliferation, and angiogenesis in CLP-induced gut barrier injury both in vivo and in vitro. Further studies have shown that PCEVs exert a protective effect on intestinal barrier function and PC recruitment. Additionally, PCEVs reduced serum inflammatory factor levels. Our data also demonstrated that the protein levels of phospho-PI3K and phospho-Akt both increased after PCEVs administration, whereas knocking out Angpt1 suppressed the protective effects of PCEVs through decreased activation of PI3K/Akt signaling. CONCLUSION PCEVs protect against sepsis by regulating the vascular endothelial barrier, promoting PC recruitment, protecting intestinal function, and restoring properties via activation of the Angpt1/PI3K/AKT pathway.
Collapse
Affiliation(s)
- Zi-Sen Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Ao Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xi Luo
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - He-Nan Zhou
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yi-Yan Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Dai-Qin Bao
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jie Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jia-Tao Zang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Qing-Hui Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Tao Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Liang-Ming Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
6
|
Wang J, Hu Q, Wang J, Lang L, Wei S, Li H, Jing M, Ma X, Zhao Y, Zhou X. Role of gut microbiota and fecal metabolites in the protective effect of soybean pulp-rich diet against estrogen-induced cholestasis in rats. Curr Res Food Sci 2025; 10:100990. [PMID: 39995468 PMCID: PMC11849669 DOI: 10.1016/j.crfs.2025.100990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/13/2025] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
This study was designed to explore the role of gut microbiota and its metabolites in the treatment of estrogen-induced cholestasis (EIC) in rats with a soybean pulp-rich diet and to clarify the effects of daidzein (DAI), a principal active ingredient of soybean pulp. The findings demonstrated that the soybean pulp-rich diet could relieve cholestasis by decreasing the levels of total bile acids (TBA) and alkaline phosphatase and enhancing the bile flow rate. Through gut microbiota and metabolomics analyses, it was revealed that this diet might alter the abundances of certain bacterial taxa including Akkermansia, Bacteroides, and Turicibacter, thus influencing lipid metabolism, tryptophan metabolism, and steroid metabolism, which led to disparities between the groups fed with and without the soybean pulp-rich diet. Moreover, the soybean pulp-rich diet could modulate the abundances of Prevotella spp. and Tyzzerella, reducing EIC by regulating lipid metabolism and short-chain fatty acids synthesis. Notably, DAI treatment significantly alleviated the abnormalities in serum TBA, alanine aminotransferase, and aspartate aminotransferase levels and mitigated the liver tissue damage in the EIC model. In summary, during cholestasis, variations in gut microbiota and metabolite profiles occurred. The intervention of soybean pulp affected the abundances of bacteria (such as Prevotella spp. and Tyzzerella) and regulated lipid metabolism-related pathways. Importantly, DAI was identified as a crucial component for the protective effects associated with the soybean pulp diet.
Collapse
Affiliation(s)
- Jiawei Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Pharmacy, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Qichao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianying Wang
- Department of Information, Medical Supplies Center of PLA General Hospital, Beijing, China
| | - Liwei Lang
- Department of Pharmacy, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Shizhang Wei
- Department of Pharmacy, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Haotian Li
- Department of Pharmacy, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Manyi Jing
- Department of Pharmacy, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanling Zhao
- Department of Pharmacy, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Xuelin Zhou
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Institute of Pharmaceutical Research, Tianjin, China
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Takasugi S, Iimura S, Yasuda M, Saito Y, Morifuji M. Key Taxa of the Gut Microbiome Associated with the Relationship Between Environmental Sensitivity and Inflammation-Related Biomarkers. Microorganisms 2025; 13:185. [PMID: 39858953 PMCID: PMC11767568 DOI: 10.3390/microorganisms13010185] [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: 12/12/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Individual differences in environmental sensitivity are linked to stress-related psychiatric symptoms. In previous research, we found that high environmental sensitivity can be a risk factor for increased inflammation and gut permeability, particularly when gut microbiome diversity is low. However, the specific gut bacterial taxa involved in this interaction remain unclear. As a preliminary study, this research aimed to identify the key gut microbiome taxa associated with this relationship. Environmental sensitivity, gut microbiome composition, gut permeability (lipopolysaccharide-binding protein, LBP), and inflammation (C-reactive protein, CRP) biomarkers were evaluated in 88 participants. The interaction between environmental sensitivity and the relative abundance of the family Marinifilaceae (genus Butyricimonas) was a predictor of CRP levels. Similarly, the interaction between environmental sensitivity and relative abundance of the family Barnesiellaceae (genus Coprobacter), the family Akkermansiaceae (genus Akkermansia), the genus Family XIII AD3011 group, the genus GCA-900066225, or the genus Ruminiclostridium 1 predicted LBP levels. Individuals with high environmental sensitivity exhibited elevated CRP or LBP levels when the relative abundance of these taxa was low. Conversely, highly sensitive individuals had lower CRP or LBP levels when the relative abundance of these taxa was high. This study suggests that specific taxa serve as one of the protective factors against inflammation and gut permeability in individuals with high environmental sensitivity. Further in-depth studies are needed to confirm these associations and understand the underlying mechanisms.
Collapse
Affiliation(s)
- Satoshi Takasugi
- R&D Division, Meiji Co., Ltd., 1-29-1 Nanakuni, Hachioji 192-0919, Japan
| | - Shuhei Iimura
- Faculty of Education, Soka University; 1-236 Tangi-machi, Hachioji 192-8577, Japan;
| | - Miyabi Yasuda
- Wellness Science Labs, Meiji Holdings Co., Ltd., 1-29-1 Nanakuni, Hachioji 192-0919, Japan; (M.Y.); (Y.S.); (M.M.)
| | - Yoshie Saito
- Wellness Science Labs, Meiji Holdings Co., Ltd., 1-29-1 Nanakuni, Hachioji 192-0919, Japan; (M.Y.); (Y.S.); (M.M.)
| | - Masashi Morifuji
- Wellness Science Labs, Meiji Holdings Co., Ltd., 1-29-1 Nanakuni, Hachioji 192-0919, Japan; (M.Y.); (Y.S.); (M.M.)
| |
Collapse
|
8
|
Wang R, Yu YF, Yu WR, Sun SY, Lei YM, Li YX, Lu CX, Zhai JN, Bai FR, Ren F, Huang JQ, Chen J. Roles of Probiotics, Prebiotics, and Postbiotics in B-Cell-Mediated Immune Regulation. J Nutr 2025; 155:37-51. [PMID: 39551357 DOI: 10.1016/j.tjnut.2024.11.011] [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: 08/20/2024] [Revised: 10/29/2024] [Accepted: 11/13/2024] [Indexed: 11/19/2024] Open
Abstract
Probiotics, prebiotics, and postbiotics can significantly influence B-cell-related diseases through their immunomodulatory effects. They enhance the immune system's function, particularly affecting B cells, which originate in the bone marrow and are crucial for antibody production and immune memory. These substances have therapeutic potential in managing allergies, autoimmune diseases, and inflammatory conditions by regulating the gut microbiota, strengthening epithelial barriers, and directly interacting with various components of the innate and adaptive immune systems. The review highlights the critical need for further research into the precise mechanisms through which probiotics, prebiotics, and postbiotics modulate B cells. Gaining this understanding could facilitate the development of more effective treatments for B-cell-related diseases by harnessing the immunomodulatory properties of these dietary components.
Collapse
Affiliation(s)
- Ran Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yifei F Yu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Weiru R Yu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Siyuan Y Sun
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yumei M Lei
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yixuan X Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Chenxu X Lu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Jianan N Zhai
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Feirong R Bai
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Fazheng Ren
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Jiaqiang Q Huang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China.
| | - Juan Chen
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China.
| |
Collapse
|
9
|
Xu MR, Lin CH, Wang CH, Wang SY. Investigate the metabolic changes in intestinal diseases by employing a 1H-NMR-based metabolomics approach on Caco-2 cells treated with cedrol. Biofactors 2025; 51:e2132. [PMID: 39415440 DOI: 10.1002/biof.2132] [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: 05/07/2024] [Accepted: 10/01/2024] [Indexed: 10/18/2024]
Abstract
Mitochondrial dysfunction may precipitate intestinal dysfunction, while inflammatory bowel disease manifests as a chronic inflammatory ailment affecting the gastrointestinal tract. This condition disrupts the barrier function of the intestinal epithelium and alters metabolic products. Increasing mitochondrial adenosine triphosphate (ATP) synthesis in intestinal epithelial cells presents a promising avenue for colitis treatments. Nevertheless, the impact of cedrol on ATP and the intestinal barrier remains unexplored. Hence, this study is dedicated to examining the cedrol's protective effect on an inflammatory cocktail (IC)-induced intestinal epithelial barrier dysfunction in Caco-2 cells. The finding reveals that cedrol enhances ATP content and the transepithelial electrical resistance value in the intestinal epithelial barrier. Moreover, cedrol mitigates the IC-induced decrease in the messenger ribonucleic acid (mRNA) expression of tight junction proteins (ZO-1, Occludin, and Claudin-1), thereby ameliorating intestinal epithelial barrier dysfunction. Furthermore, nuclear magnetic resonance (NMR)-based metabolomic analysis indicated that IC-exposed Caco-2 cells are restored by cedrol treatments. Notably, cedrol elevates metabolites such as amino acids, thereby enhancing the intestinal barrier. In conclusion, cedrol alleviates IC-induced intestinal epithelial barrier dysfunction by promoting ATP-dependent proliferation of Caco-2 cells and bolstering amino acid levels to sustain tight junction messenger ribonucleic acid expression.
Collapse
Affiliation(s)
- Mo-Rong Xu
- Doctoral Program in Microbial Genomics, National Chung Hsing University and Academia Sinica, Taichung, Taiwan
- Department of Forestry, National Chung Hsing University, Taichung, Taiwan
| | - Chia-Hsin Lin
- Department of Forestry, National Chung Hsing University, Taichung, Taiwan
| | - Chung Hsuan Wang
- Special Crop and Metabolome Discipline Cluster, Academy Circle Economy, National Chung Hsing University, Taichung, Taiwan
| | - Sheng-Yang Wang
- Doctoral Program in Microbial Genomics, National Chung Hsing University and Academia Sinica, Taichung, Taiwan
- Department of Forestry, National Chung Hsing University, Taichung, Taiwan
- Special Crop and Metabolome Discipline Cluster, Academy Circle Economy, National Chung Hsing University, Taichung, Taiwan
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
10
|
Sullivan JP, Jones MK. The Multifaceted Impact of Bioactive Lipids on Gut Health and Disease. Int J Mol Sci 2024; 25:13638. [PMID: 39769399 PMCID: PMC11728145 DOI: 10.3390/ijms252413638] [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: 12/02/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Bioactive lipids have a multifaceted role in health and disease and are recognized to play an important part in gut immunity and disease conditions such as inflammatory bowel disease and colon cancer. Advancements in lipidomics, enabled by mass spectrometry and chromatographic techniques, have enhanced our understanding of lipid diversity and functionality. Bioactive lipids, including short-chain fatty acids, saturated fatty acids, omega-3 fatty acids, and sphingolipids, exhibit diverse effects on inflammation and immune regulation. Short-chain fatty acids like butyrate demonstrate anti-inflammatory properties, enhancing regulatory T cell function, gut barrier integrity, and epigenetic regulation, making them promising therapeutic targets for inflammatory bowel disease and colon cancer. Conversely, saturated fatty acids promote inflammation by disrupting gut homeostasis, triggering oxidative stress, and impairing immune regulation. Omega-3 lipids counteract these effects, reducing inflammation and supporting immune balance. Sphingolipids exhibit complex roles, modulating immune cell trafficking and inflammation. They can exert protective effects or exacerbate colitis depending on their source and context. Additionally, eicosanoids can also prevent pathology through prostaglandin defense against damage to epithelial barriers. This review underscores the importance of dietary lipids in shaping gut health and immunity and also highlights the potential use of lipids as therapeutic strategies for managing inflammatory conditions and cancer.
Collapse
Affiliation(s)
| | - Melissa K. Jones
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL 32611, USA;
| |
Collapse
|
11
|
Horn PA, Zeni ALB, Herkenhoff ME, Curbani L, Pereira Gonçalves GH, Rutkoski CF, Israel NG, de Almeida EA. Brewer's spent yeast improves human gut microbiota and ameliorates clinical blood parameters: A randomized, double-blind, placebo-controlled trial. BIOACTIVE CARBOHYDRATES AND DIETARY FIBRE 2024; 32:100442. [DOI: 10.1016/j.bcdf.2024.100442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
12
|
Barcutean L, Farczadi L, Manescu IB, Imre S, Maier S, Balasa R. Short and Medium Chain Fatty Acids in a Cohort of Naïve Multiple Sclerosis Patients: Pre- and Post-Interferon Beta Treatment Assessment. Biologics 2024; 18:349-361. [PMID: 39569059 PMCID: PMC11577435 DOI: 10.2147/btt.s489523] [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: 07/31/2024] [Accepted: 11/06/2024] [Indexed: 11/22/2024]
Abstract
Introduction Alterations in intestinal permeability and microbiota dysregulation have been linked to the development of multiple sclerosis (MS). Short-chain fatty acids (SCFA) and medium-chain fatty acids (MCFA) are products of gut bacteria fermentation which are involved in immune regulation processes. In MS, SCFA have important immunomodulatory properties both in the periphery and the central compartment. Interferon β (IFNβ) was the first disease-modifying therapy approved for the treatment of MS and its effects on the gut microbiota are not fully elucidated. Patients and Methods We performed a prospective observational study aimed to assess peripheral levels of SCFA and MCFA in 23 newly diagnosed, treatment-naïve MS patients (nMS) before and after one year of IFNβ treatment and 23 healthy controls (HC). We investigated their associations with inflammation, interleukin-10 (IL-10), and blood-brain barrier permeability, matrix metalloproteinase 9 (MMP9). Results No significant differences in SCFA/MCFA levels were observed between baseline and after IFNβ treatment. Caproic acid levels were significantly higher in nMS compared to HC (1.64 vs 1.27 µM, p=0.005). The butyric acid/caproic acid ratio was higher in HC compared to nMS (5.47 vs 2.55, p=0.005). Correlation analysis revealed associations between SCFA/MCFA levels and inflammatory biomarkers. Conclusion nMS have a higher gut-inflammatory activity as seen by the caproic acid ratio as opposed to HC. In this cohort, IFNβ does not appear to modify the peripheral SCFA/MCFA levels after one year of treatment. The quantifications of peripheral SCFA/MCFA may prove to be a useful biomarker for gut-brain axis disruption in MS patients.
Collapse
Affiliation(s)
- Laura Barcutean
- Department of Neurology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, 540142, Romania
- Neurology 1 Clinic, Mures County Emergency Clinical Hospital, Targu Mures, Romania
| | - Lenard Farczadi
- Chromatography and Mass Spectrometry Laboratory, Center for Advanced Medical and Pharmaceutical Research, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, Mures, 540139, Romania
| | - Ion-Bogdan Manescu
- Department of Laboratory Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, 540142, Romania
| | - Silvia Imre
- Chromatography and Mass Spectrometry Laboratory, Center for Advanced Medical and Pharmaceutical Research, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, Mures, 540139, Romania
- Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, 540142, Romania
| | - Smaranda Maier
- Department of Neurology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, 540142, Romania
- Neurology 1 Clinic, Mures County Emergency Clinical Hospital, Targu Mures, Romania
| | - Rodica Balasa
- Department of Neurology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, 540142, Romania
- Neurology 1 Clinic, Mures County Emergency Clinical Hospital, Targu Mures, Romania
| |
Collapse
|
13
|
Donald K, Finlay BB. Mechanisms of microbe-mediated immune development in the context of antibiotics and asthma. FRONTIERS IN ALLERGY 2024; 5:1469426. [PMID: 39469482 PMCID: PMC11513386 DOI: 10.3389/falgy.2024.1469426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 08/28/2024] [Indexed: 10/30/2024] Open
Abstract
The gut houses 70%-80% of the body's immune cells and represents the main point of contact between the immune system and the outside world. Immune maturation occurs largely after birth and is guided by the gut microbiota. In addition to the many human clinical studies that have identified relationships between gut microbiota composition and disease outcomes, experimental research has demonstrated a plethora of mechanisms by which specific microbes and microbial metabolites train the developing immune system. The healthy maturation of the gut microbiota has been well-characterized and discreet stages marked by changes in abundance of specific microbes have been identified. Building on Chapter 8, which discusses experimental models used to study the relationship between the gut microbiota and asthma, the present review aims to dive deeper into the specific microbes and metabolites that drive key processes in immune development. The implications of microbiota maturation patterns in the context of asthma and allergies, as well as the effects of antibiotics on microbe-immune crosstalk, will also be discussed.
Collapse
Affiliation(s)
- Katherine Donald
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Departmentof Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - B. Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Departmentof Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
14
|
Dai N, Yang X, Pan P, Zhang G, Sheng K, Wang J, Liang X, Wang Y. Bacillus paralicheniformis, an acetate-producing probiotic, alleviates ulcerative colitis via protecting the intestinal barrier and regulating the NLRP3 inflammasome. Microbiol Res 2024; 287:127856. [PMID: 39079268 DOI: 10.1016/j.micres.2024.127856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 08/22/2024]
Abstract
Ulcerative colitis (UC) presents a challenging scenario in digestive health, characterized by recurrent inflammation that is often hard to manage. Bacteria capable of producing short-chain fatty acids (SCFAs) play a pivotal role in mitigating UC symptoms, rendering them promising candidates for probiotic therapy. In this investigation, we assessed the impact of Bacillus paralicheniformis HMPM220325 on dextran sodium sulfate (DSS)-induced UC in mice. Genomic analysis of the strain revealed the presence of protease genes associated with acetate and butyrate synthesis, with acetic acid detected in its fermentation broth. Administration of B. paralicheniformis HMPM220325 to UC mice ameliorated pathological manifestations of the condition and restored intestinal barrier function. Furthermore, B. paralicheniformis HMPM220325 suppressed the activation of the NLRP3 inflammasome signaling pathway and modulated the composition of the intestinal microbiota. These findings shed significant light on the potential of B. paralicheniformis as a probiotic candidate, offering a novel avenue for the prevention and therapeutic intervention of colitis.
Collapse
Affiliation(s)
- Nini Dai
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Xinting Yang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Peilong Pan
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Guanghui Zhang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Kangliang Sheng
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Jingmin Wang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Xiao Liang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China.
| | - Yongzhong Wang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China.
| |
Collapse
|
15
|
Otaru N, Bajic D, Van den Abbeele P, Vande Velde S, Van Biervliet S, Steinert RE, Rehman A. Bifidogenic Effect of Human Milk Oligosaccharides on Pediatric IBD Fecal Microbiota. Microorganisms 2024; 12:1977. [PMID: 39458287 PMCID: PMC11509818 DOI: 10.3390/microorganisms12101977] [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: 08/07/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
The prevalence of pediatric inflammatory bowel disease (pIBD) has been increasing over the last two decades. Yet, treatment strategies are still limited, in part due to the multifactorial nature of the disease and the complex interplay between genetic, environmental, dietary, immune, and gut microbial factors in its etiology. With their direct and indirect anti-inflammatory properties, human milk oligosaccharides (HMOs) are a promising treatment and management strategy for IBD. However, to date there are no insights into how HMOs may affect pIBD microbiota. Here, we compared the effects of 2'fucosyllactose (2'FL), difucosyllactose (DFL), 3'sialyllactose (3'SL), and blends thereof with fructooligosaccharide (FOS) on microbiota functionality (short- and branched-chain fatty acids, pH, and gas production) and composition (quantitative shallow shotgun sequencing) using fecal material from eight different pediatric Crohn's disease patients inoculated in the SIFR® technology. In general, all HMO treatments significantly increased total short-chain fatty acid production when compared with FOS, despite equal gas production. We found that 2'FL, either alone or in combination with DFL and 3'SL, exhibited a strong acetogenic and propiogenic effect, and 3'SL an acetogenic effect that surpassed the effects observed with FOS. No differences in overall community diversity between HMO- and FOS-treated pIBD microbiota were observed. There was, however, a stronger bifidogenic effect of 2'FL, 3'SL, 2'FL/DFL, and 2'FL/DFL + 3'SL when compared with FOS. In general, 3'SL and HMO blends enriched a broader species profile, including taxa with potentially anti-inflammatory properties, such as Faecalibacterium prausnitzii and Blautia species. This study suggests HMOs as a promising strategy to beneficially alter the gut microbial profile in pIBD.
Collapse
Affiliation(s)
- Nize Otaru
- Health, Nutrition & Care (HNC), DSM-Firmenich, 4303 Kaiseraugst, Switzerland
| | - Danica Bajic
- Health, Nutrition & Care (HNC), DSM-Firmenich, 4303 Kaiseraugst, Switzerland
| | | | - Saskia Vande Velde
- Pediatric Gastroenterology and Nutrition, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Stephanie Van Biervliet
- Pediatric Gastroenterology and Nutrition, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Robert E. Steinert
- Health, Nutrition & Care (HNC), DSM-Firmenich, 4303 Kaiseraugst, Switzerland
| | - Ateequr Rehman
- Health, Nutrition & Care (HNC), DSM-Firmenich, 4303 Kaiseraugst, Switzerland
| |
Collapse
|
16
|
da Silva VG, Smith NW, Mullaney JA, Wall C, Roy NC, McNabb WC. Food-breastmilk combinations alter the colonic microbiome of weaning infants: an in silico study. mSystems 2024; 9:e0057724. [PMID: 39191378 PMCID: PMC11406890 DOI: 10.1128/msystems.00577-24] [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: 04/22/2024] [Accepted: 07/22/2024] [Indexed: 08/29/2024] Open
Abstract
The introduction of solid foods to infants, also known as weaning, is a critical point for the development of the complex microbial community inhabiting the human colon, impacting host physiology in infancy and later in life. This research investigated in silico the impact of food-breastmilk combinations on growth and metabolite production by colonic microbes of New Zealand weaning infants using the metagenome-scale metabolic model named Microbial Community. Eighty-nine foods were individually combined with breastmilk, and the 12 combinations with the strongest influence on the microbial production of short-chain fatty acids (SCFAs) and branched-chain fatty acids (BCFAs) were identified. Fiber-rich and polyphenol-rich foods, like pumpkin and blackcurrant, resulted in the greatest increase in predicted fluxes of total SCFAs and individual fluxes of propionate and acetate when combined, respectively, with breastmilk. Identified foods were further combined with other foods and breastmilk, resulting in 66 multiple food-breastmilk combinations. These combinations altered in silico the impact of individual foods on the microbial production of SCFAs and BCFAs, suggesting that the interaction between the dietary compounds composing a meal is the key factor influencing colonic microbes. Blackcurrant combined with other foods and breastmilk promoted the greatest increase in the production of acetate and total SCFAs, while pork combined with other foods and breastmilk decreased the production of total BCFAs.IMPORTANCELittle is known about the influence of complementary foods on the colonic microbiome of weaning infants. Traditional in vitro and in vivo microbiome methods are limited by their resource-consuming concerns. Modeling approaches represent a promising complementary tool to provide insights into the behavior of microbial communities. This study evaluated how foods combined with other foods and human milk affect the production of short-chain fatty acids and branched-chain fatty acids by colonic microbes of weaning infants using a rapid and inexpensive in silico approach. Foods and food combinations identified here are candidates for future experimental investigations, helping to fill a crucial knowledge gap in infant nutrition.
Collapse
Affiliation(s)
- Vitor G da Silva
- Riddet Institute, Massey University, Palmerston North, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Nick W Smith
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Jane A Mullaney
- Riddet Institute, Massey University, Palmerston North, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
- AgResearch, Palmerston North, New Zealand
| | - Clare Wall
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
- Department of Nutrition and Dietetics, The University of Auckland, Auckland, New Zealand
| | - Nicole C Roy
- Riddet Institute, Massey University, Palmerston North, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
- Department of Human Nutrition, University of Otago, Dunedin, New Zealand
| | - Warren C McNabb
- Riddet Institute, Massey University, Palmerston North, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
| |
Collapse
|
17
|
Iacucci M, Santacroce G, Majumder S, Morael J, Zammarchi I, Maeda Y, Ryan D, Di Sabatino A, Rescigno M, Aburto MR, Cryan JF, Ghosh S. Opening the doors of precision medicine: novel tools to assess intestinal barrier in inflammatory bowel disease and colitis-associated neoplasia. Gut 2024; 73:1749-1762. [PMID: 38851294 PMCID: PMC11422792 DOI: 10.1136/gutjnl-2023-331579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/18/2024] [Indexed: 06/10/2024]
Abstract
Mounting evidence underscores the pivotal role of the intestinal barrier and its convoluted network with diet and intestinal microbiome in the pathogenesis of inflammatory bowel disease (IBD) and colitis-associated colorectal cancer (CRC). Moreover, the bidirectional association of the intestinal barrier with the liver and brain, known as the gut-brain axis, plays a crucial role in developing complications, including extraintestinal manifestations of IBD and CRC metastasis. Consequently, barrier healing represents a crucial therapeutic target in these inflammatory-dependent disorders, with barrier assessment predicting disease outcomes, response to therapy and extraintestinal manifestations.New advanced technologies are revolutionising our understanding of the barrier paradigm, enabling the accurate assessment of the intestinal barrier and aiding in unravelling the complexity of the gut-brain axis. Cutting-edge endoscopic imaging techniques, such as ultra-high magnification endocytoscopy and probe-based confocal laser endomicroscopy, are new technologies allowing real-time exploration of the 'cellular' intestinal barrier. Additionally, novel advanced spatial imaging technology platforms, including multispectral imaging, upconversion nanoparticles, digital spatial profiling, optical spectroscopy and mass cytometry, enable a deep and comprehensive assessment of the 'molecular' and 'ultrastructural' barrier. In this promising landscape, artificial intelligence plays a pivotal role in standardising and integrating these novel tools, thereby contributing to barrier assessment and prediction of outcomes.Looking ahead, this integrated and comprehensive approach holds the promise of uncovering new therapeutic targets, breaking the therapeutic ceiling in IBD. Novel molecules, dietary interventions and microbiome modulation strategies aim to restore, reinforce, or modulate the gut-brain axis. These advancements have the potential for transformative and personalised approaches to managing IBD.
Collapse
Affiliation(s)
- Marietta Iacucci
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Giovanni Santacroce
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Snehali Majumder
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Jennifer Morael
- APC Microbiome Ireland, Department of Anatomy and Neuroscience, School of Medicine, University College Cork, Cork, Ireland
| | - Irene Zammarchi
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Yasuharu Maeda
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| | - David Ryan
- Department of Radiology, School of Medicine, University College Cork, Cork, Ireland
| | - Antonio Di Sabatino
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy
- First Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Italy
| | - Maria Rescigno
- IRCSS Humanitas Research Hospital, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Maria R Aburto
- APC Microbiome Ireland, Department of Anatomy and Neuroscience, School of Medicine, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, Department of Anatomy and Neuroscience, School of Medicine, University College Cork, Cork, Ireland
| | - Subrata Ghosh
- APC Microbiome Ireland, College of Medicine and Health, University College Cork, Cork, Ireland
| |
Collapse
|
18
|
Parente IA, Chiara L, Bertoni S. Exploring the potential of human intestinal organoids: Applications, challenges, and future directions. Life Sci 2024; 352:122875. [PMID: 38942359 DOI: 10.1016/j.lfs.2024.122875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/13/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
The complex and dynamic environment of the gastrointestinal tract shapes one of the fastest renewing tissues in the human body, the intestinal epithelium. Considering the lack of human preclinical studies, reliable models that mimic the intestinal environment are increasingly explored. Patient-derived intestinal organoids are powerful tools that recapitulate in vitro many pathophysiological features of the human intestine. In this review, the possible applications of human intestinal organoids in different research fields are highlighted. From physiologically relevant to intestinal disease modeling, regenerative medicine, and toxicology studies, the potential of intestinal organoids will be here presented and discussed. Despite the remarkable opportunities offered, limitations related to ethical concerns, tissue collection, reproducibility, and methodologies may hinder the full exploitation of this cell-based model into high throughput studies and clinical practice. Currently, distinct approaches can be used to overcome the numerous challenges found along the way and to allow the full implementation of this ground-breaking technology.
Collapse
Affiliation(s)
- Inês A Parente
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Linda Chiara
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Simona Bertoni
- Department of Food and Drug, University of Parma, Parma, Italy.
| |
Collapse
|
19
|
Zhou L, Luo D, Lu W, Han J, Zhao M, Li X, Shen T, Jin Z, Zeng J, Wen Y. Gastrointestinal tract organoids as novel tools in drug discovery. Front Pharmacol 2024; 15:1463114. [PMID: 39281285 PMCID: PMC11394194 DOI: 10.3389/fphar.2024.1463114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/20/2024] [Indexed: 09/18/2024] Open
Abstract
Organoids, characterized by their high physiological attributes, effectively preserve the genetic characteristics, physiological structure, and function of the simulated organs. Since the inception of small intestine organoids, other organoids for organs including the liver, lungs, stomach, and pancreas have subsequently been developed. However, a comprehensive summary and discussion of research findings on gastrointestinal tract (GIT) organoids as disease models and drug screening platforms is currently lacking. Herein, in this review, we address diseases related to GIT organoid simulation and highlight the notable advancements that have been made in drug screening and pharmacokinetics, as well as in disease research and treatment using GIT organoids. Organoids of GIT diseases, including inflammatory bowel disease, irritable bowel syndrome, necrotizing enterocolitis, and Helicobacter pylori infection, have been successfully constructed. These models have facilitated the study of the mechanisms and effects of various drugs, such as metformin, Schisandrin C, and prednisolone, in these diseases. Furthermore, GIT organoids have been used to investigate viruses that elicit GIT reactions, including Norovirus, SARS-CoV-2, and rotavirus. Previous studies by using GIT organoids have shown that dasabuvir, gemcitabine, and imatinib possess the capability to inhibit viral replication. Notably, GIT organoids can mimic GIT responses to therapeutic drugs at the onset of disease. The GIT toxicities of compounds like gefitinib, doxorubicin, and sunset yellow have also been evaluated. Additionally, these organoids are instrumental for the study of immune regulation, post-radiation intestinal epithelial repair, treatment for cystic fibrosis and diabetes, the development of novel drug delivery systems, and research into the GIT microbiome. The recent use of conditioned media as a culture method for replacing recombinant hepatocyte growth factor has significantly reduced the cost associated with human GIT organoid culture. This advancement paves the way for large-scale culture and compound screening of GIT organoids. Despite the ongoing challenges in GIT organoid development (e.g., their inability to exist in pairs, limited cell types, and singular drug exposure mode), these organoids hold considerable potential for drug screening. The use of GIT organoids in this context holds great promises to enhance the precision of medical treatments for patients living with GIT diseases.
Collapse
Affiliation(s)
- Li Zhou
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dan Luo
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Lu
- Department of Elderly Care Center, Chengdu Pidu District Hospital of Traditional Chinese Medicine, Chengdu, China
| | - Jun Han
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Maoyuan Zhao
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xueyi Li
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tao Shen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhao Jin
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinhao Zeng
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yueqiang Wen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Pediatrics, Guang'an Hospital of Traditional Chinese Medicine, Guang'an, China
| |
Collapse
|
20
|
Deleu S, Jacobs I, Vazquez Castellanos JF, Verstockt S, Trindade de Carvalho B, Subotić A, Verstockt B, Arnauts K, Deprez L, Vissers E, Lenfant M, Vandermeulen G, De Hertogh G, Verbeke K, Matteoli G, Huys GRB, Thevelein JM, Raes J, Vermeire S. Effect of Mutant and Engineered High-Acetate-Producing Saccharomyces cerevisiae var. boulardii Strains in Dextran Sodium Sulphate-Induced Colitis. Nutrients 2024; 16:2668. [PMID: 39203805 PMCID: PMC11357622 DOI: 10.3390/nu16162668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
Acetate-producing Saccharomyces cerevisiae var. boulardii strains could exert improved effects on ulcerative colitis, which here, was preclinically evaluated in an acute dextran sodium sulphate induced model of colitis. Nine-week-old female mice were divided into 12 groups, receiving either drinking water or 2.75% dextran sodium sulphate for 7 days, combined with a daily gavage of various treatments with different levels of acetate accumulation: sham control (phosphate buffered saline, no acetate), non-probiotic control (Baker's yeast, no acetate), probiotic control (Enterol®, transient acetate), and additionally several Saccharomyces cerevisiae var. boulardii strains with respectively no, high, and extra-high acetate accumulation. Disease activity was monitored daily, and feces samples were collected at different timepoints. On day 14, the mice were sacrificed, upon which blood and colonic tissue were collected for analysis. Disease activity in inflamed mice was lower when treated with the high-acetate-producing strain compared to sham and non-probiotic controls. The non-acetate-producing strain showed higher disease activity compared to the acetate-producing strains. Accordingly, higher histologic inflammation was observed in non- or transient-acetate-producing strains compared to the sham control, whereas this increase was not observed for high- and extra-high-acetate-producing strains upon induction of inflammation. These anti-inflammatory findings were confirmed by transcriptomic analysis of differentially expressed genes. Moreover, only the strain with the highest acetate production was superior in maintaining a stable gut microbial alpha-diversity upon inflammation. These findings support new possibilities for acetate-mediated management of inflammation in inflammatory bowel disease by administrating high-acetate-producing Saccharomyces cerevisae var. boulardii strains.
Collapse
Affiliation(s)
- Sara Deleu
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Inge Jacobs
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Jorge F. Vazquez Castellanos
- VIB-KU Leuven Center for Microbiology, 3001 Leuven, Belgium (G.R.B.H.)
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, 3000 Leuven, Belgium
| | - Sare Verstockt
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | | | - Ana Subotić
- NovelYeast bv, Bio-Incubator BIO4, Gaston Geenslaan 3, Leuven-Heverlee, 3001 Leuven, Belgium
| | - Bram Verstockt
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
- Department of Gastroenterology and Hepatology, UZ Leuven, KU Leuven, 3000 Leuven, Belgium
| | - Kaline Arnauts
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Lowie Deprez
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Eva Vissers
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Matthias Lenfant
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
- Department of Gastroenterology and Hepatology, UZ Leuven, KU Leuven, 3000 Leuven, Belgium
| | - Greet Vandermeulen
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Gert De Hertogh
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
- Laboratory of Morphology and Molecular Pathology, UZ Leuven, 3000 Leuven, Belgium
| | - Kristin Verbeke
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Gianluca Matteoli
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Geert R. B. Huys
- VIB-KU Leuven Center for Microbiology, 3001 Leuven, Belgium (G.R.B.H.)
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, 3000 Leuven, Belgium
| | - Johan M. Thevelein
- NovelYeast bv, Bio-Incubator BIO4, Gaston Geenslaan 3, Leuven-Heverlee, 3001 Leuven, Belgium
| | - Jeroen Raes
- VIB-KU Leuven Center for Microbiology, 3001 Leuven, Belgium (G.R.B.H.)
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, 3000 Leuven, Belgium
| | - Séverine Vermeire
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
- Department of Gastroenterology and Hepatology, UZ Leuven, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
21
|
Behrens LMP, Gasparotto J, Rampelotto PH, Escalona MAR, da Silva LDS, Carazza-Kessler FG, Barbosa CP, Campos MS, Dorn M, Gelain DP, Moreira JCF. Sodium propionate oral supplementation ameliorates depressive-like behavior through gut microbiome and histone 3 epigenetic regulation. J Nutr Biochem 2024; 130:109660. [PMID: 38685283 DOI: 10.1016/j.jnutbio.2024.109660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 03/30/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024]
Abstract
Major depressive disorder (MDD) is a global health concern, affecting over 250 million individuals worldwide. In recent years, the gut-brain axis has emerged as a promising field for understanding the pathophysiology of MDD. Microbial metabolites, such as short-chain fatty acids (SCFAs)-acetate, butyrate, and propionate-, have gained attention for their potential to influence epigenetic modifications within the host brain. However, the precise mechanisms through which these metabolites participate in MDD pathophysiology remain elusive. This study was designed to investigate the effects of oral SCFA supplementation in adult male Wistar rats subjected to chronic unpredictable mild stress (CUMS). A subset of control and CUMS-exposed rats received different supplementations: sodium acetate (NaOAc) at a concentration of 60 mM, sodium butyrate (NaB) at 40 mM, sodium propionate (NaP) at 50 mM, or a mixture of these SCFAs. The gut microbiome was assessed through 16S rRNA sequencing, and epigenetic profiling was performed using Western blot analysis. Results demonstrated that NaP supplementation significantly alleviated anhedonia in stressed animals, as evidenced by improved performance in the sucrose consumption test. This ameliorative effect was potentially associated with the modulation of gut bacterial communities, accompanied by the attenuation of the region-specific epigenetic dysregulation in the brain of the animals exposed to chronic stress. These findings suggest a potential association between gut dysbiosis and stress response, and NaP could be a promising target for future MDD interventions. However, further studies are needed to fully elucidate the underlying mechanisms of these effects.
Collapse
Affiliation(s)
- Luiza Marques Prates Behrens
- Center of Oxidative Stress Studies, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 90035-003, Brazil; Graduate Program in Biological Sciences: Biochemistry, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 90035-003, Brazil; Graduate Program in Cellular and Molecular Biology, Center of Biotechnology, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 91501-970, Brazil.
| | - Juciano Gasparotto
- Center of Oxidative Stress Studies, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 90035-003, Brazil
| | - Pabulo Henrique Rampelotto
- Bioinformatics and Biostatistics Core Facility, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 91501-907, Brazil
| | - Manuel Adrian Riveros Escalona
- Graduate Program in Veterinary Sciences, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 91540-000, Brazil
| | - Lucas Dos Santos da Silva
- Center of Oxidative Stress Studies, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 90035-003, Brazil; Graduate Program in Cellular and Molecular Biology, Center of Biotechnology, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 91501-970, Brazil
| | - Flávio Gabriel Carazza-Kessler
- Center of Oxidative Stress Studies, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 90035-003, Brazil; Graduate Program in Biological Sciences: Biochemistry, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 90035-003, Brazil
| | - Camila Pocharski Barbosa
- Center of Oxidative Stress Studies, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 90035-003, Brazil
| | - Marlene Soares Campos
- Center of Oxidative Stress Studies, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 90035-003, Brazil
| | - Márcio Dorn
- Department of Theoretical Informatics, Institute of Informatics, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 91501-970, Brazil; Center of Biotechnology, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 91501-970, Brazil
| | - Daniel Pens Gelain
- Center of Oxidative Stress Studies, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 90035-003, Brazil; Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 90035-003, Brazil
| | - José Cláudio Fonseca Moreira
- Center of Oxidative Stress Studies, Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 90035-003, Brazil; Department of Biochemistry, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre 90035-003, Brazil
| |
Collapse
|
22
|
Peña-Vázquez GI, Arredondo-Arenillas A, Serrano-Sandoval SN, Antunes-Ricardo M. Functional foods lipids: unraveling their role in the immune response in obesity. Crit Rev Food Sci Nutr 2024:1-22. [PMID: 39073763 DOI: 10.1080/10408398.2024.2382942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Functional lipids are lipids that are found in food matrices and play an important role in influencing human health as their role goes beyond energy storage and structural components. Ongoing research into functional lipids has highlighted their potential to modulate immune responses and other mechanisms associated with obesity, along with its comorbidities. These lipids represent a new field that may offer new therapeutic and preventive strategies for these diseases by understanding their contribution to health. In this review, we discussed in-depth the potential food sources of functional lipids and their reported potential benefit of the major lipid classification: based on their composition such as simple, compound, and derived lipids, and based on their function such as storage and structural, by investigating the intricate mechanisms through which these lipids interact in the human body. We summarize the key insights into the bioaccessibility and bioavailability of the most studied functional lipids. Furthermore, we review the main immunomodulatory mechanisms reported in the literature in the past years. Finally, we discuss the perspectives and challenges faced in the food industry related to functional lipids.
Collapse
Affiliation(s)
- Gloria Itzel Peña-Vázquez
- Tecnologico de Monterrey, Centro de Biotecnología FEMSA, Escuela de Ingeniería y Ciencias, Monterrey, NL, México
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, Monterrey, NL, México
| | - Ana Arredondo-Arenillas
- Tecnologico de Monterrey, Centro de Biotecnología FEMSA, Escuela de Ingeniería y Ciencias, Monterrey, NL, México
| | - Sayra N Serrano-Sandoval
- Tecnologico de Monterrey, Centro de Biotecnología FEMSA, Escuela de Ingeniería y Ciencias, Monterrey, NL, México
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, Monterrey, NL, México
| | - Marilena Antunes-Ricardo
- Tecnologico de Monterrey, Centro de Biotecnología FEMSA, Escuela de Ingeniería y Ciencias, Monterrey, NL, México
- Tecnologico de Monterrey, Institute for Obesity Research, Monterrey, Monterrey, NL, México
| |
Collapse
|
23
|
Barrong H, Coven H, Lish A, Fessler SN, Jasbi P, Johnston CS. Daily Vinegar Ingestion Improves Depression and Enhances Niacin Metabolism in Overweight Adults: A Randomized Controlled Trial. Nutrients 2024; 16:2305. [PMID: 39064748 PMCID: PMC11280469 DOI: 10.3390/nu16142305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/15/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Depressive disorders are the most prevalent mental health conditions in the world. The commonly prescribed antidepressant medications can have serious side effects, and their efficacy varies widely. Thus, simple, effective adjunct therapies are needed. Vinegar, a fermented acetic acid solution, is emerging as a healthful dietary supplement linked to favorable outcomes for blood glucose management, heart disease risk, and adiposity reduction, and a recent report suggests vinegar may improve symptoms of depression. This randomized controlled study examined the 4-week change in scores for the Center for Epidemiological Studies Depression (CES-D) questionnaire and the Patient Health Questionnaire (PHQ-9) in healthy overweight adults ingesting 2.95 g acetic acid (4 tablespoons vinegar) vs. 0.025 g acetic acid (one vinegar pill) daily. A secondary objective explored possible underlying mechanisms using metabolomics analyses. At week 4, mean CES-D scores fell 26% and 5% for VIN and CON participants respectively, a non-significant difference between groups, and mean PHQ-9 scores fell 42% and 18% for VIN and CON participants (p = 0.036). Metabolomics analyses revealed increased nicotinamide concentrations and upregulation of the NAD+ salvage pathway for VIN participants compared to controls, metabolic alterations previously linked to improved mood. Thus, daily vinegar ingestion over four weeks improved self-reported depression symptomology in healthy overweight adults, and enhancements in niacin metabolism may factor into this improvement.
Collapse
Affiliation(s)
- Haley Barrong
- Nutrition Program, College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (H.B.); (H.C.); (A.L.); (S.N.F.)
| | - Hannah Coven
- Nutrition Program, College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (H.B.); (H.C.); (A.L.); (S.N.F.)
| | - Alexandra Lish
- Nutrition Program, College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (H.B.); (H.C.); (A.L.); (S.N.F.)
| | - Samantha N. Fessler
- Nutrition Program, College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (H.B.); (H.C.); (A.L.); (S.N.F.)
| | - Paniz Jasbi
- Systems Precision Engineering and Advanced Research (SPEAR), Theriome Inc., Phoenix, AZ 85004, USA;
| | - Carol S. Johnston
- Nutrition Program, College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (H.B.); (H.C.); (A.L.); (S.N.F.)
| |
Collapse
|
24
|
Custers E, Vreeken D, Schuren F, van den Broek TJ, van Dongen L, Geenen B, de Blaauw I, Wiesmann M, Hazebroek EJ, Kleemann R, Kiliaan AJ. Impact of Microbiota and Metabolites on Intestinal Integrity and Inflammation in Severe Obesity. Pharmaceuticals (Basel) 2024; 17:918. [PMID: 39065768 PMCID: PMC11279642 DOI: 10.3390/ph17070918] [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: 06/18/2024] [Revised: 07/03/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Obesity is a multifactorial disease associated with low-grade inflammation. The gut is thought to be involved in obesity-related inflammation, as it is continuously exposed to antigens from food, microbiota and metabolites. However, the exact underlying mechanisms are still unknown. Therefore, we examined the relation between gut pathology, microbiota, its metabolites and cytokines in adults with severe obesity. Individuals eligible for bariatric surgery were included. Fecal and plasma samples were collected at surgery timepoint, to assess microbiota and metabolite composition. Jejunal biopsies were collected during surgery and stained for cytotoxic T cells, macrophages, mast cells and tight junction component zonula occludens-1. Based on these stainings, the cohort was divided into four groups: high versus low intestinal inflammation and high versus low intestinal integrity. We found no significant differences in microbiota diversity between groups, nor for individual bacterial species. No significant differences in metabolites were observed between the intestinal inflammatory groups. However, some metabolites and cytokines differed between the intestinal integrity groups. Higher plasma levels of interleukin-8 and tauro-chenodeoxycholic acid were found, whereas isovaleric acid and acetic acid were lower in the high intestinal integrity group. As the results were very subtle, we suggest that our cohort shows very early and minor intestinal pathology.
Collapse
Affiliation(s)
- Emma Custers
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Radboud Alzheimer Center, Donders Institute for Brain Cognition and Behaviour, Center for Medical Neuroscience, 6500 HB Nijmegen, The Netherlands; (E.C.); (B.G.); (M.W.)
- Department of Bariatric Surgery, Vitalys, Rijnstate Hospital, 6815 AD Arnhem, The Netherlands;
| | - Debby Vreeken
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Radboud Alzheimer Center, Donders Institute for Brain Cognition and Behaviour, Center for Medical Neuroscience, 6500 HB Nijmegen, The Netherlands; (E.C.); (B.G.); (M.W.)
- Department of Bariatric Surgery, Vitalys, Rijnstate Hospital, 6815 AD Arnhem, The Netherlands;
| | - Frank Schuren
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands; (F.S.); (R.K.)
| | - Tim J. van den Broek
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands; (F.S.); (R.K.)
| | - Lieke van Dongen
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Radboud Alzheimer Center, Donders Institute for Brain Cognition and Behaviour, Center for Medical Neuroscience, 6500 HB Nijmegen, The Netherlands; (E.C.); (B.G.); (M.W.)
| | - Bram Geenen
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Radboud Alzheimer Center, Donders Institute for Brain Cognition and Behaviour, Center for Medical Neuroscience, 6500 HB Nijmegen, The Netherlands; (E.C.); (B.G.); (M.W.)
| | - Ivo de Blaauw
- Division of Pediatric Surgery, Department of Surgery, Radboudumc-Amalia Children’s Hospital, 6525 GA Nijmegen, The Netherlands;
| | - Maximilian Wiesmann
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Radboud Alzheimer Center, Donders Institute for Brain Cognition and Behaviour, Center for Medical Neuroscience, 6500 HB Nijmegen, The Netherlands; (E.C.); (B.G.); (M.W.)
| | - Eric J. Hazebroek
- Department of Bariatric Surgery, Vitalys, Rijnstate Hospital, 6815 AD Arnhem, The Netherlands;
- Division of Human Nutrition and Health, Wageningen University and Research, 6708 WE Wageningen, The Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands; (F.S.); (R.K.)
| | - Amanda J. Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Radboud Alzheimer Center, Donders Institute for Brain Cognition and Behaviour, Center for Medical Neuroscience, 6500 HB Nijmegen, The Netherlands; (E.C.); (B.G.); (M.W.)
| |
Collapse
|
25
|
Du Y, He C, An Y, Huang Y, Zhang H, Fu W, Wang M, Shan Z, Xie J, Yang Y, Zhao B. The Role of Short Chain Fatty Acids in Inflammation and Body Health. Int J Mol Sci 2024; 25:7379. [PMID: 39000498 PMCID: PMC11242198 DOI: 10.3390/ijms25137379] [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: 05/24/2024] [Revised: 06/27/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Short chain fatty acids (SCFAs), mainly including acetate, propionate and butyrate, are produced by intestinal bacteria during the fermentation of partially digested and indigestible polysaccharides. SCFAs play an important role in regulating intestinal energy metabolism and maintaining the homeostasis of the intestinal environment and also play an important regulatory role in organs and tissues outside the gut. In recent years, many studies have shown that SCFAs can regulate inflammation and affect host health, and two main signaling mechanisms have also been identified: the activation of G-protein coupled receptors (GPCRs) and inhibition of histone deacetylase (HDAC). In addition, a growing body of evidence highlights the importance of every SCFA in influencing health maintenance and disease development. In this review, we summarized the recent advances concerning the biological properties of SCFAs and their signaling pathways in inflammation and body health. Hopefully, it can provide a systematic theoretical basis for the nutritional prevention and treatment of human diseases.
Collapse
Affiliation(s)
- Yuhang Du
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Changhao He
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yongcheng An
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yan Huang
- College of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Huilin Zhang
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wanxin Fu
- College of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Menglu Wang
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Ziyi Shan
- College of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jiamei Xie
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yang Yang
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Baosheng Zhao
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|
26
|
Wang Q, Guo F, Zhang Q, Hu T, Jin Y, Yang Y, Ma Y. Organoids in gastrointestinal diseases: from bench to clinic. MedComm (Beijing) 2024; 5:e574. [PMID: 38948115 PMCID: PMC11214594 DOI: 10.1002/mco2.574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/15/2024] [Accepted: 04/26/2024] [Indexed: 07/02/2024] Open
Abstract
The etiology of gastrointestinal (GI) diseases is intricate and multifactorial, encompassing complex interactions between genetic predisposition and gut microbiota. The cell fate change, immune function regulation, and microenvironment composition in diseased tissues are governed by microorganisms and mutated genes either independently or through synergistic interactions. A comprehensive understanding of GI disease etiology is imperative for developing precise prevention and treatment strategies. However, the existing models used for studying the microenvironment in GI diseases-whether cancer cell lines or mouse models-exhibit significant limitations, which leads to the prosperity of organoids models. This review first describes the development history of organoids models, followed by a detailed demonstration of organoids application from bench to clinic. As for bench utilization, we present a layer-by-layer elucidation of organoid simulation on host-microbial interactions, as well as the application in molecular mechanism analysis. As for clinical adhibition, we provide a generalized interpretation of organoid application in GI disease simulation from inflammatory disorders to malignancy diseases, as well as in GI disease treatment including drug screening, immunotherapy, and microbial-targeting and screening treatment. This review draws a comprehensive and systematical depiction of organoids models, providing a novel insight into the utilization of organoids models from bench to clinic.
Collapse
Affiliation(s)
- Qinying Wang
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of Cancer InstituteFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Fanying Guo
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Qinyuan Zhang
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - TingTing Hu
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - YuTao Jin
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Yongzhi Yang
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Yanlei Ma
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
27
|
de Carvalho BT, Subotić A, Vandecruys P, Deleu S, Vermeire S, Thevelein JM. Enhancing probiotic impact: engineering Saccharomyces boulardii for optimal acetic acid production and gastric passage tolerance. Appl Environ Microbiol 2024; 90:e0032524. [PMID: 38752748 PMCID: PMC11218656 DOI: 10.1128/aem.00325-24] [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: 02/22/2024] [Accepted: 04/18/2024] [Indexed: 06/19/2024] Open
Abstract
Saccharomyces boulardii has been a subject of growing interest due to its potential as a probiotic microorganism with applications in gastrointestinal health, but the molecular cause for its probiotic potency has remained elusive. The recent discovery that S. boulardii contains unique mutations causing high acetic acid accumulation and inhibition of bacterial growth provides a possible clue. The natural S. boulardii isolates Sb.P and Sb.A are homozygous for the recessive mutation whi2S270* and accumulate unusually high amounts of acetic acid, which strongly inhibit bacterial growth. However, the homozygous whi2S270* mutation also leads to acetic acid sensitivity and acid sensitivity in general. In the present study, we have constructed a new S. boulardii strain, derived from the widely therapeutically used CMCN I-745 strain (isolated from the pharmaceutical product Enterol), producing even higher levels of acetic acid while keeping the same tolerance toward low pH as the parent Enterol (ENT) strain. This newly engineered strain, named ENT3, has a homozygous deletion of ACH1 and strong overexpression of ALD4. It is also able to accumulate much higher acetic acid concentrations when growing on low glucose levels, in contrast to the ENT wild-type and Sb.P strains. Moreover, we show the antimicrobial capacity of ENT3 against gut pathogens in vitro and observed that higher acetic acid production might correlate with better persistence in the gut in healthy mice. These findings underscore the possible role of the unique acetic acid production and its potential for improvement of the probiotic action of S. boulardii.IMPORTANCESuperior variants of the probiotic yeast Saccharomyces boulardii produce high levels of acetic acid, which inhibit the growth of bacterial pathogens. However, these strains also show increased acid sensitivity, which can compromise the viability of the cells during their passage through the stomach. In this work, we have developed by genetic engineering a variant of Saccharomyces boulardii that produces even higher levels of acetic acid and does not show enhanced acid sensitivity. We also show that the S. boulardii yeasts with higher acetic acid production persist longer in the gut, in agreement with a previous work indicating competition between probiotic yeast and bacteria for residence in the gut.
Collapse
Affiliation(s)
| | - Ana Subotić
- NovelYeast bv, Bio-Incubator BIO4, Leuven-Heverlee, Belgium
| | - Paul Vandecruys
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven-Heverlee, Belgium
| | - Sara Deleu
- Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Séverine Vermeire
- Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Johan M. Thevelein
- NovelYeast bv, Bio-Incubator BIO4, Leuven-Heverlee, Belgium
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven-Heverlee, Belgium
- Center for Microbiology, VIB, Leuven-Heverlee, Belgium
| |
Collapse
|
28
|
Ge X, Liu H, Wu Y, Liu W, Qi W, Ye L, Cao Q, Lian H, Bai R, Zhou W. Parenteral n-3 polyunsaturated fatty acids supplementation improves postoperative recovery for patients with Crohn's disease after bowel resection: a randomized, unblinded controlled clinical trial. Am J Clin Nutr 2024; 119:1027-1035. [PMID: 38569774 DOI: 10.1016/j.ajcnut.2023.12.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND The postoperative inflammatory response is associated with postoperative recovery in surgery. n-3 (ω-3) polyunsaturated fatty acids have been reported to lower inflammation. The postoperative role of parenteral n-3 polyunsaturated fatty acids supplementation on outcomes in Crohn's disease after bowel resection is unclear. OBJECTIVES We aimed to investigate the effects of postoperative parenteral n-3 polyunsaturated fatty acids supplementation in Crohn's disease. METHODS A prospective randomized, unblinded controlled clinical trial was conducted for patients with Crohn's disease who underwent bowel resection between May 2019 and February 2022. Postoperative complications, complete blood count, serum biochemical values, and cytokine concentrations were compared in patients with and without parenteral n-3 polyunsaturated fatty acids supplementation for 5 d postoperatively. RESULTS There were 268 patients randomly assigned in the analysis, with 134 in the control group (a mix of long-chain and medium-chain fats at 1.0 g/kg/d) and 134 in the treatment group (long-chain, medium-chain, and n-3 polyunsaturated fats at 1.2 g/kg/d). Twenty-six did not complete the allocated treatment, and 8 patients were lost to follow-up. The intention-to-treat analysis and the per-protocol analysis showed that there were a significant reduction in overall complication rates (22.4% compared with 49.3%; P < 0.001 and 21.8% compared with 38.2%; P = 0.006) and postoperative stay (8.8 ± 4.5 d compared with 11.2 ± 6.8 d; P = 0.001 and 8.7 ± 4.0 d compared with 11.5 ± 7.3 d; P < 0.001) in patients with parenteral n-3 polyunsaturated fatty acids supplementation compared with patients in the control group. In the secondary outcomes, the mean ± standard deviation of interleukin (IL)-6 (17.11 ± 2.14 pg/mL compared with 30.50 ± 5.14 pg/mL; P = 0.014), IL-1β (2.01 ± 0.05 pg/mL compared with 2.24 ± 0.09 pg/mL; P = 0.019), tumor necrosis factor-α (2.09 ± 0.06 pg/mL compared with 2.29 ± 0.06 pg/mL; P = 0.029), and C-reactive protein concentrations (51.3 ± 4.2 mg/L compared with 64.4 ± 5.3 mg/L; P = 0.050) on postoperative day 5 in the treatment group were much lower than those in the control group. CONCLUSIONS Parenteral n-3 polyunsaturated fatty acids supplementation promotes postoperative recovery in patients with Crohn's disease following bowel resection, with fewer complications and reduced inflammatory cytokines. This trial was registered at clinicaltrials.gov as NCT03901937 at https://classic. CLINICALTRIALS gov/ct2/show/NCT03901937?term=NCT03901937&cond=Crohn+Disease&draw=2&rank=1.
Collapse
Affiliation(s)
- Xiaolong Ge
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huaying Liu
- Department of Medicine, Guangxi Medical College, Nanning, China
| | - Yan Wu
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Liu
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weilin Qi
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Linna Ye
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qian Cao
- Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haifeng Lian
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China.
| | - Rongpan Bai
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Wei Zhou
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Inflammatory Bowel Disease Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
29
|
Kong L, Chen S, Huang S, Zheng A, Gao S, Ye J, Hua C. Challenges and opportunities in inflammatory bowel disease: from current therapeutic strategies to organoid-based models. Inflamm Res 2024; 73:541-562. [PMID: 38345635 DOI: 10.1007/s00011-024-01854-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/20/2024] [Accepted: 01/24/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is an increasingly prevalent global health concern that has garnered substantial attention. However, the underlying mechanisms are still unclear and the current treatments have significant limitations. Intestinal organoids provide an in vitro model to explore the pathogenesis, test the therapeutic effects, and develop regenerative treatments as well as offer the potential to transform drug discovery of IBD. METHODS To advance our understanding of the whole story of IBD spanning from the pathogenesis to the current therapeutic strategies and latest advancements, a comprehensive search of major databases including PubMed, Scopus, and Web of Science was conducted to retrieve original articles and reviews related to IBD, organoids, pathogenesis and therapy. RESULTS This review deciphers the etiopathogenesis and the current therapeutic approaches in the treatment of IBD. Notably, critical aspects of intestinal organoids in IBD, such as their potential applications, viability, cell renewal ability, and barrier functionality are highlighted. We also discuss the advances, limitations, and prospects of intestinal organoids for precision medicine. CONCLUSION The latest strides made in research about intestinal organoids help elucidate intricate aspects of IBD pathogenesis, and pave the prospective avenues for novel therapeutic interventions.
Collapse
Affiliation(s)
- Lingjie Kong
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Siyan Chen
- School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Shenghao Huang
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Anzhe Zheng
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Sheng Gao
- Laboratory Animal Center, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Jianzhong Ye
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Chunyan Hua
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
30
|
Xiao J, Guo X, Wang Z. Crosstalk between hypoxia-inducible factor-1α and short-chain fatty acids in inflammatory bowel disease: key clues toward unraveling the mystery. Front Immunol 2024; 15:1385907. [PMID: 38605960 PMCID: PMC11007100 DOI: 10.3389/fimmu.2024.1385907] [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: 02/14/2024] [Accepted: 03/19/2024] [Indexed: 04/13/2024] Open
Abstract
The human intestinal tract constitutes a complex ecosystem, made up of countless gut microbiota, metabolites, and immune cells, with hypoxia being a fundamental environmental characteristic of this ecology. Under normal physiological conditions, a delicate balance exists among these complex "residents", with disruptions potentially leading to inflammatory bowel disease (IBD). The core pathology of IBD features a disrupted intestinal epithelial barrier, alongside evident immune and microecological disturbances. Central to these interconnected networks is hypoxia-inducible factor-1α (HIF-1α), which is a key regulator in gut cells for adapting to hypoxic conditions and maintaining gut homeostasis. Short-chain fatty acids (SCFAs), as pivotal gut metabolites, serve as vital mediators between the host and microbiota, and significantly influence intestinal ecosystem. Recent years have seen a surge in research on the roles and therapeutic potential of HIF-1α and SCFAs in IBD independently, yet reviews on HIF-1α-mediated SCFAs regulation of IBD under hypoxic conditions are scarce. This article summarizes evidence of the interplay and regulatory relationship between SCFAs and HIF-1α in IBD, pivotal for elucidating the disease's pathogenesis and offering promising therapeutic strategies.
Collapse
Affiliation(s)
- Jinyin Xiao
- Graduate School, Hunan University of Traditional Chinese Medicine, Changsha, China
- Department of Anorectal, the Second Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Xiajun Guo
- Department of Geriatric, the First People’s Hospital of Xiangtan City, Xiangtan, China
| | - Zhenquan Wang
- Department of Anorectal, the Second Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, China
| |
Collapse
|
31
|
Jardou M, Brossier C, Marquet P, Picard N, Druilhe A, Lawson R. Solid organ transplantation and gut microbiota: a review of the potential immunomodulatory properties of short-chain fatty acids in graft maintenance. Front Cell Infect Microbiol 2024; 14:1342354. [PMID: 38476165 PMCID: PMC10927761 DOI: 10.3389/fcimb.2024.1342354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Transplantation is the treatment of choice for several end-stage organ defects: it considerably improves patient survival and quality of life. However, post-transplant recipients may experience episodes of rejection that can favor or ultimately lead to graft loss. Graft maintenance requires a complex and life-long immunosuppressive treatment. Different immunosuppressive drugs (i.e., calcineurin inhibitors, glucocorticoids, biological immunosuppressive agents, mammalian target of rapamycin inhibitors, and antiproliferative or antimetabolic agents) are used in combination to mitigate the immune response against the allograft. Unfortunately, the use of these antirejection agents may lead to opportunistic infections, metabolic (e.g., post-transplant diabetes mellitus) or cardiovascular (e.g., arterial hypertension) disorders, cancer (e.g., non-Hodgkin lymphoma) and other adverse effects. Lately, immunosuppressive drugs have also been associated with gut microbiome alterations, known as dysbiosis, and were shown to affect gut microbiota-derived short-chain fatty acids (SCFA) production. SCFA play a key immunomodulatory role in physiological conditions, and their impairment in transplant patients could partly counterbalance the effect of immunosuppressive drugs leading to the activation of deleterious pathways and graft rejection. In this review, we will first present an overview of the mechanisms of graft rejection that are prevented by the immunosuppressive protocol. Next, we will explain the dynamic changes of the gut microbiota during transplantation, focusing on SCFA. Finally, we will describe the known functions of SCFA in regulating immune-inflammatory reactions and discuss the impact of SCFA impairment in immunosuppressive drug treated patients.
Collapse
Affiliation(s)
| | | | | | | | | | - Roland Lawson
- National Institute of Health and Medical Research (FRANCE) (INSERM), Univ. Limoges, Pharmacology & Transplantation, U1248, Limoges, France
| |
Collapse
|
32
|
Yao Y, Liu Y, Xu Q, Mao L. Short Chain Fatty Acids: Essential Weapons of Traditional Medicine in Treating Inflammatory Bowel Disease. Molecules 2024; 29:379. [PMID: 38257292 PMCID: PMC10818876 DOI: 10.3390/molecules29020379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic and recurrent intestinal inflammatory disease, mainly including Crohn's disease (CD) and ulcerative colitis (UC). In recent years, the incidence and prevalence of IBD have been on the rise worldwide and have become a significant concern of health and a huge economic burden on patients. The occurrence and development of IBD involve a variety of pathogenic factors. The changes in short-chain fatty acids (SCFAs) are considered to be an important pathogenic mechanism of this disease. SCFAs are important metabolites in the intestinal microbial environment, which are closely involved in regulating immune, anti-tumor, and anti-inflammatory activities. Changes in metabolite levels can reflect the homeostasis of the intestinal microflora. Recent studies have shown that SCFAs provide energy for host cells and intestinal microflora, shape the intestinal environment, and regulate the immune system, thereby regulating intestinal physiology. SCFAs can effectively reduce the incidence of enteritis, cardiovascular disease, colon cancer, obesity, and diabetes, and also play an important role in maintaining the balance of energy metabolism (mainly glucose metabolism) and improving insulin tolerance. In recent years, many studies have shown that numerous decoctions and natural compounds of traditional Chinese medicine have shown promising therapeutic activities in multiple animal models of colitis and thus attracted increasing attention from scientists in the study of IBD treatment. Some of these traditional Chinese medicines or compounds can effectively alleviate colonic inflammation and clinical symptoms by regulating the generation of SCFAs. This study reviews the effects of various traditional Chinese medicines or bioactive substances on the production of SCFAs and their potential impacts on the severity of colonic inflammation. On this basis, we discussed the mechanism of SCFAs in regulating IBD-associated inflammation, as well as the related regulatory factors and signaling pathways. In addition, we provide our understanding of the limitations of current research and the prospects for future studies on the development of new IBD therapies by targeting SCFAs. This review may widen our understanding of the effect of traditional medicine from the view of SCFAs and their role in alleviating IBD animal models, thus contributing to the studies of IBD researchers.
Collapse
Affiliation(s)
- Yuan Yao
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China; (Y.Y.); (Y.L.)
| | - Yongchao Liu
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China; (Y.Y.); (Y.L.)
| | - Qiuyun Xu
- Basic Medical Research Center, School of Medicine, Nantong University, Nantong 226019, China
| | - Liming Mao
- Department of Immunology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, China; (Y.Y.); (Y.L.)
| |
Collapse
|
33
|
Hedin KA, Mirhakkak MH, Vaaben TH, Sands C, Pedersen M, Baker A, Vazquez-Uribe R, Schäuble S, Panagiotou G, Wellejus A, Sommer MOA. Saccharomyces boulardii enhances anti-inflammatory effectors and AhR activation via metabolic interactions in probiotic communities. THE ISME JOURNAL 2024; 18:wrae212. [PMID: 39488793 PMCID: PMC11631509 DOI: 10.1093/ismejo/wrae212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/17/2024] [Accepted: 11/01/2024] [Indexed: 11/04/2024]
Abstract
Metabolic exchanges between strains in gut microbial communities shape their composition and interactions with the host. This study investigates the metabolic synergy between potential probiotic bacteria and Saccharomyces boulardii, aiming to enhance anti-inflammatory effects within a multi-species probiotic community. By screening a collection of 85 potential probiotic bacterial strains, we identified two strains that demonstrated a synergistic relationship with S. boulardii in pairwise co-cultivation. Furthermore, we computationally predicted cooperative communities with symbiotic relationships between S. boulardii and these bacteria. Experimental validation of 28 communities highlighted the role of S. boulardii as a key player in microbial communities, significantly boosting the community's cell number and production of anti-inflammatory effectors, thereby affirming its essential role in improving symbiotic dynamics. Based on our observation, one defined community significantly activated the aryl hydrocarbon receptor-a key regulator of immune response-280-fold more effectively than the community without S. boulardii. This study underscores the potential of microbial communities for the design of more effective probiotic formulations.
Collapse
Affiliation(s)
- Karl Alex Hedin
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Mohammad H Mirhakkak
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute (Leibniz-HKI), Jena 07745, Germany
| | - Troels Holger Vaaben
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Carmen Sands
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Mikael Pedersen
- National Food Institute, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Adam Baker
- Human Health Biosolution, Novonesis, Hørsholm 2970, Denmark
| | - Ruben Vazquez-Uribe
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby 2800, Denmark
- Center for Microbiology, VIB-KU Leuven, Leuven 3001, Belgium
| | - Sascha Schäuble
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute (Leibniz-HKI), Jena 07745, Germany
| | - Gianni Panagiotou
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology—Hans Knöll Institute (Leibniz-HKI), Jena 07745, Germany
- Faculty of Biological Sciences, Institute of Microbiology, Friedrich Schiller University, Jena 07743, Germany
- Jena University Hospital, Friedrich Schiller University, Jena 07743, Germany
- Department of Medicine, University of Hong Kong, Hong Kong (SAR), China
| | - Anja Wellejus
- Human Health Biosolution, Novonesis, Hørsholm 2970, Denmark
| | - Morten Otto Alexander Sommer
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| |
Collapse
|
34
|
Ni M, He H, Chen M, Li Z, Cai H, Chen Z, Li M, Xu H. Supplementation of sodium acetate improves the growth performance and intestinal health of rabbits through Wnt/β-catenin signaling pathway. J Anim Sci 2024; 102:skae197. [PMID: 39037212 PMCID: PMC11337008 DOI: 10.1093/jas/skae197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 07/21/2024] [Indexed: 07/23/2024] Open
Abstract
Acetic acid, which is one of the most abundant short-chain fatty acids (SCFA) in rabbits' cecum, has been reported to play an important function during various physiological metabolic processes. The present study was conducted to elucidate the effects of sodium acetate on growth performance and intestinal health by evaluating feed intake and efficiency, diarrhea score, serum and cecum metabolites, cecal pH and SCFA, histological staining, nutritional composition of meat and gene expression profile of cecum in rabbits. As a result of sodium acetate supplement, the feed conversion ratio, diarrhea score, and diameter of muscle fiber were significantly decreased (P < 0.05). Additionally, dietary sodium acetate significantly increased in total area of muscle fibers and content of crude ash (P < 0.05). Dietary sodium acetate significantly increased serum glucose, total bile acid, and total cholesterol levels and decreased amylase, lipase, and tCO2 content (P < 0.05). Further examination suggested that sodium acetate supplementation enhanced the micro-environment of cecum, evidenced by significantly increased levels of total antioxidant capacity, total superoxide dismutase, and glutathione peroxidase, and decreased pH and amylase levels (P < 0.05). According to transcriptome sequencing of cecal tissues, differentially expressed genes were predominantly enriched in cell cycle, ABC transporters, and chemokine signaling pathways. Sodium acetate was further suggested to stimulate the proliferation and migration of rabbits' cecum epithelial cells by activating Wnt/β-catenin pathway both in vivo and in vitro. In conclusion, dietary sodium acetate supplementation improved growth performance and intestinal health in rabbits.
Collapse
Affiliation(s)
- Mengke Ni
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Hui He
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Mengjuan Chen
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Zhichao Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Hanfang Cai
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Zhi Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Ming Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Huifen Xu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| |
Collapse
|
35
|
Dinu LD, Gatea F, Matei F, Banciu C, Vamanu E. Gut Microbiota Modulation by Selenium and Zinc Enrichment Postbiotic on Dysbiosis Associated with Hypertension. Curr Vasc Pharmacol 2024; 22:365-374. [PMID: 38779729 DOI: 10.2174/0115701611290537240509061549] [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: 11/30/2023] [Revised: 04/03/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Targeting gut dysbiosis to treat chronic diseases or to alleviate the symptoms is a new direction for medical adjuvant therapies. Recently, postbiotics have received considerable attention as they are non-viable probiotic preparations that confer various health benefits to the host without the safety problems associated with using live microbial cells. OBJECTIVE The aim of the study is to obtain selenium (Se) and zinc (Zn) enriched Saccharomyces boulardii postbiotic biomass and to analyze its modulation effect because these minerals play an important role in reducing gut dysbiosis linked to cardiovascular (CV) diseases. METHOD The effect of the S. boulardii and Se/Zn enriched yeast postbiotics on CV microbial fingerprint was studied in vitro using the gastrointestinal system (GIS 1) and analyzed by microbiological, chemical, and qPCR methods. RESULT There was a 2.2 log CFU/mL increase in the total bacterial load after SeZn postbiotic treatment and in the qPCR counts of Firmicutes phyla for both treatments. Beneficial taxa, Bifidobacterium spp. and Lactobacillus spp., as well as Bacteroidesspp. were up to 1.5 log higher after mineral- enriched postbiotic application, while the acetic acid level increased. CONCLUSION These preliminary studies highlight the therapeutic potential of using Se/Zn enriched yeast postbiotics as adjuvants for clinical treatments of CV diseases.
Collapse
Affiliation(s)
- Laura-Dorina Dinu
- Department of Industrial Biotechnology, Faculty of Biotechnology, University of Agronomic Sciences and Veterinary Medicine, Bucharest, Romania
| | - Florentina Gatea
- Department of Biotechnology, Centre of Bioanalysis, National Institute of Research and Development for Biological Sciences, Bucharest, Romania
| | - Florentina Matei
- Faculty of Food Industry and Tourism, Transylvania University of Brasov, Brasov, Romania
| | - Cristian Banciu
- Department of Ecology, Institute of Biology of Romanian Academy, Bucharest, Romania
| | - Emanuel Vamanu
- Department of Industrial Biotechnology, Faculty of Biotechnology, University of Agronomic Sciences and Veterinary Medicine, Bucharest, Romania
| |
Collapse
|
36
|
Yan D, Ye S, He Y, Wang S, Xiao Y, Xiang X, Deng M, Luo W, Chen X, Wang X. Fatty acids and lipid mediators in inflammatory bowel disease: from mechanism to treatment. Front Immunol 2023; 14:1286667. [PMID: 37868958 PMCID: PMC10585177 DOI: 10.3389/fimmu.2023.1286667] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023] Open
Abstract
Inflammatory Bowel Disease (IBD) is a chronic, relapsing inflammatory disorder of the gastrointestinal tract. Though the pathogenesis of IBD remains unclear, diet is increasingly recognized as a pivotal factor influencing its onset and progression. Fatty acids, essential components of dietary lipids, play diverse roles in IBD, ranging from anti-inflammatory and immune-regulatory functions to gut-microbiota modulation and barrier maintenance. Short-chain fatty acids (SCFAs), products of indigestible dietary fiber fermentation by gut microbiota, have strong anti-inflammatory properties and are seen as key protective factors against IBD. Among long-chain fatty acids, saturated fatty acids, trans fatty acids, and ω-6 polyunsaturated fatty acids exhibit pro-inflammatory effects, while oleic acid and ω-3 polyunsaturated fatty acids display anti-inflammatory actions. Lipid mediators derived from polyunsaturated fatty acids serve as bioactive molecules, influencing immune cell functions and offering both pro-inflammatory and anti-inflammatory benefits. Recent research has also highlighted the potential of medium- and very long-chain fatty acids in modulating inflammation, mucosal barriers, and gut microbiota in IBD. Given these insights, dietary intervention and supplementation with short-chain fatty acids are emerging as potential therapeutic strategies for IBD. This review elucidates the impact of various fatty acids and lipid mediators on IBD and delves into potential therapeutic avenues stemming from these compounds.
Collapse
Affiliation(s)
- Dong Yan
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Shuyu Ye
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Cancer Research Institute, Central South University, Changsha, China
| | - Yue He
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Sidan Wang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Cancer Research Institute, Central South University, Changsha, China
| | - Yi Xiao
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Cancer Research Institute, Central South University, Changsha, China
| | - Xin Xiang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Minzi Deng
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Cancer Research Institute, Central South University, Changsha, China
| | - Weiwei Luo
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Cancer Research Institute, Central South University, Changsha, China
| | - Xuejie Chen
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Cancer Research Institute, Central South University, Changsha, China
| | - Xiaoyan Wang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Cancer Research Institute, Central South University, Changsha, China
| |
Collapse
|
37
|
Dinu LD, Gatea F, Roaming Israel F, Lakicevic M, Dedović N, Vamanu E. The Modulation Effect of a Fermented Bee Pollen Postbiotic on Cardiovascular Microbiota and Therapeutic Perspectives. Biomedicines 2023; 11:2712. [PMID: 37893086 PMCID: PMC10604238 DOI: 10.3390/biomedicines11102712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Hypertension is a frequent comorbidity in patients with heart failure; therefore, blood pressure management for these patients is widely recommended in medical guidelines. Bee pollen and postbiotics that contain inactivated probiotic cells and their metabolites have emerged as promising bioactive compounds sources, and their potential role in mitigating cardiovascular (CV) risks is currently being unveiled. Therefore, this preliminary study aimed to investigate the impact of a lactic-fermented bee pollen postbiotic (FBPP) on the CV microbiota via in vitro tests. A new isolated Lactobacillus spp. strain from the digestive tract of bees was used to ferment pollen, obtaining liquid and dried atomized caps postbiotics. The modulating effects on a CV microbiota that corresponds to the pathophysiology of hypertension were investigated using microbiological methods and qPCR and correlated with the metabolic profile. Both liquid and dried FBPPs increased the number of the beneficial Lactobacillus spp. and Bifidobacterium spp. bacteria by up to 2 log/mL, while the opportunistic pathogen E. coli, which contributes to CV pathogenesis, decreased by 3 log/mL. The short-chain fatty acid (SCFA) profile revealed a significant increase in lactic (6.386 ± 0.106 g/L) and acetic (4.284 ± 0.017 g/L) acids, both with known antihypertensive effects, and the presence of isovaleric acid, which promotes a healthy gut microbiota. Understanding the impact of the FBPP on gut microbiota could lead to innovative strategies for promoting heart health and preventing cardiovascular diseases.
Collapse
Affiliation(s)
- Laura-Dorina Dinu
- Faculty of Biotechnology, University of Agricultural Sciences and Veterinary Medicine, 011464 Bucharest, Romania;
| | - Florentina Gatea
- Centre of Bioanalysis, National Institute for Biological Sciences, 060031 Bucharest, Romania;
| | - Florentina Roaming Israel
- Faculty of Biotechnology, University of Agricultural Sciences and Veterinary Medicine, 011464 Bucharest, Romania;
| | - Milena Lakicevic
- Faculty of Agriculture, University of Novi Sad, 21000 Novi Sad, Serbia; (M.L.); (N.D.)
| | - Nebojša Dedović
- Faculty of Agriculture, University of Novi Sad, 21000 Novi Sad, Serbia; (M.L.); (N.D.)
| | - Emanuel Vamanu
- Faculty of Biotechnology, University of Agricultural Sciences and Veterinary Medicine, 011464 Bucharest, Romania;
| |
Collapse
|
38
|
Ma M, Quan M, Zhang J, Zhang A, Gao P, Shang Q, Yu G. In Vitro Fermentation of Polysaccharide from Edible Alga Enteromorpha clathrata by the Gut Microbiota of Patients with Ulcerative Colitis. Nutrients 2023; 15:4122. [PMID: 37836407 PMCID: PMC10574352 DOI: 10.3390/nu15194122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/18/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Dietary intake of the sulfated polysaccharide from edible alga E. clathrata (ECP) has recently been illustrated to attenuate ulcerative colitis (UC) by targeting gut dysbiosis in mice. However, ECP is not easily absorbed in the gut and, as a potential candidate for next-generation prebiotics development, how it is fermented by human gut microbiota has not been characterized. Here, using in vitro anaerobic fermentation and 16S high-throughput sequencing, we illustrate for the first time the detailed fermentation characteristics of ECP by the gut microbiota of nine UC patients. Our results indicated that, compared to that of glucose, fermentation of ECP by human gut microbiota produced a higher amount of anti-inflammatory acetate and a lower amount of pro-inflammatory lactate. Additionally, ECP fermentation helped to shape a more balanced microbiota composition with increased species richness and diversity. Moreover, ECP significantly stimulated the growth of anti-colitis bacteria in the human gut, including Bacteroides thetaiotaomicron, Bacteroides ovatus, Blautia spp., Bacteroides uniformis, and Parabacteroides spp. Altogether, our study provides the first evidence for the prebiotic effect of ECP on human gut microbiota and sheds new light on the development of ECP as a novel prebiotic candidate for the prevention and potential treatment of UC.
Collapse
Affiliation(s)
- Mingfeng Ma
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (M.M.); (M.Q.); (J.Z.)
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - Min Quan
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (M.M.); (M.Q.); (J.Z.)
| | - Jiaxue Zhang
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (M.M.); (M.Q.); (J.Z.)
| | - Aijun Zhang
- Qilu Hospital of Shandong University (Qingdao), Qingdao 266035, China; (A.Z.); (P.G.)
| | - Puyue Gao
- Qilu Hospital of Shandong University (Qingdao), Qingdao 266035, China; (A.Z.); (P.G.)
| | - Qingsen Shang
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (M.M.); (M.Q.); (J.Z.)
- Qingdao Marine Biomedical Research Institute, Qingdao 266071, China
| | - Guangli Yu
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (M.M.); (M.Q.); (J.Z.)
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
| |
Collapse
|
39
|
Huan Q, Peng J, Chang Y, Zhang Q, Xing T, Jiang D, Chen W, Shen X, Bian Z, Xiao H. Activation of P2Y1R impedes intestinal mucosa repair during colitis. Int J Biol Sci 2023; 19:4360-4375. [PMID: 37781034 PMCID: PMC10535714 DOI: 10.7150/ijbs.82302] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 08/01/2023] [Indexed: 10/03/2023] Open
Abstract
Delayed intestinal mucosal healing is one of the pathogenic bases for the recurrence of inflammatory bowel disease (IBD), but how the IBD inflammatory environment impedes intestinal mucosa repair remains unclear. Adenosine diphosphate (ADP) is an endogenous ligand of P2Y1R that is highly produced at sites of inflammation. We herein identify a novel role of ADP to directly facilitate inflammation-induced epithelial permeability, delay wound healing, and disrupt tight junction integrity, and we found that P2Y1R, a receptor preferentially activated by ADP, was significantly upregulated in the colonic mucosa of ulcerative colitis (UC) patients and in colonic epithelial cells of colitis mice. Inhibition of P2Y1R significantly increased the epithelial permeability, decreased the wound healing capacity, and impaired the tight junction integrity in TNF-α-challenged Caco-2 cells. In parallel, the same effects in promoting intestinal mucosa repair were observed in DSS-induced colitis in P2Y1R-/- mice. Mechanistic investigation revealed that P2Y1R inhibition facilitated epithelial AMP-activated protein kinase (AMPK) phosphorylation and gut microbiota homeostasis reconstruction. Taken together, these findings highlight that P2Y1R activation plays an important role in impeding intestinal mucosa repair during colitis, and that P2Y1R is an attractive target for the therapy of IBD.
Collapse
Affiliation(s)
- Qiuchan Huan
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants and The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China
| | - Jiao Peng
- Department of Pharmacy, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yaoyao Chang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Qiansheng Zhang
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tianhang Xing
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Danling Jiang
- Department of Gastroenterology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Wenke Chen
- Department of Gastroenterology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants and The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China
| | - Zhaoxiang Bian
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
| | - Haitao Xiao
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
40
|
Aguilera M, Daddaoua A. Prebiotics and Probiotics: Healthy Biotools for Molecular Integrative and Modulation Approaches. Int J Mol Sci 2023; 24:ijms24087559. [PMID: 37108721 PMCID: PMC10144313 DOI: 10.3390/ijms24087559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
The scope of this Special Issue is to highlight and expand our knowledge on the molecular mechanisms of prebiotics and probiotics, as well as to offer a broad overview of current advancements and future directions in this research field [...].
Collapse
Affiliation(s)
- Margarita Aguilera
- Department of Microbiología, Pharmacy School, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS), 18014 Granada, Spain
- Institute of Nutrition and Food Technology "José Mataix", Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. Armilla, 18016 Granada, Spain
| | - Abdelali Daddaoua
- Instituto de Investigación Biosanitaria (IBS), 18014 Granada, Spain
- Institute of Nutrition and Food Technology "José Mataix", Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. Armilla, 18016 Granada, Spain
- Department of Biochemistry and Molecular Biology II, Pharmacy School, University of Granada, 18071 Granada, Spain
| |
Collapse
|