1
|
Oliveira M, Barbosa J, Teixeira P. Listeria monocytogenes gut interactions and listeriosis: Gut modulation and pathogenicity. Microbiol Res 2025; 297:128187. [PMID: 40279724 DOI: 10.1016/j.micres.2025.128187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
Following ingestion via contaminated food, Listeria monocytogenes faces multiple hurdles through the human digestive system, thereby influencing its capacity to cause infection. This review provides a comprehensive overview of the multifaceted mechanisms employed by L. monocytogenes to overcome gastrointestinal hurdles and interact with the host's microbiota, facing chemical and physical barriers such as saliva, stomach acidity, bile salts and mechanical clearance. Proposed evasion strategies will be highlighted, exploring the bacteriocins produced by L. monocytogenes, such as the well-described bacteriocin Listeriolysin S (LLS), a bacteriocin that inhibits inflammogenic species - Lmo2776, and a phage tail-like bacteriocin, monocin. The competitive dynamic interactions within the gut microbiota, as well as the modulation of microbiota composition and immune responses, will also be explored. Finally, the adhesion and invasion of the intestinal epithelium by L. monocytogenes is described, exploring the mechanism of pathogenesis, biofilm and aggregation capacities and other virulence factors. Unlike previous reviews that may focus on individual aspects of L. monocytogenes pathogenicity, this review offers a holistic perspective on the bacterium's ability to persist and cause infection, integrating information about survival strategies, including bacteriocin production, immune modulation, and virulence factors. By connecting recent findings on microbial interactions and infection dynamics, this review incorporates recent developments in the field and connects various lines of research that explore both host and microbial factors influencing infection outcomes.
Collapse
Affiliation(s)
- M Oliveira
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho 1327, 4169-005, Porto, Portugal
| | - J Barbosa
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho 1327, 4169-005, Porto, Portugal
| | - P Teixeira
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua de Diogo Botelho 1327, 4169-005, Porto, Portugal.
| |
Collapse
|
2
|
Liang S, Zhao D, Liu X, Liu B, Li Y. The stomach, small intestine, and colon-specific gastrointestinal tract delivery systems for bioactive nutrients. Adv Colloid Interface Sci 2025; 341:103503. [PMID: 40209595 DOI: 10.1016/j.cis.2025.103503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 02/13/2025] [Accepted: 03/29/2025] [Indexed: 04/12/2025]
Abstract
Oral administration is a convenient way to deliver bioactive nutrients. However, the complex and dynamic environment of the gastrointestinal (GI) tract poses distinct challenges. These include the acidic environment of the stomach, limited transport across the GI mucosa, and the risk of enzymatic degradation, all of which can compromise the nutritional effectiveness of orally delivered nutrients. In response to these challenges, various GI tract delivery systems have been developed to target specific regions, such as the stomach, small intestine, or colon, to precisely control the release of bioactive nutrients and enhance their health-promoting benefits. This review critically examines the principles underlying stomach-, small intestine-, and colon-targeted delivery systems, highlighting the selection of appropriate wall materials and the interactions between delivery systems and the mucosal epithelial barrier. Moreover, we describe relevant biological models and quantitative analyses to measure these interactions. In particular, we emphasize the significant advantages offered by colon-targeted delivery systems in maintaining a healthy colonic microenvironment. This review aims to inspire novel concepts and stimulate further research into GI tract delivery systems, offering promising avenues for maximizing the therapeutic effects of bioactive nutrients in practical applications.
Collapse
Affiliation(s)
- Shuang Liang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; Central Laboratory, NMPA Key Laboratory for Dental Materials, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Dongyu Zhao
- Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Xiangyu Liu
- Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Bin Liu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yuan Li
- Research Center of Food Colloids and Delivery of Functionality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
3
|
Fan S, Cai X, Cui W, Ma P, Wu M, Guo J, Zhang Y, Xuan K, Li Z. Restoration of gut integrity by Bacteroides acidifaciens in water-deprived conditions. Biochem Biophys Res Commun 2025; 767:151917. [PMID: 40318377 DOI: 10.1016/j.bbrc.2025.151917] [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: 03/30/2025] [Revised: 04/19/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Water scarcity exerts profound physiological impacts, yet its effects on intestinal microbiota-barrier crosstalk remain poorly understood. This study investigates water-deprivation-induced gut dysbiosis and identifies Bacteroides acidifaciens (B.acidifaciens) as a critical mediator of epithelial barrier resilience. Using a murine model of graded water restriction, we observed colon shortening, villus atrophy, goblet cell loss, and barrier disruption correlated with water-deprivation severity. Oral supplementation of B.acidifaciens restored colonic architecture, enhanced mucin secretion, and ameliorated barrier dysfunction in water-deprived mice. Mechanistically, B.acidifaciens maintained barrier homeostasis by directly stimulating mucus production in intestinal epithelial cells and upregulating the expression of tight junction proteins. These findings establish B.acidifaciens as a critical species in mitigating water-deprivation-induced colon injury and advance our understanding of microbiota-directed interventions for dehydration-associated gastrointestinal disorders.
Collapse
Affiliation(s)
- Siyuan Fan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Disease, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Xinyue Cai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Disease, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Wenbo Cui
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Disease, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Peisen Ma
- Faculty of Medicine and Health University of Sydney, Camperdown, NSW, 2050, Sydney, Australia
| | - Meiling Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Disease, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jing Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Disease, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yipeng Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Disease, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Kun Xuan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Disease, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Zihan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Disease, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
4
|
Castranova D, Kenton MI, Kraus A, Dell CW, Park JS, Venero Galanternik M, Park G, Lumbantobing DN, Dye L, Marvel M, Iben J, Taimatsu K, Pham V, Willms RJ, Blevens L, Robertson TF, Hou Y, Huttenlocher A, Foley E, Parenti LR, Frazer JK, Narayan K, Weinstein BM. The axillary lymphoid organ is an external, experimentally accessible immune organ in the zebrafish. J Exp Med 2025; 222:e20241435. [PMID: 40167600 PMCID: PMC11960710 DOI: 10.1084/jem.20241435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/13/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Lymph nodes and other secondary lymphoid organs play critical roles in immune surveillance and immune activation in mammals, but the deep internal locations of these organs make it challenging to image and study them in living animals. Here, we describe a previously uncharacterized external immune organ in the zebrafish ideally suited for studying immune cell dynamics in vivo, the axillary lymphoid organ (ALO). This small, translucent organ has an outer cortex teeming with immune cells, an inner medulla with a mesh-like network of fibroblastic reticular cells along which immune cells migrate, and a network of lymphatic vessels draining to a large adjacent lymph sac. Noninvasive high-resolution imaging of transgenically marked immune cells can be carried out in ALOs of living animals, which are readily accessible to external treatment. This newly discovered tissue provides a superb model for dynamic live imaging of immune cells and their interaction with pathogens and surrounding tissues, including blood and lymphatic vessels.
Collapse
Affiliation(s)
- Daniel Castranova
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Madeleine I. Kenton
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Aurora Kraus
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Christopher W. Dell
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Jong S. Park
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Marina Venero Galanternik
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Gilseung Park
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Daniel N. Lumbantobing
- Division of Fishes, Department of Vertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA
| | - Louis Dye
- Microscopy and Imaging Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Miranda Marvel
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - James Iben
- Molecular Genomics Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Kiyohito Taimatsu
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Van Pham
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Reegan J. Willms
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Lucas Blevens
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tanner F. Robertson
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Yiran Hou
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Lynne R. Parenti
- Division of Fishes, Department of Vertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA
| | - J. Kimble Frazer
- Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kedar Narayan
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Brant M. Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
5
|
Chen LR, Zhou SS, Yang JX, Liu XQ. Effect of hypoxia on the mucus system and intragastric microecology in the gastrointestinal tract. Microb Pathog 2025; 205:107615. [PMID: 40355054 DOI: 10.1016/j.micpath.2025.107615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 04/03/2025] [Accepted: 04/17/2025] [Indexed: 05/14/2025]
Abstract
Digestive diseases have a high incidence worldwide, with various geographic, age, and gender factors influencing the occurrence and development of the diseases. The main etiologic factors involve genetics, environment, lifestyle, and dietary habits. In a low-oxygen environment, however, the body's tissue cells activate hypoxia-inducible factor (HIF), which produces different inflammatory mediators. Hypoxia impacts health at the molecular level by modulating cellular stress responses, metabolic pathways, and immune functions. It also alters gene expression and cellular behavior, thereby affecting gastrointestinal function. Under normal physiological conditions, the gastrointestinal mucus system serves as a crucial protective barrier, defending against mechanical injury, pathogenic invasion, and exposure to harmful chemicals. The integrity and functionality of this barrier are dependent on the synthesis and regulation of mucins and mucus, which are influenced by multiple factors. Additionally, the composition and diversity of the gastric microbiota are shaped by factors such as Helicobacter pylori infection, diet, and lifestyle. A balanced gastric microbiota supports gastrointestinal health and fortifies the mucus barrier. However, hypoxia can disrupt this equilibrium, leading to inflammation, alterations in the mucus layer, and destabilization of the gastric microbiota. Understanding the interplay between hypoxia, the mucus system, and the gastric microbiota is essential for identifying novel therapeutic strategies. Future research should elucidate the mechanisms through which hypoxia influences these systems and develop interventions to mitigate its adverse effects on gastrointestinal health. We examined the impact of hypoxia on the gastrointestinal mucus system and gastric microbiota, highlighting its implications for human health and potential therapeutic approaches.
Collapse
Affiliation(s)
- Li Rong Chen
- Qinghai University, Xining, 810001, PR China; Affiliated People's Hospital of Qinghai University, Xining, 810001, PR China
| | - Si Si Zhou
- Affiliated People's Hospital of Qinghai University, Xining, 810001, PR China; Department of Gastroenterology, Qinghai Provincial People's Hospital, Xining, 810001, PR China; Qinghai Provincial Clinical Medical Research Center for Digestive Diseases, Xining, 810001, PR China.
| | - Ji Xiang Yang
- Qinghai University, Xining, 810001, PR China; Affiliated People's Hospital of Qinghai University, Xining, 810001, PR China
| | - Xiao Qian Liu
- Qinghai University, Xining, 810001, PR China; Affiliated People's Hospital of Qinghai University, Xining, 810001, PR China
| |
Collapse
|
6
|
Schwerdtfeger LA, Montini F, Chitnis T, Cox LM, Weiner HL. Faecal mucoprotein MUC2 is decreased in multiple sclerosis and is associated with mucin degrading bacteria. EBioMedicine 2025; 116:105721. [PMID: 40344717 DOI: 10.1016/j.ebiom.2025.105721] [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: 12/30/2024] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND The gut microbiome is altered in MS and may contribute to disease by disrupting the intestinal barrier. The colonic mucus barrier, which is primarily composed of mucin protein 2 (MUC2), plays a crucial role in providing a barrier between colonic epithelial cells and the microbiome. Disruption of intestinal epithelial and mucus barriers has been reported in inflammatory bowel disease (IBD) and Parkinson's disease (PD) but has not been studied in the context of the microbiome in multiple sclerosis (MS). METHODS We investigated the epithelial tight junction protein zonulin occludins 1 (ZO-1), mucus protein MUC2, inflammatory stool markers (calprotectin), and gut microbiota composition in a cohort of subjects with relapsing and progressive MS. FINDINGS MUC2 was decreased in stool of subjects with both relapsing and progressive MS. ZO-1 was elevated in the serum of subjects with progressive MS but was not altered in the stool. Inflammatory markers typically elevated in IBD and PD, including calprotectin, were not altered in MS stool, suggesting disease specificity of altered gut physiology in MS. Microbiota with known mucus degrading capacity were elevated in the stool of subjects with MS and negatively correlated with mucus protein levels. INTERPRETATION Taken together, these findings suggest reduced gut barrier function in MS which is linked to increased mucin degrading bacteria. FUNDING This work was supported by grants from the National MS Society, the NIH/NINDS, the Nancy Davis Race to Erase MS Young Investigator Award, the Water Cove Charitable Foundation, and the Clara E. and John H. Ware Jr. FOUNDATION
Collapse
Affiliation(s)
- Luke A Schwerdtfeger
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Federico Montini
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Tanuja Chitnis
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Laura M Cox
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
7
|
Fan F, Guo R, Pan K, Xu H, Chu X. Mucus and mucin: changes in the mucus barrier in disease states. Tissue Barriers 2025:2499752. [PMID: 40338015 DOI: 10.1080/21688370.2025.2499752] [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: 12/17/2024] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 05/09/2025] Open
Abstract
In this review we discuss mucus, the viscoelastic secretion from goblet or mucous producing cells that covers and protects all non-keratinized wet epithelial surfaces. In addition to the surface of organs directly contacting with the external environment such as the eyes, this layer provides protection to the underlying gastrointestinal, respiratory and female reproductive tracts by trapping pathogens, irritants, environmental fine particles and potentially harmful foreign substances. Mucins, the primary structural components of mucus, form structurally different mucus layers at different sites in a process regulated by a variety of factors. Currently, more and more studies have shown that the mucus barrier is not only closely related to various intestinal mucus diseases, but also involved in the occurrence and development of various airway diseases and mucus-related diseases, thus it may become a new target for the treatment of various related diseases in the future. Since the dysfunction of the mucous layer is closely related to various pathological processes, in-depth understanding of its molecular mechanism and physiological role is of great theoretical and practical significance for disease prevention and treatment. Here, we discuss different aspects of the mucus layer by focusing on its chemical composition, synthetic pathways, and some of the characteristics of the mucus layer in physiological and pathological situations.
Collapse
Affiliation(s)
- Fangfang Fan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Ruihan Guo
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Kun Pan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Hongye Xu
- Quality Assurance department, Tongling Institutes for Food and Drug Control, Tongling, China
| | - Xiaoqin Chu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China
- Engineering Technology Research Center of Modern Pharmaceutical Preparation, Hefei, Anhui Province, China
| |
Collapse
|
8
|
Plenge M, Schnepel N, Müsken M, Rohde J, Goethe R, Breves G, Mazzuoli-Weber G, Benz P. Development and characterization of a 2D porcine colonic organoid model for studying intestinal physiology and barrier function. PLoS One 2025; 20:e0312989. [PMID: 40333830 PMCID: PMC12057940 DOI: 10.1371/journal.pone.0312989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 04/03/2025] [Indexed: 05/09/2025] Open
Abstract
The porcine colon epithelium plays a crucial role in nutrient absorption, ion transport, and barrier function. However ethical concerns necessitate the development of alternatives to animal models for its study. The objective of this study was to develop and characterize a two-dimensional (2D) in vitro model of porcine colonic organoids that closely mimics native colon tissue, thereby supporting in vitro research in gastrointestinal physiology, pathology, and pharmacology. Porcine colonic crypts were isolated and cultured in three-dimensional (3D) organoid systems, which were subsequently disaggregated to form 2D monolayers on transwell inserts. The integrity of the monolayers was evaluated through the measurement of transepithelial electrical resistance (TEER) and electron microscopy. The functional prerequisites of the model were evaluated through the measurement of the mRNA expression of key ion channels and transporters, using quantitative RT-PCR. Ussing chamber experiments were performed to verify physiological activity. The 2D monolayer displayed robust TEER values and retained structural characteristics, including microvilli and mucus-secreting goblet cells, comparable to those observed in native colon tissue. Gene expression analysis revealed no significant differences between the 2D organoid model and native tissue with regard to critical transporters. Ussing chamber experiments demonstrated physiological responses that were consistent with those observed in native colonic tissue. In conclusion, 2D porcine colonic organoid model can be recommended as an accurate representation of the physiological and functional attributes of the native colon epithelium. This model offers a valuable tool for investigating intestinal barrier properties, ion transport, and the pathophysiology of gastrointestinal diseases, while adhering to the 3R principles.
Collapse
Affiliation(s)
- Masina Plenge
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Nadine Schnepel
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Mathias Müsken
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Judith Rohde
- Institute of Microbiology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Ralph Goethe
- Institute of Microbiology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Gerhard Breves
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Gemma Mazzuoli-Weber
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Pascal Benz
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
9
|
Pletsch EA, Dawson HD, Cheung L, Ragonese JS, Chen CT, Smith AD. A type 4 resistant potato starch alters the cecal microbiome, gene expression and resistance to colitis in mice fed a Western diet based on NHANES data. Food Funct 2025; 16:3439-3464. [PMID: 40207550 DOI: 10.1039/d4fo04697h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Four major types of resistant starch (RS1-4) are present in foods and can be fermented to produce short-chain fatty acids (SCFAs), alter the microbiome and modulate post-prandial glucose metabolism. While studies in rodents have examined the effects of RS4 consumption on the microbiome, fewer have examined its effect on gene expression in the cecum or colon or resistance to bacterial-induced colitis, and those that have, use diets that do not reflect what is typically consumed by humans. Here we fed mice a Total Western Diet (TWD), based on National Health and Nutrition Examination Survey (NHANES) data for 6-7 weeks and then supplemented their diet with 0, 2, 5, or 10% of the RS4, Versafibe 1490™ (VF), a phosphorylated and cross-linked potato starch. After three weeks, mice were infected with Citrobacter rodentium (Cr) to induce colitis. Infected mice fed the 10% VF diet had the highest levels of Cr fecal excretion at days 4, 7 and 11 post-infection. Infected mice fed the 5% and 10%VF diets had increased hyperplasia and colonic damage compared with the control. Changes in bacterial genera relative abundance, and alpha and beta diversity due to diet were most evident in mice fed 10% VF. Cr infection also resulted in specific changes to the microbiome and gene expression both in the cecum and the colon compared with diet alone, including the expression of multiple antimicrobial genes, Reg3b, Reg3g, NOS2 and Ifng. These results demonstrate that VF, a RS4, alters cecal and colonic gene expression, the microbiome composition and resistance to bacterial-induced colitis.
Collapse
Affiliation(s)
- Elizabeth A Pletsch
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Rm. 228, Bldg. 307C, BARC-East, 10300 Baltimore Ave., Beltsville, MD, 20705, USA.
| | - Harry D Dawson
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Rm. 228, Bldg. 307C, BARC-East, 10300 Baltimore Ave., Beltsville, MD, 20705, USA.
| | - Lumei Cheung
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Rm. 228, Bldg. 307C, BARC-East, 10300 Baltimore Ave., Beltsville, MD, 20705, USA.
| | - Jack S Ragonese
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Rm. 228, Bldg. 307C, BARC-East, 10300 Baltimore Ave., Beltsville, MD, 20705, USA.
| | - Celine T Chen
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Rm. 228, Bldg. 307C, BARC-East, 10300 Baltimore Ave., Beltsville, MD, 20705, USA.
| | - Allen D Smith
- Diet, Genomics, and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, United States Department of Agriculture, Rm. 228, Bldg. 307C, BARC-East, 10300 Baltimore Ave., Beltsville, MD, 20705, USA.
| |
Collapse
|
10
|
Venkatasamy L, Iannucci J, Pereverzev A, Hoar J, Huber E, Ifegbo A, Dominy R, El-Hakim Y, Mani KK, Dabney A, Pilla R, Sohrabji F, Shapiro LA. Systemic IGF-1 administration prevents traumatic brain injury induced gut permeability, dysmorphia, dysbiosis, and the increased number of immature dentate granule cells. Acta Neuropathol Commun 2025; 13:90. [PMID: 40319295 PMCID: PMC12049052 DOI: 10.1186/s40478-025-01998-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/03/2025] [Indexed: 05/07/2025] Open
Abstract
Traumatic brain injury (TBI) occurs in 2-3 million Americans each year and is a leading cause of death and disability. Among the many physiological consequences of TBI, the hypothalamic pituitary axis (HPA) is particularly vulnerable, including a reduction in growth hormone (GH) and insulin-like growth factor (IGF-1). Clinical and preclinical supplementation of IGF-1 after TBI has exhibited beneficial effects. IGF-1 receptors are prominently observed in many tissues, including in the brain and in the gastrointestinal (GI) system. In addition to causing damage in the brain, TBI also induces GI system damage, including inflammation and alterations to intestinal permeability and the gut microbiome. The goal of this study was to assess the effects of systemic IGF-1 treatment in a rat model of TBI on GI outcomes. Because GI dysfunction has been linked to hippocampal dysfunction, we also examined proliferation and immature granule cells in the hippocampal dentate gyrus. 10-week-old male rats were treated with an intraperitoneal (i.p.) dose of IGF-1 at 4 and 24 h after lateral fluid percussion injury (FPI). At 3- and 35-days post-injury (DPI), gut permeability, gut dysmorphia, the fecal microbiome, and the hippocampus were assessed. FPI-induced permeability of the blood-gut-barrier, as measured by elevated gut metabolites in the blood, and this was prevented by the IGF-1 treatment. Gut dysmorphia and alterations to the microbiome were also observed after FPI and these effects were ameliorated by IGF-1, as was the increase in immature granule cells in the hippocampus. These findings suggest that IGF-1 can target gut dysfunction and damage after TBI, in addition to its role in influencing adult hippocampal neurogenesis.
Collapse
Affiliation(s)
- Lavanya Venkatasamy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A and M University, 8447 Riverside Pkwy, Bryan, TX, 77807-3260, USA
| | - Jaclyn Iannucci
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A and M University, 8447 Riverside Pkwy, Bryan, TX, 77807-3260, USA
| | - Aleksandr Pereverzev
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A and M University, 8447 Riverside Pkwy, Bryan, TX, 77807-3260, USA
| | - Jonathan Hoar
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A and M University, 8447 Riverside Pkwy, Bryan, TX, 77807-3260, USA
| | - Emily Huber
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A and M University, 8447 Riverside Pkwy, Bryan, TX, 77807-3260, USA
| | - Angel Ifegbo
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A and M University, 8447 Riverside Pkwy, Bryan, TX, 77807-3260, USA
| | - Reagan Dominy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A and M University, 8447 Riverside Pkwy, Bryan, TX, 77807-3260, USA
| | - Yumna El-Hakim
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A and M University, 8447 Riverside Pkwy, Bryan, TX, 77807-3260, USA
| | - Kathiresh Kumar Mani
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A and M University, 8447 Riverside Pkwy, Bryan, TX, 77807-3260, USA
| | - Alan Dabney
- Department of Statistics, College of Arts and Sciences, Texas A and M University, College Station, TX, USA
| | - Rachel Pilla
- Gastrointestinal Laboratory, College of Veterinary Medicine and Biomedical Sciences, Texas A and M University, College Station, TX, USA
| | - Farida Sohrabji
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A and M University, 8447 Riverside Pkwy, Bryan, TX, 77807-3260, USA
| | - Lee A Shapiro
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A and M University, 8447 Riverside Pkwy, Bryan, TX, 77807-3260, USA.
| |
Collapse
|
11
|
Park I, Nam H, Ravichandran S, Wall EH, Lillehoj HS. Phytochemicals act holistically to enhance host defenses during poultry coccidiosis. Poult Sci 2025; 104:105042. [PMID: 40120241 PMCID: PMC11987653 DOI: 10.1016/j.psj.2025.105042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/12/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025] Open
Abstract
This study was conducted to investigate the effects of a phytochemical mixture containing full spectrum cinnamon, clove, and oregano essential oils (CCO) on the growth performance, intestinal immunity, and intestinal integrity of broilers infected with coccidiosis. In chicken macrophage cells (CMCs), inflammation was induced with 1.0 µg/mL LPS, followed by stimulating with CCO at three concentrations (1.0, 10.0, and 100 µg/mL) and measuring the gene expression levels of IL-1β and IL-8. In chicken intestinal epithelial cells (IECs), CCO was added and cultured, and the gene expression levels of occludin, ZO-1, and MUC2 were measured. In the in vivo experiment, one hundred and twenty male broiler chickens (0-day-old) were allocated into three treatment groups: (1) basal diet without infection (NC), (2) basal diet with E. maxima infection (PC), and (3) CCO at 4.5 mg/kg feed with E. maxima infection (CCO). Body weight (BW) was measured on days 0, 7, 14, 20, and 22. PC and CCO groups were orally infected with E. maxima on day 14. Jejunal samples were collected on day 22 to conduct gene expression analysis of cytokines, TJ proteins, and antioxidant enzymes. CCO significantly decreased IL-1β and IL-8 in CMCs and increased ZO-1 and MUC2 in IECs in a dose-dependent manner. In the E. maxima-infected groups, dietary CCO tended to mitigate BW loss due to infection. Upon infection, proinflammatory cytokines were suppressed in the CCO group compared to the PC group. Dietary CCO also increased the expression of occludin and JAM-2 in the jejunum. However, CCO did not reduce the oocyst number in coccidiosis-infected chickens. These results suggest that dietary CCO supplementation may improve intestinal immunity and permeability, helping to reduce productivity losses in E. maxima-infected broilers through gut physiological responses, rather than direct antimicrobial effects. These results show the advantage of using in vitro screening based on host-mediated responses, and not on direct pathogen killing, when exploring new phytochemicals to mitigate disease response to reduce economic losses due to coccidiosis.
Collapse
Affiliation(s)
- Inkyung Park
- Animal Bioscience and Biotechnology Laboratory, United States Department of Agriculture, Beltsville Agricultural Research Center, Beltsville, MD 20705, USA
| | - Hyoyoun Nam
- Animal Bioscience and Biotechnology Laboratory, United States Department of Agriculture, Beltsville Agricultural Research Center, Beltsville, MD 20705, USA
| | | | - Emma H Wall
- Nutreco Exploration, Nutreco Switzerland GmbH, 9320 Arbon, Switzerland
| | - Hyun S Lillehoj
- Animal Bioscience and Biotechnology Laboratory, United States Department of Agriculture, Beltsville Agricultural Research Center, Beltsville, MD 20705, USA.
| |
Collapse
|
12
|
Sharafian Z, Littlejohn PT, Michalski C, Sousa JA, Cheung J, Hill M, Piper H, Jacobson K, Lavoie PM, Allaire JM, Vallance BA. Crosstalk with infant-derived Th17 cells, as well as exposure to IL-22 promotes maturation of intestinal epithelial cells in an enteroid model. Front Immunol 2025; 16:1582688. [PMID: 40375988 PMCID: PMC12078324 DOI: 10.3389/fimmu.2025.1582688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/08/2025] [Indexed: 05/18/2025] Open
Abstract
Introduction The intestinal epithelium of human infants is developmentally immature compared to that of adults. Exactly how this immaturity affects key epithelial functions and their interactions with nearby immune cells remains an understudied area of research, partly due to limited access to non-diseased infant gut tissues. Human intestinal organoids, or "mini guts" generated from tissue stem cells, are promising models for investigating intestinal biology and disease mechanisms. These three-dimensional structures closely mimic their tissue of origin, including cellular physiology and genetics. We have also previously shown that neonatal Th17 cells represent a distinct cell population with a cytokine profile skewed toward IL-22 production rather than IL-17A, as seen in adult Th17 cells. Methods In this study, we sought to model the impact of neonatal-derived Th17 cytokine, namely IL-22 and the intestinal epithelium using infant-derived ileal enteroids. We generated enteroids from ileal biopsies from infants (< 6 months old) and cultured them for seven days with standard organoid growth media, organoid media supplemented with conditioned media from cord-blood-derived Th17 cells, or media supplemented with recombinant IL-22. We assessed morphological changes and conducted transcriptomics profiling via RNAseq. Results Exposing enteroids to neonatal Th17-cells-derived conditioned media led to enhanced growth, maturation, and differentiation as compared to control media. These effects were ablated when an IL-22 neutralizing antibody was used, while conversely, supplementing with recombinant IL-22 mimicked the Th17 effects, increasing intestinal epithelial cell proliferation and inducing marked differentiation of secretory cells. Our transcriptomic profiling similarly demonstrated significant changes in response to IL-22 with downregulation of Wnt and Notch signaling and upregulation of immune pathways, particularly interferon signaling. The transcriptomic data also suggested that IL-22 treatment led to changes in cell type composition with an increase in stem- and progenitor cells at the expense of enterocytes. Conclusion Taken together, our data suggests that early-life intestinal development is likely influenced by IL-22-dependent crosstalk between the infant epithelium and exposure to neighboring Th17 cells. This promotes epithelial cell maturation and immune readiness, reflected at both the morphological and molecular levels. Our work also provides a relevant framework for studying healthy infant gut development, which can be further leveraged to examine early-life gastrointestinal disorders, model complex human disease, and therapeutic testing while reducing reliance on animal models.
Collapse
Affiliation(s)
- Zohreh Sharafian
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Paula T. Littlejohn
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Vancouver, BC, Canada
| | - Christina Michalski
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - James A. Sousa
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Janelle Cheung
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Mariana Hill
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Hannah Piper
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Kevan Jacobson
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Pascal M. Lavoie
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Joannie M. Allaire
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Bruce A. Vallance
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
| |
Collapse
|
13
|
Zhang Y, Zhou Z, Zhang Z, Liu Y, Ji W, Wang J, Wang K, Li Q. Lentinan mitigates ulcerative colitis via the IL-22 pathway to repair the compromised mucosal barrier and enhance antimicrobial defense. Int J Biol Macromol 2025; 307:141784. [PMID: 40054799 DOI: 10.1016/j.ijbiomac.2025.141784] [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: 11/04/2024] [Revised: 02/15/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
Ulcerative colitis (UC) involves chronic, complex pathology of the intestinal mucosa. Current treatments are limited in efficacy and associated with adverse effects, highlighting the urgent need for improved therapeutic options. Lentinan (LNT), a polysaccharide drug commonly used in clinical immune modulation therapies, shows potential for UC treatment, though its specific targets and mechanisms remain unclear. In this study, LNT administration effectively mitigated DSS-induced colitis in mice, enhanced mucosal barrier function and antimicrobial defense. Specifically, LNT modulated the balance between tissue-resident and infiltrating macrophages, thereby improving pathogen clearance and enhancing the immunological barrier. Notably, we identified a novel effect of LNT in regulating the macrophage Dectin-1-ILC3 axis to increase IL-22 secretion. This led to the modulation of epithelial O-glycan fucosylation, antimicrobial peptides, and epithelial stem cells, thereby strengthening antimicrobial defenses and the physicochemical barrier. Neutralization with anti-IL-22 antibodies diminished the therapeutic effect of LNT in UC, underscoring the critical role of IL-22 in LNT-mediated treatment. Overall, this study highlights the potential of LNT as a novel therapeutic agent for UC, offering new insights into its molecular mechanisms and clinical application.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030 Wuhan, China
| | - Zhihong Zhou
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Zeming Zhang
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Yan Liu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030 Wuhan, China
| | - Wenting Ji
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Jinglin Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030 Wuhan, China.
| | - Kaiping Wang
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, 430030 Wuhan, China.
| | - Qiang Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030 Wuhan, China.
| |
Collapse
|
14
|
Huyghe P, Ceulemans M, Keita ÅV, Söderholm J, Depoortere I, Tack J, Wauters L, Vanuytsel T. The Duodenal Microenvironment in Functional Dyspepsia. J Neurogastroenterol Motil 2025; 31:186-198. [PMID: 40205896 PMCID: PMC11986653 DOI: 10.5056/jnm24176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/20/2025] [Accepted: 02/22/2025] [Indexed: 04/11/2025] Open
Abstract
Functional dyspepsia (FD) is a chronic gastrointestinal disorder without a readily identifiable organic cause, resulting in bothersome upper abdominal symptoms. It is a highly prevalent disorder of which the pathophysiology remains mostly elusive, despite intensive research efforts. However, recent studies have found alterations in the microenvironment of the duodenum in patients with FD. In this review we summarize the duodenal microenvironment in homeostatic conditions and the alterations found in patients with FD, highlighting the similarities and discrepancies between different studies. The most consistent findings, being an impaired duodenal barrier and duodenal immune activation, are reviewed. We discuss the potential triggers for these observed alterations, including psychological comorbidities, luminal alterations and food related triggers. In summary, this review presents the evidence of molecular and cellular changes in patients with FD, with an impaired duodenal barrier and activated mucosal eosinophils and mast cells, challenging the notion that FD is purely functional, and offering different targets for potential future treatments.
Collapse
Affiliation(s)
- Pauline Huyghe
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Matthias Ceulemans
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Åsa V Keita
- Department of Biomedical and Clinical Sciences and Department of Surgery, Linköping University, Linköping, Sweden
| | - Johan Söderholm
- Department of Biomedical and Clinical Sciences and Department of Surgery, Linköping University, Linköping, Sweden
| | - Inge Depoortere
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Jan Tack
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Lucas Wauters
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Tim Vanuytsel
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
15
|
Kovanda L, Hejna M, Du T, Liu Y. Butyrate Derivatives Exhibited Anti-Inflammatory Effects and Enhanced Intestinal Barrier Integrity in Porcine Cell Culture Models. Animals (Basel) 2025; 15:1289. [PMID: 40362102 PMCID: PMC12071038 DOI: 10.3390/ani15091289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/25/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Butyrate and its derivatives may influence inflammatory status and physiology in a variety of organisms and organ systems. Inflammatory conditions of the gastrointestinal tract, such as post-weaning diarrhea, negatively impact swine. Dietary intervention with butyrate-based compounds should be considered a strategy to improve disease resistance in pigs. We aimed to assess the properties of different forms of butyrate treatments using porcine cell culture experiments. This assessment may inform future in vivo feed experiments designed to determine its potential application of the dietary supplements for pigs. An intestinal porcine enterocyte cell line, IPEC-J2, was seeded at 5 × 103 cells/mL in 96-well plates to confirm cell viability by MTT assay for each dose range used in the current experiments (0, 0.5, 1, 2, 4 mM butyric acid or tributyrin; 0, 1, 2, 4, 8 mM sodium butyrate or monobutyrin). For transepithelial electrical resistance (TEER) analysis, IPEC-J2 was seeded at 5 × 105 cells/mL in 12-well transwell inserts and treated with 5 levels of each butyrate derivative after adherence (n = 5). TEER was measured at 24, 48, and 72 h post-treatment to quantify intestinal barrier integrity of IPEC-J2 monolayers. Butyric acid, sodium butyrate, and monobutyrin significantly increased (p < 0.05) TEER in IPEC-J2 at different time points compared with control. Further, porcine alveolar macrophages (PAMs) were harvested from donor weaned piglets (n = 6) via bronchoalveolar lavage and isolated for primary culture (6 × 105 cells/well, 6-well plates). PAMs were treated with five levels of each butyrate derivative with or without lipopolysaccharide (LPS, 1 μg/mL) challenge. The concentrations of TNF-α and IL-1β in cell culture supernatants were measured by enzyme-linked immunosorbent assay (ELISA). Butyric acid and sodium butyrate treatments reduced the production of TNF-α in LPS-challenged PAMs (linear; p < 0.05). Different butyrate derivatives exerted anti-inflammatory properties and improved intestinal barrier integrity.
Collapse
Affiliation(s)
- Lauren Kovanda
- Department of Animal Science, University of California, Davis, CA 95616, USA; (L.K.); (T.D.)
| | - Monika Hejna
- Department of Biotechnology and Nutrigenomics, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Postępu 36A, 05-552 Jastrzębiec, Poland;
| | - Tina Du
- Department of Animal Science, University of California, Davis, CA 95616, USA; (L.K.); (T.D.)
- Department of Nutrition, University of California, Davis, CA 95616, USA
| | - Yanhong Liu
- Department of Animal Science, University of California, Davis, CA 95616, USA; (L.K.); (T.D.)
| |
Collapse
|
16
|
Wei FH, Xie WY, Zhao PS, Ji ZH, Gao F, Chen CZ, Zhang Z, Gao W, Yuan B. Crataegus pinnatifida polysaccharide alleviates DSS-induced colitis in mice by regulating the intestinal microbiota and enhancing arginine biosynthesis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156794. [PMID: 40315641 DOI: 10.1016/j.phymed.2025.156794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/08/2025] [Accepted: 04/20/2025] [Indexed: 05/04/2025]
Abstract
BACKGROUND The development of effective and safe dietary supplements is essential for both the prevention and management of ulcerative colitis (UC), as its pathogenesis is intricate and difficult to completely resolve. Crataegus pinnatifida, a medicinal food with a long history of use, has broad medicinal value. Recent research has revealed promising insights into the role of polysaccharide derived from Crataegus pinnatifida on modulating short-chain fatty acids (SCFAs) to alleviate UC inflammation. However, the mechanisms by which CPP regulates the intestinal microbiota and key metabolites during the antagonistic phase of UC have yet to be elucidated. OBJECTIVE This research elucidated the protective role of CPP in relation to UC, highlighted the mechanisms through which CPP operates, particularly regarding gut microbiota and metabolism, and offered a theoretical foundation for the potential use of CPP as a dietary supplement aimed at preventing UC. METHODS The impact of CPP on acute UC induced by 3 % DSS in mice was examined through the evaluation of the disease activity index, measurement of colon length, and observation of body weight changes. Enzyme-linked immunosorbent assay (ELISA) was used to measure inflammatory factor levels in both serum and colon, as well as to assess oxidative stress mediators. The intestinal histological damage, mucus layer damage and the level of tight junction protein were analyzed by histopathological staining and western blot (WB). The impact of gut microbiota on CPP in colitis was evaluated using 16S rRNA sequencing, microbiota depletion experiments, and fecal microbiota transplantation (FMT) studies. The key metabolic pathways and key metabolites affected by CPP in the treatment of UC were analyzed through untargeted metabolomics sequencing, ELISA, and WB assays. RESULTS Prophylactic dietary supplementation with Crataegus pinnatifida polysaccharide (CPP) notably reduced the fundamental clinical manifestations of UC induced by DSS, including DAI score, reduced colon length, and weight loss, as well as inflammation and oxidative stress. CPP promoted the expression of Claudin-1, ZO-1 and Occludin and promoted mucin secretion, which contributed to the mitigation of intestinal barrier damage caused by DSS. 16S sequencing results and metabolomics results revealed that CPP intervention upregulated the relative abundance of Lactobacillus, thereby reshaping the intestinal microbiota and activate the arginine biosynthesis pathway. The results of fecal microbiota transplantation and antibiotic clearance experiments indicated that the alleviating effect of CPP on UC was dependent on the intestinal microbiota and this alleviating effect was transferred through fecal microbiota transplantation. Mechanistically, CPP indirectly promoted the expression of the rate-limiting enzyme argininosuccinate synthase 1 (ASS1) in the intestinal Arginine biosynthesis pathway by reshaping the intestinal microbiota, thereby increasing intestinal Arginine level and alleviating the inflammatory response and oxidative stress induced by DSS and intestinal barrier damage. CONCLUSION Our research findings demonstrate that CPP is a plant-derived polysaccharide that alleviates UC by modulating the gut microbiota and enhancing arginine biosynthesis.
Collapse
Affiliation(s)
- Fan-Hao Wei
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China.
| | - Wen-Yin Xie
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China.
| | - Pei-Sen Zhao
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China.
| | - Zhong-Hao Ji
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China.
| | - Fei Gao
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China.
| | - Cheng-Zhen Chen
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China.
| | - Zhe Zhang
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China.
| | - Wei Gao
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China.
| | - Bao Yuan
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China.
| |
Collapse
|
17
|
Zhang S, Cao Y, Huang Y, Zhang X, Mou C, Qin T, Chen Z, Bao W. Abortive PDCoV infection triggers Wnt/β-catenin pathway activation, enhancing intestinal stem cell self-renewal and promoting chicken resistance. J Virol 2025; 99:e0013725. [PMID: 40135895 PMCID: PMC11998530 DOI: 10.1128/jvi.00137-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 02/28/2025] [Indexed: 03/27/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging coronavirus causing economic losses to swine industries worldwide. PDCoV can infect chickens under laboratory conditions, usually with no symptoms or mild symptoms, and may cause outbreaks in backyard poultry and wildfowl, posing a potential risk of significant economic loss to the commercial poultry industry. However, the reasons for such a subdued reaction after infection are not known. Here, using chicken intestinal organoid monolayers, we found that although PDCoV infects them nearly as well as porcine intestinal organoid monolayers, infection did not result in detectable amounts of progeny virus. In ex vivo and in vivo experiments using chickens, PDCoV infection failed to initiate interferon and inflammatory responses. Additionally, infection did not result in a disrupted intestinal barrier nor a reduced number of goblet cells and mucus secretion, as in pigs. In fact, the number of goblet cells increased as did the secreted mucus, thereby providing an enhanced protective barrier. Ex vivo PDCoV infection in chicken triggered activation of the Wnt/β-catenin pathway with the upregulation of Wnt/β-catenin pathway genes (Wnt3a, Lrp5, β-catenin, and TCF4) and Wnt target genes (Lgr5, cyclin D1, and C-myc). This activation stimulates the self-renewal of intestinal stem cells (ISCs), accelerating ISC-mediated epithelial regeneration by significant up-regulation of PCNA (transiently amplifying cells), BMI1 (ISCs), and Lyz (Paneth cells). Our data demonstrate that abortive infection of PDCoV in chicken cells activates the Wnt/β-catenin pathway, which facilitates the self-renewal and proliferation of ISCs, contributing to chickens' resistance to PDCoV infection.IMPORTANCEThe intestinal epithelium is the main target of PDCoV infection and serves as a physical barrier against pathogens. Additionally, ISCs are charged with tissue repair after injury, and promoting rapid self-renewal of intestinal epithelium will help to re-establish the physical barrier and maintain intestinal health. We found that PDCoV infection in chicken intestinal organoid monolayers resulted in abortive infection and failed to produce infectious virions, disrupt the intestinal barrier, reduce the number of goblet cells and mucus secretion, and induce innate immunity, but rather increased goblet cell numbers and mucus secretion. Abortive PDCoV infection activated the Wnt/β-catenin pathway, enhancing ISC renewal and accelerating the renewal and replenishment of shed PDCoV-infected intestinal epithelial cells, thereby enhancing chicken resistance to PDCoV infection. This study provides novel insights into the mechanisms underlying the mild or asymptomatic response to PDCoV infection in chickens, which is critical for understanding the virus's potential risks to the poultry industry.
Collapse
Affiliation(s)
- Shuai Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yanan Cao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yanjie Huang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Xueli Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Chunxiao Mou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Tao Qin
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhenhai Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Wenbin Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| |
Collapse
|
18
|
Ma S, Duan Y, Yu Y, Hu Q, Tao Q, Li X, Kimatu BM, Ma G. Effects and Mechanisms of Pleurotus eryngii Polysaccharide on Intestinal Barrier Damage: Based on the Perspective of Its Interaction with Intestinal Mucus during Gut Digestion. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:7755-7773. [PMID: 40109172 DOI: 10.1021/acs.jafc.4c11339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
In this study, pathways and mechanisms of action of a new type of Pleurotus eryngii polysaccharide (PEP) with known structural characteristics and probiotic properties in the intestine were investigated. An in vitro cell model was used to investigate the protective effects of complexes formed between PEPs and their related products with mucin against gut barrier damage. Dextran sulfate sodium salt-induced colitis was used to determine the characteristics of the interaction between PEPs and intestinal mucus (IMs) at different consumption times. Finally, the protective effect of PEPs against intestinal barrier damage was investigated, as mediated by IMs. The result showed that complexes of PEP-related products and mucin improved damage to the intestinal barrier. PEPs exhibited differential functional activities at different stages. In normal and colitis mice, the interactions between IMs and PEPs showed different characteristics. From the transport and absorption standpoint, the role of PEPs in driving intestinal health was also clarified in this study.
Collapse
Affiliation(s)
- Sai Ma
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Yaning Duan
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Yunyan Yu
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Qiuhui Hu
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Qi Tao
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Xinyi Li
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Benard Muinde Kimatu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
- Department of Dairy and Food Science and Technology, Egerton University, P.O. Box, 536-20115, Egerton, Kenya
| | - Gaoxing Ma
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| |
Collapse
|
19
|
Kaden T, Alonso‐Román R, Stallhofer J, Gresnigt MS, Hube B, Mosig AS. Leveraging Organ-on-Chip Models to Investigate Host-Microbiota Dynamics and Targeted Therapies for Inflammatory Bowel Disease. Adv Healthc Mater 2025; 14:e2402756. [PMID: 39491534 PMCID: PMC12004439 DOI: 10.1002/adhm.202402756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/29/2024] [Indexed: 11/05/2024]
Abstract
Inflammatory bowel disease (IBD) is an idiopathic gastrointestinal disease with drastically increasing incidence rates. Due to its multifactorial etiology, a precise investigation of the pathogenesis is extremely difficult. Although reductionist cell culture models and more complex disease models in animals have clarified the understanding of individual disease mechanisms and contributing factors of IBD in the past, it remains challenging to bridge research and clinical practice. Conventional 2D cell culture models cannot replicate complex host-microbiota interactions and stable long-term microbial culture. Further, extrapolating data from animal models to patients remains challenging due to genetic and environmental diversity leading to differences in immune responses. Human intestine organ-on-chip (OoC) models have emerged as an alternative in vitro model approach to investigate IBD. OoC models not only recapitulate the human intestinal microenvironment more accurately than 2D cultures yet may also be advantageous for the identification of important disease-driving factors and pharmacological interventions targets due to the possibility of emulating different complexities. The predispositions and biological hallmarks of IBD focusing on host-microbiota interactions at the intestinal mucosal barrier are elucidated here. Additionally, the potential of OoCs to explore microbiota-related therapies and personalized medicine for IBD treatment is discussed.
Collapse
Affiliation(s)
- Tim Kaden
- Dynamic42 GmbH07745JenaGermany
- Institute of Biochemistry IICenter for Sepsis Control and CareJena University Hospital07747JenaGermany
| | - Raquel Alonso‐Román
- Department of Microbial Pathogenicity MechanismsLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Junior Research Group Adaptive Pathogenicity StrategiesLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
| | | | - Mark S. Gresnigt
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Junior Research Group Adaptive Pathogenicity StrategiesLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
| | - Bernhard Hube
- Department of Microbial Pathogenicity MechanismsLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Institute of MicrobiologyFaculty of Biological SciencesFriedrich Schiller University07743JenaGermany
| | - Alexander S. Mosig
- Institute of Biochemistry IICenter for Sepsis Control and CareJena University Hospital07747JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
| |
Collapse
|
20
|
Wu T, Song F, Huang J, Cui S, Wang L, Yang Q, Wu Y, Li B, Tu Y, Wan X, Liu J. Gut microbiota: The pivotal conduit in the onset of constipation and its alleviation by tea flower polysaccharides (TFP) in a mouse model. Int J Biol Macromol 2025; 304:140808. [PMID: 39924042 DOI: 10.1016/j.ijbiomac.2025.140808] [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: 12/10/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
Plant-derived bioactive components, such as polysaccharides, provide promising alleviating effects on constipation with minimal side-effects compared to pharmacological interventions. This study aimed to explore the therapeutic potential of tea flower polysaccharides (TFP) on constipation and the involved mechanisms. In a loperamide-induced constipation mouse model, TFP administration significantly increased fecal water content from 54.23-57.30 % to 63.70-79.36 %, enhanced intestinal transit rate from 30.80 % to 38.81 %, and reduced gastrointestinal (GI) transit time from 234.4 min to 186.2 min. TFP restored levels of both excitatory and inhibitory hormones related to GI motility. Transcriptomic analysis of colonic epithelial cells revealed that TFP restored expression of 544 genes involved in various pathways, including the NF-κB and JAK-STAT signaling pathways, which are associated with the improvement of constipation. Gut microbiota analysis demonstrated that TFP mitigated dysbiosis by normalizing the Firmicutes/Bacteroidota ratio, inhibiting pathogenic genera (e.g., Helicobacter), and promoting beneficial genera (e.g., Muribaculaceae, Bacteroides, Parabacteroides). The mediating role of gut microbiota in the onset of constipation and its alleviation was confirmed through fecal microbiota transplantation (FMT). Furthermore, TFP and its combination with anti-constipation drugs alleviated constipation-induced hepatorenal damage. This study highlights TFP's potential in treating constipation and underscores the essential role of gut microbiota in its therapeutic effects.
Collapse
Affiliation(s)
- Tingbo Wu
- Department of Tea Science, Zhejiang University, Hangzhou 310058, Zhejiang, PR China
| | - Fanfen Song
- Research Unit VEG-i-TEC, Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Campus Kortrijk, St-Martems Latemlaan 2B, Kortrijk 8500, Belgium
| | - Jiahong Huang
- Department of Tea Science, Zhejiang University, Hangzhou 310058, Zhejiang, PR China
| | - Shumao Cui
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, PR China
| | - Linlin Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu, PR China
| | - Qin Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, PR China
| | - Yuanyuan Wu
- Department of Tea Science, Zhejiang University, Hangzhou 310058, Zhejiang, PR China
| | - Bo Li
- Department of Tea Science, Zhejiang University, Hangzhou 310058, Zhejiang, PR China
| | - Youying Tu
- Department of Tea Science, Zhejiang University, Hangzhou 310058, Zhejiang, PR China
| | - Xiaochun Wan
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, Hefei 230036, Anhui, PR China
| | - Junsheng Liu
- Department of Tea Science, Zhejiang University, Hangzhou 310058, Zhejiang, PR China.
| |
Collapse
|
21
|
Shen C, Zhang S, Di H, Wang S, Wang Y, Guan F. The Role of Triterpenoids in Gastric Ulcer: Mechanisms and Therapeutic Potentials. Int J Mol Sci 2025; 26:3237. [PMID: 40244034 PMCID: PMC11990034 DOI: 10.3390/ijms26073237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/25/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Gastric ulcer (GU) is a prevalent gastrointestinal disorder impacting millions worldwide, with complex pathogenic mechanisms that may progress to severe illnesses. Conventional therapies relying on anti-secretory agents and antibiotics are constrained by drug abuse and increased bacterial resistance, highlighting the urgent need for safer therapeutic alternatives. Natural medicinal compounds, particularly triterpenoids derived from plants and herbs, have gained significant attention in recent years due to their favorable efficacy and reduced toxicity profiles. Emerging evidence indicates that triterpenoids exhibit potent anti-ulcer properties across various experimental models, modulating key pathways involved in inflammation, oxidative stress, apoptosis, and mucosal protection. Integrating current knowledge of these bioactive compounds facilitates the development of natural triterpenoids, addresses challenges in their clinical translation, deepens mechanistic understanding of GU pathogenesis, and drives innovation of therapeutic strategies for GU. This review comprehensively evaluates the progress of research on triterpenoids in GU treatment since 2000, discussing their biological sources, structural characteristics, pharmacological activities, and mechanisms of action, the animal models employed in the studies, current limitations, and the challenges associated with their clinical application.
Collapse
Affiliation(s)
- Congcong Shen
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (C.S.); (S.Z.); (H.D.); (S.W.)
| | - Shengyu Zhang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (C.S.); (S.Z.); (H.D.); (S.W.)
| | - Han Di
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (C.S.); (S.Z.); (H.D.); (S.W.)
| | - Shuang Wang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (C.S.); (S.Z.); (H.D.); (S.W.)
| | - Yanhong Wang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (C.S.); (S.Z.); (H.D.); (S.W.)
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Feng Guan
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (C.S.); (S.Z.); (H.D.); (S.W.)
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| |
Collapse
|
22
|
Yakut A. Gut microbiota in the development and progression of chronic liver diseases: Gut microbiota-liver axis. World J Hepatol 2025; 17:104167. [PMID: 40177197 PMCID: PMC11959663 DOI: 10.4254/wjh.v17.i3.104167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/28/2025] [Accepted: 02/25/2025] [Indexed: 03/26/2025] Open
Abstract
The gut microbiota (GM) is a highly dynamic ecology whose density and composition can be influenced by a wide range of internal and external factors. Thus, "How do GM, which can have commensal, pathological, and mutualistic relationships with us, affect human health?" has become the most popular research issue in recent years. Numerous studies have demonstrated that the trillions of microorganisms that inhabit the human body can alter host physiology in a variety of systems, such as metabolism, immunology, cardiovascular health, and neurons. The GM may have a role in the development of a number of clinical disorders by producing bioactive peptides, including neurotransmitters, short-chain fatty acids, branched-chain amino acids, intestinal hormones, and secondary bile acid conversion. These bioactive peptides enter the portal circulatory system through the gut-liver axis and play a role in the development of chronic liver diseases, cirrhosis, and hepatic encephalopathy. This procedure is still unclear and quite complex. In this study, we aim to discuss the contribution of GM to the development of liver diseases, its effects on the progression of existing chronic liver disease, and to address the basic mechanisms of the intestinal microbiota-liver axis in the light of recent publications that may inspire the future.
Collapse
Affiliation(s)
- Aysun Yakut
- Department of Gastroenterology, İstanbul Medipol University Sefakoy Health Practice Research Center, İstanbul 38000, Türkiye.
| |
Collapse
|
23
|
Roy A, Meregini S, Cho HJ, Chen Z, Zaki A, Argula T, Beutler B, SoRelle JA. N-glycosylation enzyme Mpi is essential for mucin O-glycosylation, host-microbe homeostasis, Paneth cell defense, and metabolism. RESEARCH SQUARE 2025:rs.3.rs-6222474. [PMID: 40195978 PMCID: PMC11975007 DOI: 10.21203/rs.3.rs-6222474/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Intestinal homeostasis relies on a protective mucus layer that separates bacteria from the host, with Muc2 as its primary component. This secreted, gel-forming mucin is heavily O-glycosylated, allowing it to retain water and support beneficial bacteria. For the first time, we demonstrate that Muc2 N-glycosylation plays a critical in mucin maturation, O-glycosylation, barrier integrity, and the prevention of dysbiosis. Using mouse models with global and intestine-specific N-glycan deficiency- caused by the loss of the mannose producing enzyme, Mpi- we uncover an unexpected link between N-glycosylation and intestinal homeostasis. Our findings reveal that Mpi hypomorphic mice are highly sensitive to DSS-induced colitis, while Mpi flox; Villin Cre mice spontaneously develop disease, exhibiting increased ER stress and dysbiosis. Additionally, electron microscopy, proteomics, and gene expression analyses of goblet and Paneth cells indicate immaturity, mitochondrial loss, and disruptions in lipid metabolism. These results highlight the fundamental role of N-glycosylation in maintaining intestinal homeostasis.
Collapse
Affiliation(s)
- Avishek Roy
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Steve Meregini
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Hye-Jeong Cho
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Zhenglan Chen
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Aariz Zaki
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Tandav Argula
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Bruce Beutler
- Center for Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX
| | - Jeffrey A SoRelle
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
- Center for Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX
- Department of Pediatrics, Division of Allergy and Immunology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
24
|
Quintieri L, Luparelli A, Caputo L, Schirinzi W, De Bellis F, Smiriglia L, Monaci L. Unraveling the Biological Properties of Whey Peptides and Their Role as Emerging Therapeutics in Immune Tolerance. Nutrients 2025; 17:938. [PMID: 40289962 PMCID: PMC11946102 DOI: 10.3390/nu17060938] [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: 02/02/2025] [Revised: 02/20/2025] [Accepted: 03/04/2025] [Indexed: 04/30/2025] Open
Abstract
Whey is a natural by-product of the cheese-making process and represents a valuable source of nutrients, including vitamins, all essential amino acids and proteins with high quality and digestibility characteristics. Thanks to its different techno-functional characteristics, such as solubility, emulsification, gelling and foaming, it has been widely exploited in food manufacturing. Also, advances in processing technologies have enabled the industrial production of a variety of whey-based products exerting biological activities. The beneficial properties of whey proteins (WPs) include their documented effects on cardiovascular, digestive, endocrine, immune and nervous systems, and their putative role in the prevention and treatment of non-communicable diseases (NCDs). In this regard, research on their application for health enhancement, based on the optimization of product formulation and the development of pharmaceuticals, is highly relevant. Beyond the health and nutritionally relevant effects as in in vivo animal studies, the allergenicity of WPs and WP hydrolysates is also herein tackled and discussed, as well as their potential role as therapeutics for immune tolerance and so-called tolerogenic effects. Grounded on the WPs' health-promoting functions, this paper presents the latest research showing the potential of whey-derived peptides as an alternative strategy in NCD treatment. This work also reports a careful analysis of their current use, also revealing which obstacles limit their full exploitation, thus highlighting the future challenges in the field. Concluding, safety considerations, encompassing WP allergenicity, are also discussed, providing some insights on the role of WPs and peptides in milk allergen immunotolerance.
Collapse
Affiliation(s)
- Laura Quintieri
- Institute of Sciences of Food Production, National Research Council (ISPA-CNR), Via G. Amendola, 122/O, 70126 Bari, Italy; (L.Q.); (A.L.); (L.C.); (W.S.); (F.D.B.)
| | - Anna Luparelli
- Institute of Sciences of Food Production, National Research Council (ISPA-CNR), Via G. Amendola, 122/O, 70126 Bari, Italy; (L.Q.); (A.L.); (L.C.); (W.S.); (F.D.B.)
| | - Leonardo Caputo
- Institute of Sciences of Food Production, National Research Council (ISPA-CNR), Via G. Amendola, 122/O, 70126 Bari, Italy; (L.Q.); (A.L.); (L.C.); (W.S.); (F.D.B.)
| | - William Schirinzi
- Institute of Sciences of Food Production, National Research Council (ISPA-CNR), Via G. Amendola, 122/O, 70126 Bari, Italy; (L.Q.); (A.L.); (L.C.); (W.S.); (F.D.B.)
| | - Federica De Bellis
- Institute of Sciences of Food Production, National Research Council (ISPA-CNR), Via G. Amendola, 122/O, 70126 Bari, Italy; (L.Q.); (A.L.); (L.C.); (W.S.); (F.D.B.)
| | - Leonardo Smiriglia
- Parafarmacia Smiriglia Leonardo, Via San Giorgio 19/B, 70019 Bari, Italy;
| | - Linda Monaci
- Institute of Sciences of Food Production, National Research Council (ISPA-CNR), Via G. Amendola, 122/O, 70126 Bari, Italy; (L.Q.); (A.L.); (L.C.); (W.S.); (F.D.B.)
| |
Collapse
|
25
|
El-Baz AM, El-Mahmoudy AA, Saber S, ElRakaiby MT. The coadministration of Lactobacillus probiotic augments the antitumor effect of telmisartan in rats. AMB Express 2025; 15:38. [PMID: 40044961 PMCID: PMC11883082 DOI: 10.1186/s13568-025-01843-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 02/07/2025] [Indexed: 03/09/2025] Open
Abstract
Colorectal cancer (CRC) is a prevalent disease with a high mortality rate and is significantly affected by microbial dysbiosis. Recent research suggests that modulation of the gut microbiome can have therapeutic benefits and that Angiotensin-II Type 1 Receptor (AT1R) can stimulate cell growth, angiogenesis, and resistance to apoptosis in various cancers. In this study, the adjunctive administration of Lactobacillus spp. and telmisartan, an AT1R blocker, was explored in the treatment of CRC. The effect of telmisartan and a mixture of probiotic species, Lactobacillus delbrueckii and Lactobacillus fermentum, was assessed on key biomarkers and selected gut microbiota taxa in 1,2-dimethylhydrazine-induced CRC in rats. Angiogenesis, inflammation, and apoptosis were assessed by measuring vascular endothelial growth factor (VEGF), carcinoembryonic antigen (CEA), Interleukin 6 (IL-6), and Annexin V levels, respectively. The relative abundance of selected gut microbial taxa, including Bacteroides spp., Clostridium spp., Clostridium coccoides, Ruminococcus spp., and Lactobacillus spp. was analyzed to determine the change in the microbial composition in the different experimental groups of the animal model. This study demonstrated that the unique combination therapy using a Lactobacillus mixture and telmisartan effectively reduced VEGF and IL-6 levels, indicating decreased angiogenesis and inflammation. Lactobacillus spp. co-administration with telmisartan boosted programmed cell death, reversed dysbiosis, improved histopathological outcomes, and reduced CEA levels. These findings offer a new perspective on the role of Lactobacillus spp. and telmisartan in CRC treatment. Further research on their adjunctive use and therapeutic potential are needed to enhance clinical efficacy.
Collapse
Affiliation(s)
- Ahmed M El-Baz
- Department of Microbiology and Immunology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt.
| | - Amany A El-Mahmoudy
- Dakahliya Health Directorate, Ministry of Health and Population, Dakahliya, 35931, Egypt.
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt
| | - Marwa T ElRakaiby
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
26
|
El-Baz AM, El-Mahmoudy AA, Saber S, ElRakaiby MT. The coadministration of Lactobacillus probiotic augments the antitumor effect of telmisartan in rats. AMB Express 2025; 15:38. [DOI: https:/doi.org/10.1186/s13568-025-01843-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 02/07/2025] [Indexed: 05/14/2025] Open
Abstract
Abstract
Colorectal cancer (CRC) is a prevalent disease with a high mortality rate and is significantly affected by microbial dysbiosis. Recent research suggests that modulation of the gut microbiome can have therapeutic benefits and that Angiotensin-II Type 1 Receptor (AT1R) can stimulate cell growth, angiogenesis, and resistance to apoptosis in various cancers. In this study, the adjunctive administration of Lactobacillus spp. and telmisartan, an AT1R blocker, was explored in the treatment of CRC. The effect of telmisartan and a mixture of probiotic species, Lactobacillus delbrueckii and Lactobacillus fermentum, was assessed on key biomarkers and selected gut microbiota taxa in 1,2-dimethylhydrazine-induced CRC in rats. Angiogenesis, inflammation, and apoptosis were assessed by measuring vascular endothelial growth factor (VEGF), carcinoembryonic antigen (CEA), Interleukin 6 (IL-6), and Annexin V levels, respectively. The relative abundance of selected gut microbial taxa, including Bacteroides spp., Clostridium spp., Clostridium coccoides, Ruminococcus spp., and Lactobacillus spp. was analyzed to determine the change in the microbial composition in the different experimental groups of the animal model. This study demonstrated that the unique combination therapy using a Lactobacillus mixture and telmisartan effectively reduced VEGF and IL-6 levels, indicating decreased angiogenesis and inflammation. Lactobacillus spp. co-administration with telmisartan boosted programmed cell death, reversed dysbiosis, improved histopathological outcomes, and reduced CEA levels. These findings offer a new perspective on the role of Lactobacillus spp. and telmisartan in CRC treatment. Further research on their adjunctive use and therapeutic potential are needed to enhance clinical efficacy.
Collapse
|
27
|
Yang S, Liu H, Liu Y. Advances in intestinal epithelium and gut microbiota interaction. Front Microbiol 2025; 16:1499202. [PMID: 40104591 PMCID: PMC11914147 DOI: 10.3389/fmicb.2025.1499202] [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: 09/20/2024] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
The intestinal epithelium represents a critical interface between the host and external environment, serving as the second largest surface area in the human body after the lungs. This dynamic barrier is sustained by specialized epithelial cell types and their complex interactions with the gut microbiota. This review comprehensively examines the recent advances in understanding the bidirectional communication between intestinal epithelial cells and the microbiome. We briefly highlight the role of various intestinal epithelial cell types, such as Paneth cells, goblet cells, and enteroendocrine cells, in maintaining intestinal homeostasis and barrier function. Gut microbiota-derived metabolites, particularly short-chain fatty acids and bile acids, influence epithelial cell function and intestinal barrier integrity. Additionally, we highlight emerging evidence of the sophisticated cooperation between different epithelial cell types, with special emphasis on the interaction between tuft cells and Paneth cells in maintaining microbial balance. Understanding these complex interactions has important implications for developing targeted therapeutic strategies for various gastrointestinal disorders, including inflammatory bowel disease, metabolic disorders, and colorectal cancer.
Collapse
Affiliation(s)
- Sen Yang
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Pediatrics, The Fifth Peoples Hospital of Chengdu, Chengdu, China
| | - Hanmin Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yang Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
28
|
Xie C, Qi C, Zhang J, Wang W, Meng X, Aikepaer A, Lin Y, Su C, Liu Y, Feng X, Gao H. When short-chain fatty acids meet type 2 diabetes mellitus: Revealing mechanisms, envisioning therapies. Biochem Pharmacol 2025; 233:116791. [PMID: 39894305 DOI: 10.1016/j.bcp.2025.116791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/19/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Evidence is accumulating that short-chain fatty acids (SCFAs) produced by the gut microbiota play pivotal roles in host metabolism. They contribute to the metabolic regulation and energy homeostasis of the host not only by preserving intestinal health and serving as energy substrates but also by entering the systemic circulation as signaling molecules, affecting the gut-brain axis and neuroendocrine-immune network. This review critically summarizes the current knowledge regarding the effects of SCFAs in the fine-tuning of the pathogenesis of type 2 diabetes mellitus (T2DM) and insulin resistance, with an emphasis on the complex relationships among diet, microbiota-derived metabolites, T2DM inflammation, glucose metabolism, and the underlying mechanisms involved. We hold an optimistic view that elucidating how diet can influence gut bacterial composition and activity, SCFA production, and metabolic functions in the host will advance our understanding of the mutual interactions of the intestinal microbiota with other metabolically active organs, and may pave the way for harnessing these pathways to develop novel personalized therapeutics for glucometabolic disorders.
Collapse
Affiliation(s)
- Cong Xie
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China
| | - Cong Qi
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China
| | - Jianwen Zhang
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; School of Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617 China
| | - Wei Wang
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China
| | - Xing Meng
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; School of Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617 China
| | - Aifeila Aikepaer
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; Dongzhimen Hospital, the First Clinical Medical School of Beijing University of Chinese Medicine, Beijing 100700 China
| | - Yuhan Lin
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; Dongzhimen Hospital, the First Clinical Medical School of Beijing University of Chinese Medicine, Beijing 100700 China
| | - Chang Su
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730124 China
| | - Yunlu Liu
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700 China
| | - Xingzhong Feng
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China.
| | - Huijuan Gao
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China.
| |
Collapse
|
29
|
Xiao K, Park S, Stachowiak JC, Rangamani P. Biophysical modeling of membrane curvature generation and curvature sensing by the glycocalyx. Proc Natl Acad Sci U S A 2025; 122:e2418357122. [PMID: 39969997 PMCID: PMC11873937 DOI: 10.1073/pnas.2418357122] [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: 09/09/2024] [Accepted: 01/11/2025] [Indexed: 02/21/2025] Open
Abstract
Generation of membrane curvature is fundamental to cellular function. Recent studies have established that the glycocalyx, a sugar-rich polymer layer at the cell surface, can generate membrane curvature. While there have been some theoretical efforts to understand the interplay between the glycocalyx and membrane bending, there remain open questions about how the properties of the glycocalyx affect membrane bending. For example, the relationship between membrane curvature and the density of glycosylated proteins on its surface remains unclear. In this work, we use polymer brush theory to develop a detailed biophysical model of the energetic interactions of the glycocalyx with the membrane. Using this model, we identify the conditions under which the glycocalyx can both generate and sense curvature. Our model predicts that the extent of membrane curvature generated depends on the grafting density of the glycocalyx and the backbone length of the polymers constituting the glycocalyx. Furthermore, when coupled with the intrinsic membrane properties such as spontaneous curvature and a line tension along the membrane, the curvature generation properties of the glycocalyx are enhanced. These predictions were tested experimentally by examining the propensity of glycosylated transmembrane proteins to drive the assembly of highly curved filopodial protrusions at the plasma membrane of adherent mammalian cells. Our model also predicts that the glycocalyx has curvature-sensing capabilities, in agreement with the results of our experiments. Thus, our study develops a quantitative framework for mapping the properties of the glycocalyx to the curvature generation capability of the membrane.
Collapse
Affiliation(s)
- Ke Xiao
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA92093
| | - Sujeong Park
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX78712
| | - Jeanne C. Stachowiak
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX78712
- McKetta Department of Chemical Engineering, University of Texas at Austin, Austin, TX78712
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA92093
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA92093
| |
Collapse
|
30
|
Fu K, Cui J, Li Y, Zhang Y, Wang Y, Wu J, Chen X, Xue F, Ren J, Dai J, Tang F. Escherichia coli phage ΦPNJ-9 adheres to mucus via a variant Hoc protein. J Virol 2025; 99:e0178924. [PMID: 39723818 PMCID: PMC11853027 DOI: 10.1128/jvi.01789-24] [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: 10/13/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024] Open
Abstract
Phages, as antagonists of bacteria, hold significant promise for combating drug-resistant bacterial infections. Their host specificity allows phages to target pathogenic bacteria without disrupting the gut microbiota, offering distinct advantages in the prevention and control of intestinal pathogens. The interaction between the phage and the gut plays a crucial role in the efficacy of phage-mediated bacterial killing. However, the mechanisms underlying these interactions remain poorly understood. In this study, we demonstrate that the clinically isolated T4-like phage, ΦPNJ-9, effectively adheres to the intestinal mucosa in vivo. This adhesion is mediated by the phage's Hoc protein, which interacts with MUC2 in the mucus. The Hoc protein of ΦPNJ-9 represents a variant, consisting of only three domains and lacking Domain 3, in contrast to phage T4. The key interacting sites on ΦPNJ-9 Hoc are amino acids S183, L184, and T185 within Domain 2. Displaying Domain 2 of ΦPNJ-9 Hoc on the surface of M13 phage significantly enhances its adhesion to the intestinal mucosa. Additionally, we identify fucose residues in MUC2 as the critical binding sites for the phage. Through this adhesion, the phage occupies the intestinal niche, thereby protecting the mucosal layer from pathogenic Escherichia coli infections. Our findings highlight the role of Hoc proteins in phage adhesion to intestinal mucus and the variation in binding sites, providing key insights for phage-based strategies aimed at preventing and controlling intestinal pathogens.IMPORTANCEThe rise in antibiotic-resistant pathogenic bacteria has sparked renewed interest in phage therapy as a promising alternative, particularly for targeting intestinal pathogens due to phage's host specificity. However, clinical applications have revealed that many phages are ineffective in eliminating bacteria within the gut, primarily due to the complex interactions between the phage and the gut environment. However, the mechanisms underlying these interactions remain poorly understood. Our previous study demonstrated that a T4-like phage adheres to the intestinal mucosa through the interaction between its Hoc protein and MUC2 in the mucus. Whether this model is widespread among T4-like phages remains unknown. Here, we characterize a variant Hoc protein from a T4-like phage, and identify new binding sites within this protein. Our findings suggest that the interaction between Hoc and MUC2 is likely common, but the critical binding sites vary depending on the specific phage.
Collapse
Affiliation(s)
- Kailai Fu
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Jiaqi Cui
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Yao Li
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Yuhan Zhang
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Yang Wang
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Jiaoling Wu
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Xinru Chen
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Feng Xue
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Jianluan Ren
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Jianjun Dai
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Fang Tang
- Key Laboratory of Animal Bacteriology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
31
|
Kumar S, Mukherjee R, Gaur P, Leal É, Lyu X, Ahmad S, Puri P, Chang CM, Raj VS, Pandey RP. Unveiling roles of beneficial gut bacteria and optimal diets for health. Front Microbiol 2025; 16:1527755. [PMID: 40041870 PMCID: PMC11877911 DOI: 10.3389/fmicb.2025.1527755] [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: 11/18/2024] [Accepted: 02/03/2025] [Indexed: 04/02/2025] Open
Abstract
The gut microbiome plays a pivotal role in human health, influencing digestion, immunity, and disease prevention. Beneficial gut bacteria such as Akkermansia muciniphila, Adlercreutzia equolifaciens, and Christensenella minuta contribute to metabolic regulation and immune support through bioactive metabolites like short-chain fatty acids (SCFAs). Dietary patterns rich in prebiotics, fermented foods, and plant-based bioactive compounds, including polyphenols and flavonoids, promote microbiome diversity and stability. However, challenges such as individual variability, bioavailability, dietary adherence, and the dynamic nature of the gut microbiota remain significant. This review synthesizes current insights into gut bacteria's role in health, emphasizing the mechanisms by which dietary interventions modulate microbiota. Additionally, it highlights advancements in microbiome-targeted therapies and the transformative potential of personalized nutrition, leveraging microbiota profiling and artificial intelligence (AI) to develop tailored dietary strategies for optimizing gut health and mitigating chronic inflammatory disorders. Addressing these challenges requires a multidisciplinary approach that integrates scientific innovation, ethical frameworks, and practical implementation strategies.
Collapse
Affiliation(s)
- Suresh Kumar
- National Institute of Biologicals, Ministry of Health & Family Welfare, Govt. of India, Noida, India
| | - Riya Mukherjee
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan, Taiwan
| | - Pratibha Gaur
- Centre for Drug Design Discovery and Development (C4D), SRM University Delhi-NCR, Sonepat, India
- Department of Biotechnology and Microbiology, SRM University Delhi-NCR, Sonepat, India
| | - Élcio Leal
- Laboratório de Diversidade Viral, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, Brazil
| | - Xiaoming Lyu
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Saheem Ahmad
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Ha'il, Saudi Arabia
| | - Paridhi Puri
- University Centre for Research and Development, Chandigarh University, Mohali, India
| | - Chung-Ming Chang
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan, Taiwan
- Master & Ph.D Program in Biotechnology Industry, Chang Gung University, Taoyuan, Taiwan
| | - V. Samuel Raj
- Department of Biotechnology and Microbiology, SRM University Delhi-NCR, Sonepat, India
- Laboratório de Diversidade Viral, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, Brazil
| | - Ramendra Pati Pandey
- Department of Biotechnology and Microbiology, SRM University Delhi-NCR, Sonepat, India
- Laboratório de Diversidade Viral, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, Brazil
| |
Collapse
|
32
|
Rubio-Casillas A, Rodríguez-Quintero CM, Hromić-Jahjefendić A, Uversky VN, Redwan EM, Brogna C. The essential role of prebiotics in restoring gut health in long COVID. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:385-411. [PMID: 40246350 DOI: 10.1016/bs.pmbts.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The gut microbiota (GM) plays an essential role in human health, influencing not only digestive processes but also the immune system´s functionality. The COVID-19 pandemic has highlighted the complex interaction between viral infections and the GM. Emerging evidence has demonstrated that SARS-CoV-2 can disrupt microbial homeostasis, leading to dysbiosis and compromised immune responses. The severity of COVID-19 has been associated with a reduction in the abundance of several beneficial bacteria in the gut. It has been proposed that consuming probiotics may help to re-colonize the GM. Although probiotics are important, prebiotics are essential for their metabolism, growth, and re-colonization capabilities. This chapter delves into the critical role of prebiotics in restoring GM after COVID-19 disease. The mechanisms by which prebiotics enhance the metabolism of beneficial bacteria will be described, and how prebiotics mediate the re-colonization of the gut with beneficial bacteria, thereby restoring microbial diversity and promoting the resilience of the gut-associated immune system. The benefits of consuming prebiotics from natural sources are superior to those from chemically purified commercial products.
Collapse
Affiliation(s)
- Alberto Rubio-Casillas
- Autlan Regional Hospital, Jalisco Health Services, Autlan, Jalisco, Mexico; Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Jalisco, Mexico.
| | | | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina.
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States; Laboratory of New Methods in Biology, Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Russia.
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria, Egypt
| | - Carlo Brogna
- Craniomed Group Srl, Research Facility, Montemiletto (Av), Italy
| |
Collapse
|
33
|
Soltani N, Rahimi S, Khaki P, Karimi Torshizi MA, Eskandari B, Grimes J. Efficacy of hyperimmunized egg yolk antibodies (IgY) against Campylobacter jejuni: In Vitro and In Vivo evaluations. Poult Sci 2025; 104:104718. [PMID: 39787828 PMCID: PMC11761915 DOI: 10.1016/j.psj.2024.104718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025] Open
Abstract
Campylobacter infections are a prevalent cause of diarrheal disease in humans and are the most significant zoonotic pathogens worldwide. Human campylobacteriosis is generally via ingestion of contaminated poultry products. However, based on recent studies chicken egg yolk antibody (IgY) powder has great potential to reduce the cecum load of Campylobacter jejuni (C. jejuni) in broilers. To understand the effective and economically feasible dosage, two immunization and challenge studies were conducted using 30 layer hens and 250 broiler chickens and found a scientific approach, starting with in vitro evaluations and progressing with in vivo studies confirmed. In this study it was demonstrated that specific IgY powder (SIgY), produced by immunized hens via bacterin, was highly effective in inhibiting bacterial growth and adhesion, as well as exhibiting bactericidal and agglutination properties (P < 0.05). Notably, doses of 0.5 % and 1 % SIgY significantly enhanced both the height and width of intestinal villi, along with improving the villus height-to-crypt depth ratio when compared to the positive control group (P < 0.05). Furthermore, medium and high doses of SIgY were effective in preserving the integrity of the intestinal epithelium, as evidenced by a reduction in crypt depth and the number of goblet cells, which serve as important markers in the immune system (P < 0.01). Additionally, analyses of cecal and liver bacterial counts in response to the 0.5 % SIgY treatment revealed a significant reduction in C. jejuni counts compared to other challenged groups throughout the 28 d experiment (P < 0.01). Based on these results, it may be concluded that specific antibodies play a crucial role in maintaining the integrity of intestinal villi, support the health of the intestinal epithelium, and reduce the colonization of C. jejuni. These findings could form the basis for developing an economical and effective strategy to enhance poultry and human health in the context of C. jejuni infection.
Collapse
Affiliation(s)
- Nazanin Soltani
- Department of Poultry Science, Faculty of Agriculture, Tarbiat Modares University, Postal code: 1411713116, Tehran, Iran
| | - Shaban Rahimi
- Department of Poultry Science, Faculty of Agriculture, Tarbiat Modares University, Postal code: 1411713116, Tehran, Iran
| | - Pejvak Khaki
- Department of Microbiology, Razi Vaccine and Serum Production Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Postal Code: 3197619751, Karaj, Iran
| | - Mohammad Amir Karimi Torshizi
- Department of Poultry Science, Faculty of Agriculture, Tarbiat Modares University, Postal code: 1411713116, Tehran, Iran
| | - Bahareh Eskandari
- Department of Poultry Science, Faculty of Agriculture, Tarbiat Modares University, Postal code: 1411713116, Tehran, Iran
| | - Jesse Grimes
- Prestage Department of Poultry Science, College of Agriculture and Life Sciences, North Carolina State University, Raleigh, NC 27695-7608.
| |
Collapse
|
34
|
Zhang S, Zhou R, Xie X, Xiong S, Li L, Li Y. Polysaccharides from Lycium barbarum, yam, and sunflower ameliorate colitis in a structure and intrinsic flora-dependent manner. Carbohydr Polym 2025; 349:122905. [PMID: 39643421 DOI: 10.1016/j.carbpol.2024.122905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 12/09/2024]
Abstract
Polysaccharides have been suggested to ameliorate metabolic diseases. However, their differential colitis-mitigating effects in mouse models with different colony structures remain poorly understood. Therefore, this study investigated the effects of polysaccharides from Lycium barbarum (LBP), sunflower (SP), and yam (YP) on colitis in C57BL/6 J (B6) mice born via vaginal delivery (VD) and in both caesarean section (CS)- and VD-born Institute of Cancer Research (ICR) mice. LBP was mainly composed of glucose (30.2 %), galactose (27.5 %), and arabinose (26.9 %). The main components of SP and YP were galacturonic acid (75.8 %) and glucose (98.1 %), respectively. Interestingly, LBP effectively alleviated body weight loss, reduced inflammatory cytokine levels, and restored intestinal barrier function in all three mouse models. Moreover, LBP decreased the abundance of norank_f__norank_o__Clostridia_UCG-014, Coriobacteriaceae_UCG-002, and norank_f_Eubacterium_coprostanoligenes_group in B6 mice, and the abundance of these genera positively correlated with pro-inflammatory cytokine levels. LBP increased the abundance of Lactobacillus, which was positively correlated with the levels of the protective factor, IL-10, in CS-born ICR mice. Collectively, our study suggests the potential application of LBP in the treatment of ulcerative colitis. We also provide an alternative method for restoring intestinal homeostasis in CS-born offspring.
Collapse
Affiliation(s)
- Shanshan Zhang
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ruchen Zhou
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaoran Xie
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Shanshan Xiong
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Lixiang Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Shandong Provincial Clinical Research Center for Digestive Disease, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Yanqing Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Laboratory of Translational Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China; Shandong Provincial Clinical Research Center for Digestive Disease, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
35
|
Yang D, Li MM, Xu HX, Wang WJ, Yin ZP, Zhang QF. Retrograded starch as colonic delivery carrier of taxifolin for treatment of DSS-induced ulcerative colitis in mice. Int J Biol Macromol 2025; 288:138602. [PMID: 39672437 DOI: 10.1016/j.ijbiomac.2024.138602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/27/2024] [Accepted: 12/07/2024] [Indexed: 12/15/2024]
Abstract
Taxifolin, a natural dihydroflavonol compound, possesses notable anti-inflammatory properties and regulatory effects on intestinal microbiota. In this study, gelatinized-retrograded corn starch (GCS) was utilized as a carrier for colonic delivery of taxifolin, and its therapeutic efficacy against dextran sulfate sodium (DSS)-induced colitis in mice were systematically investigated. Taxifolin can integrate into the helical structure of starch, and the formation of GCS-Taxifolin complexes (GCS-Tax) significantly delayed the release of taxifolin in vitro. After oral administration of GCS-Tax, fecal excretion of taxifolin increased from 0.42 % to 10.89 % within 24 h compared to free taxifolin. Moreover, GCS-Tax facilitated the production of short-chain fatty acid in mice and effectively alleviated DSS-induced colitis symptoms, including weight loss, bloody stools, and colonic tissue damage. Additionally, GCS-Tax significantly suppressed proinflammatory factors such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and lipopolysaccharide (LPS), while elevating anti-inflammatory interleukin-10 (IL-10) level in mice serum. Furthermore, it restored intestinal mucosal barrier function by upregulating the expression of Mucin 2, Occludin, and zonula occludens-1 (ZO-1), reducing Beclin 1 expression, and exhibited hepatoprotective effects by enhancing total antioxidant capacity (T-AOC), catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) activities. High-throughput sequencing analysis revealed that GCS-Tax improved intestinal flora diversity, reducing inflammation-related Bacterium 1 and Staphylococcus, while promoting the abundance of beneficial bacteria like Lachnospiraceae.
Collapse
Affiliation(s)
- Dan Yang
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Mang-Mang Li
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Hai-Xia Xu
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Wen-Jun Wang
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Zhong-Ping Yin
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China
| | - Qing-Feng Zhang
- Jiangxi Key Laboratory of Natural Product and Functional Food, College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang 330045, China.
| |
Collapse
|
36
|
Jiang B, Peng C, Li X, Sun C, Lu W, Fang Y. Banana Starch Nanoparticles Disrupt the Integrity of the Intestinal Barrier by Opening Tight Junctions in Mice. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408298. [PMID: 39668449 DOI: 10.1002/smll.202408298] [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: 09/12/2024] [Revised: 12/04/2024] [Indexed: 12/14/2024]
Abstract
The banana-derived starch nanoparticles have been extensively used in food and biomedicine industries, due to their unique physicochemical properties and functional benefits. With their pervasive presence in food, people are significantly exposed to these nanoparticles, raising concerns about their potential health risks and impact on intestinal health. However, there is still limited understanding of the direct interaction between native banana starch nanoparticles (BSNs) and the intestinal systems. Here, it is demonstrated that BSNs can cause tight junctions to loosen, increase intestinal permeability, and disrupt the intestinal barrier. This increased permeability is closely linked to the size of BSNs, with smaller BSNs (d = 60 nm) having a stronger effect on permeation. Furthermore, the disruption of the intestinal barrier integrity caused by BSNs is connected to a reduced amount of tight junction proteins. Mechanistically, BSNs disrupt tight junctions by affecting mitochondrial function and activating myosin light chain kinase (MLCK) signaling pathway. These findings indicate that BSNs have the potential to pose health risks by compromising the integrity of the intestinal barrier, reminding the safety of food biopolymer nanoparticles in living organisms needs to be re-assessed.
Collapse
Affiliation(s)
- Bing Jiang
- Department of food science and engineering, School of agriculture and biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chenglu Peng
- Department of food science and engineering, School of agriculture and biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoyang Li
- Department of food science and engineering, School of agriculture and biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Cuixia Sun
- Department of food science and engineering, School of agriculture and biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wei Lu
- Department of food science and engineering, School of agriculture and biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yapeng Fang
- Department of food science and engineering, School of agriculture and biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
37
|
Zhu X, Cheng F, Duan H, Fu S, Zhao C. Novel insights into the study of goblet cell hypersecretion in allergic rhinitis. Front Immunol 2025; 16:1525928. [PMID: 39958344 PMCID: PMC11825788 DOI: 10.3389/fimmu.2025.1525928] [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: 11/10/2024] [Accepted: 01/06/2025] [Indexed: 02/18/2025] Open
Abstract
Goblet cell hypersecretion is a hallmark of airway inflammation and is driven by complex neuroimmune regulation involving submucosal glands and goblet cells. Although studies have focused on mast cell degranulation as a critical driver of nasal secretion, the role of goblet cells in this process is relatively under-researched. In allergic airway inflammation, goblet cells exhibit metaplasia and hypersecretion. However, allergen exposure does not directly trigger goblet cell degranulation, raising questions regarding the underlying mechanisms of these reactions. The activation of enteric neurons promotes goblet cell degranulation by stimulating the calcitonin gene-related peptide (CGRP)-receptor active modification protein-1 (RAMP1) axis. Meanwhile, airway goblet cells express various neuropeptide receptors, and their activation by neuropeptides such as substance P and CGRP induces mucus secretion, exacerbating allergic rhinitis-associated hypersecretion. Thus, although previously less recognised, the neuron-goblet cell signalling axis plays a critical role in allergic rhinitis mucus secretion. This review highlights current research on the neuroimmune mechanisms underlying goblet cell metaplasia and degranulation, focusing on allergic rhinitis, so as to guide clinical treatment strategies.
Collapse
Affiliation(s)
- Xiaojia Zhu
- Department of Otolaryngology–Head and Neck Surgery, The Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Medical University, Taiyuan, China
| | - Fengli Cheng
- Department of Otolaryngology–Head and Neck Surgery, The Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Hongying Duan
- Department of Otolaryngology–Head and Neck Surgery, The Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Medical University, Taiyuan, China
| | - Sirui Fu
- Department of Otolaryngology–Head and Neck Surgery, The Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Medical University, Taiyuan, China
| | - Changqing Zhao
- Department of Otolaryngology–Head and Neck Surgery, The Second Hospital, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
38
|
Zhu H, Xu G. Electrochemical biosensors for dopamine. Clin Chim Acta 2025; 566:120039. [PMID: 39550057 DOI: 10.1016/j.cca.2024.120039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/08/2024] [Accepted: 11/10/2024] [Indexed: 11/18/2024]
Abstract
Dopamine (DA), a key catecholamine, plays a pivotal role in the regulation of human cognition and emotions. It has profound effects on the hormonal, memory, and cardiovascular systems. Anomalies like Alzheimer's, Parkinson's, schizophrenia, and senile dementia are linked to abnormal DA levels. Consequently, the precise determination of DA levels in biological systems is critical for the accurate diagnosis and treatment of these disorders. Among all analytical techniques, electrochemical studies provide the most selective and highly sensitive methods for detecting DA in biological samples. Ascorbic acid and uric acid are two examples of small biomolecules that can obstruct the detection of DA in biological fluids. To address this issue, numerous attempts have been made to modify bare electrodes to separate the signals of these substances and enhance the electrocatalytic activity towards DA. Various surface modifiers, including coatings, conducting polymers, ionic liquids, nanomaterials, and inorganic complexes, have been employed in the modification process. Despite the reported success in DA detection using electrochemical sensors, many of these approaches are deemed too complex and costly for real-world applications. Therefore, this review aims to provide an overview of DA electrochemical biosensors that are practical for real-world applications.
Collapse
Affiliation(s)
- Hang Zhu
- School of Pharmacy and Medical Technology, Putian University, Putian, Fujian 351100, China; Key Laboratory of Pharmaceutical Analysis and Laboratory Medicine, Putian University, Putian, Fujian 351100, China.
| | - Guifen Xu
- School of Pharmacy and Medical Technology, Putian University, Putian, Fujian 351100, China
| |
Collapse
|
39
|
Chen L, Tang J, Xia Y, Wang J, Xia LN. Mechanistic study of the effect of a high-salt diet on the intestinal barrier. Sci Rep 2025; 15:3826. [PMID: 39885261 PMCID: PMC11782509 DOI: 10.1038/s41598-025-88291-y] [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: 07/01/2024] [Accepted: 01/28/2025] [Indexed: 02/01/2025] Open
Abstract
Despite the established link between chronic high salt diet (HSD) and an increase in gut inflammation, the effect of HSD on the integrity of the intestinal barrier remains understudied. The present study aims to investigate the impact of HSD on the intestinal barrier in rats, encompassing its mechanical, mucous, and immune components. Expression levels of intestinal tight junction proteins and mucin-2 (MUC2) in SD rats were analyzed using immunofluorescence. The expression area of goblet cell mucopolysaccharides was assessed through PAS staining. Additionally, serum D-lactic acid, SIgA, β-defensin, and colonic tissue cytokines were measured using ELISA. Rats fed with HSD exhibited decreased expression of tight junction proteins, particularly Occludin, resulting in impairment of the intestinal epithelial barrier and an elevated serum D-lactic acid level. Furthermore, a notable reduction in the expression of goblet cell mucopolysaccharides, along with lower β-defensin and MUC2 levels, was observed. Notably, the SIgA and immune-related cytokines were significantly reduced in the HSD group. HSD disrupts the intestinal barrier in rats, leading to increased permeability and the entry of inflammatory factors into the bloodstream. This finding suggests that HSD may contribute to the pathogenesis of various diseases.
Collapse
Affiliation(s)
- Li Chen
- School of Health Preservation and Rehabilitation, Chengdu University of TCM, Shierqiao Road, Chengdu, 610075, Sichuan, People's Republic of China
- Key Laboratory of Traditional Chinese Medicine Regimen and Health Industry Development, State Administration of TCM, Chengdu, 610075, Sichuan, People's Republic of China
| | - Junrui Tang
- School of Health Preservation and Rehabilitation, Chengdu University of TCM, Shierqiao Road, Chengdu, 610075, Sichuan, People's Republic of China
- Key Laboratory of Traditional Chinese Medicine Regimen and Health Industry Development, State Administration of TCM, Chengdu, 610075, Sichuan, People's Republic of China
| | - Yanglin Xia
- School of Health Preservation and Rehabilitation, Chengdu University of TCM, Shierqiao Road, Chengdu, 610075, Sichuan, People's Republic of China
- Key Laboratory of Traditional Chinese Medicine Regimen and Health Industry Development, State Administration of TCM, Chengdu, 610075, Sichuan, People's Republic of China
| | - Jie Wang
- School of Health Preservation and Rehabilitation, Chengdu University of TCM, Shierqiao Road, Chengdu, 610075, Sichuan, People's Republic of China
- Key Laboratory of Traditional Chinese Medicine Regimen and Health Industry Development, State Administration of TCM, Chengdu, 610075, Sichuan, People's Republic of China
| | - Li-Na Xia
- School of Health Preservation and Rehabilitation, Chengdu University of TCM, Shierqiao Road, Chengdu, 610075, Sichuan, People's Republic of China.
- Key Laboratory of Traditional Chinese Medicine Regimen and Health Industry Development, State Administration of TCM, Chengdu, 610075, Sichuan, People's Republic of China.
| |
Collapse
|
40
|
Huang MY, Smieska LM, Tako E. Intestinal Effects of Brewers' Spent Grain Extract In Ovo ( Gallus gallus)-A Pilot Study. Animals (Basel) 2025; 15:303. [PMID: 39943073 PMCID: PMC11816252 DOI: 10.3390/ani15030303] [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: 12/19/2024] [Revised: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Upcycling brewers' spent grain (BSG) into poultry feed needs to be optimized. Since broiler chickens inefficiently digest fiber, we created a water-soluble BSG extract (BSGE) to explore this fraction's potential nutritional benefits. We utilized intra-amniotic administration (in ovo) to target the gastrointestinal tract of broiler embryos. BSGE increased villus surface area and goblet cell quantity and size, implying improved duodenal development. The extract also changed cecal Escherichia coli (E. coli) and Clostridium abundances. Synchrotron X-ray fluorescence microscopy, along with zinc and iron transporter relative expression, did not reveal significant changes by BSGE. These findings highlight the potential for BSGE to be a functional feed component, underscoring the potential value of upcycling this byproduct. This pilot study supports future work exploring the impact of BSGE within feed and its effects over long-term consumption.
Collapse
Affiliation(s)
- Melissa Y. Huang
- Department of Food Science, Cornell University, Ithaca, NY 14853, USA;
| | - Louisa M. Smieska
- Cornell High Energy Synchrotron Source, Cornell University, Ithaca, NY 14853, USA;
| | - Elad Tako
- Department of Food Science, Cornell University, Ithaca, NY 14853, USA;
| |
Collapse
|
41
|
Zhao H, Zhang Z, Liu H, Ma M, Sun P, Zhao Y, Liu X. Multi-omics perspective: mechanisms of gastrointestinal injury repair. BURNS & TRAUMA 2025; 13:tkae057. [PMID: 39845194 PMCID: PMC11752642 DOI: 10.1093/burnst/tkae057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 01/24/2025]
Abstract
In this review, we examine the significance of multi-omics technologies in understanding the plethora of intricate processes that activate gastrointestinal (GI) injury repair. Multi-omics, which includes genomics, transcriptomics, proteomics, and metabolomics, allows intricate mapping of cellular responses and molecular pathways involved in GI repair. We highlight the potential of multi-omics to discover previously unknown therapeutic targets or elucidate the molecular basis of the pathogenesis of GI. Furthermore, we explore the possibilities of integrating omics data to improve prediction models, and summarize the state-of-the-art technological developments and persisting obstacles that hinder the translation of multi-omics into clinical practice. Finally, innovative multi-omics approaches that can improve patient outcomes and advance therapeutic strategies in GI medicine are discussed.
Collapse
Affiliation(s)
- Haibin Zhao
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Zhigang Zhang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Hongyu Liu
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Mingxiu Ma
- Department of General Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Peng Sun
- Department of General Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Yang Zhao
- Department of General Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| | - Xun Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
| |
Collapse
|
42
|
Silva-Veiga FM, Marinho TS, de Souza-Mello V, Aguila MB, Mandarim-de-Lacerda CA. Tirzepatide, a dual agonist of glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1), positively impacts the altered microbiota of obese, diabetic, ovariectomized mice. Life Sci 2025; 361:123310. [PMID: 39675551 DOI: 10.1016/j.lfs.2024.123310] [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: 09/28/2024] [Revised: 11/26/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
The study aimed to verify the effect of Tirzepatide (Tzp, a dual agonist GIP/GLP-1) on intestinal health and microbiota balance in an obese diabetic ovariectomized (Ovx) mice model. Female C57BL/6 mice with Ovx and diet-induced obesity with diabetes were treated with Tzp (10 nmol/kg) for four weeks. Control (C) and obese-diabetic subgroups (Od) were formed (group abbreviations: O, Ovx; T, Tzp; n = 30/group): C, CT, CO, COT, Od, OdT, OdO, OdOT. The ileum was structurally and molecularly studied, and cecal feces had microbial DNA determined. Tzp improved the intestinal barrier structure and protection. Cldn12 (Claudin 12) increased, and Muc2 (Mucin 2) decreased. JamA (junctional adhesion molecules) and Ocln (Occludin) increased. Tzp mitigated macrophage activation and inflammation, altered composition, and the contribution to microbiota: Firmicutes decreased, and Bacteroidetes increased, changing the Firmicutes / Bacteroidetes ratio. Proteobacteria, Actinobacteria, Bifidobacterium, and Clostridium increased. In addition, Bacteroides, Prevotella, and Akkermansia increased. PCA indicated a significant action of Cd14, Muc2, and Tlr4 on CO and Il17 on OdO; Il10, Cd206, Cd12, Ocln, and JamA in Od. Bacteroides, Bifidobacterium, Clostridium, Actinobacteria, and Bacteroides were enhanced in CT and COT, Provotella, Proteobacteria, and Firmicutes in CO, Od, OdT, OdO, and Akkermansia in OdOT. In conclusion, the intestinal barrier function in our model is compromised by alterations in phylogenetic diversity and intestinal microbiota, which characterize dysbiosis and potentially enable the influx of toxins into other tissues. Treatment with Tzp demonstrated the ability to reverse intestinal dysbiosis, help repair intestinal barrier integrity, and mitigate possible endotoxemia through anti-inflammatory signaling pathways.
Collapse
Affiliation(s)
- Flavia Maria Silva-Veiga
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thatiany Souza Marinho
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vanessa de Souza-Mello
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Carlos Alberto Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
43
|
Vallejos OP, Bueno SM, Kalergis AM. Probiotics in inflammatory bowel disease: microbial modulation and therapeutic prospects. Trends Mol Med 2025:S1471-4914(24)00338-1. [PMID: 39814640 DOI: 10.1016/j.molmed.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/18/2025]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder that represents a significant public health challenge worldwide. This multifactorial condition results from complex interactions among genetic, environmental, immune, and microbial factors. Some beneficial microbes, known as probiotics, have been identified as promising therapeutic agents for inflammatory conditions, such as IBD. In this review, we explore the potential of probiotics as a therapeutic strategy for managing IBD. Probiotics have shown promise due to their ability to modulate the gut microbiota, regulate histamine levels, and enhance vitamin D metabolism, thereby promoting a tolerant immune profile and reducing inflammation. While the exact mechanisms underlying these benefits remain incompletely understood, probiotics represent a novel and emerging approach for alleviating the exacerbated inflammation characteristic of this disorder.
Collapse
Affiliation(s)
- Omar P Vallejos
- Millennium Institute of Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute of Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Alexis M Kalergis
- Millennium Institute of Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
44
|
Xu J, Abe K, Kodama T, Sultana M, Chac D, Markiewicz SM, Matsunami H, Kuba E, Tsunoda S, Alam M, Weil AA, Nakamura S, Yamashiro T. The role of morphological adaptability in Vibrio cholerae's motility. mBio 2025; 16:e0246924. [PMID: 39611848 PMCID: PMC11708025 DOI: 10.1128/mbio.02469-24] [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: 08/13/2024] [Accepted: 11/12/2024] [Indexed: 11/30/2024] Open
Abstract
Vibrio cholerae, the causative agent of cholera, displays remarkable adaptability to diverse environmental conditions through morphological changes that enhance its pathogenicity and influence the global epidemiology of the disease. This study examines the motility differences between filamentous and comma-shaped forms of the V. cholerae O1 strain under various viscosity conditions. Utilizing the El Tor strain, we induced filamentous transformation and conducted a comparative analysis with the canonical comma-shaped morphology. Our methodology involved assessing motility patterns, swimming speeds, rotation rates, kinematics, and reversal frequencies using dark-field microscopy and high-speed imaging techniques. The results show that filamentous V. cholerae cells retain enhanced motility in viscous environments, indicating an evolutionary adaptation for survival in varied habitats, particularly the human gastrointestinal tract. Filamentous forms exhibited increased reversal behavior at mucin interfaces, suggesting an advantage in penetrating the mucus layer. Furthermore, the presence of filamentous cells in bile-supplemented medium underscores their relevance in natural infection scenarios. IMPORTANCE This study highlights the enhanced motility of filamentous Vibrio cholerae in viscous environments, an adaptation that may provide a survival advantage in the human gastrointestinal tract. By demonstrating increased reversal behavior at mucin interfaces, filamentous V. cholerae cells exhibit a superior ability to penetrate the mucus layer, which is crucial for effective colonization and infection. Filamentous cells in bile-supplemented media further underscores their potential role in disease pathogenesis. These findings offer critical insights into the morphological flexibility of V. cholerae and its potential implications for infection dynamics, paving the way for more effective strategies in managing and preventing cholera outbreaks.
Collapse
Affiliation(s)
- Jun Xu
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa, Japan
| | - Keigo Abe
- Department of Applied Physics, Graduate School of Engineering, Tohoku University, Sendai, Miyagi, Japan
| | - Toshio Kodama
- Department of Bacteriology, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Marzia Sultana
- Infectious Diseases Division, International Center for Diarrheal Disease Research, Bangladesh, Bangladesh, Dhaka
| | - Denise Chac
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | - Hideyuki Matsunami
- Molecular Cryo-Electron Microscopy Unit, Okinawa Institute of Science and Technology, Okinawa, Japan
| | - Erika Kuba
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa, Japan
| | - Shiyu Tsunoda
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa, Japan
| | - Munirul Alam
- Infectious Diseases Division, International Center for Diarrheal Disease Research, Bangladesh, Bangladesh, Dhaka
| | - Ana A. Weil
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Shuichi Nakamura
- Department of Applied Physics, Graduate School of Engineering, Tohoku University, Sendai, Miyagi, Japan
| | - Tetsu Yamashiro
- Department of Bacteriology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa, Japan
| |
Collapse
|
45
|
Luo Y, He B, Li ZP, Zhong Q, Liu YC, Zhang HY, Li Y, Yan HL, Hu YL, Zheng ZJ, Ren H, Liao XP, Sun J. Rutin Synergizes with Colistin to Eradicate Salmonellosis in Mice by Enhancing the Efficacy and Reducing the Toxicity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:438-449. [PMID: 39699161 DOI: 10.1021/acs.jafc.4c06751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
The wide dissemination of multidrug-resistant (MDR) Gram-negative bacteria poses a significant global health and security concern. As developing new antibiotics is generally costly, fastidious, and time-consuming, there is an urgent need for alternative therapeutic strategies to address the gap in antibiotic discovery void. This study aimed to investigate the activity of colistin (CS) in combination with a natural product, rutin (RT), to combat against Salmonella Typhimurium (S. Tm) in vitro and in vivo. The results showed that a combination with RT enabled the potentiation of CS efficacy. Further mechanistic analysis indicated that RT disrupted iron homeostasis to inactivate the PmrA/PmrB system, thereafter reducing the bacterial membrane modifications for enhancing CS binding. Besides enhancing bactericidal activity of CS, RT was also observed to mitigate the CS-induced nephrotoxicity, by which the dosing limitation of CS was overcome for better pathogen clearance. The animal trial eventually confirmed the in vivo synergistic interaction of RT with CS to treat the bacterial infection. To sum up, the present study uncovered the potential of RT as a viable adjuvant of CS to eradicate the infection and protect the hosts, which might serve as a promising alternative to combat infections caused by MDR Gram-negative bacteria.
Collapse
Affiliation(s)
- Yang Luo
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, PR China
| | - Bing He
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, PR China
| | - Zhi-Peng Li
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, PR China
| | - Qin Zhong
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, PR China
| | - Yu-Chen Liu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, PR China
| | - Hai-Yi Zhang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, PR China
| | - Yan Li
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, PR China
| | - Hui-Lin Yan
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, PR China
| | - Ya-Lin Hu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, PR China
| | - Zi-Jian Zheng
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, PR China
| | - Hao Ren
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, PR China
| | - Xiao-Ping Liao
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, PR China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China
| | - Jian Sun
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics, Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, PR China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China
| |
Collapse
|
46
|
Termite F, Archilei S, D’Ambrosio F, Petrucci L, Viceconti N, Iaccarino R, Liguori A, Gasbarrini A, Miele L. Gut Microbiota at the Crossroad of Hepatic Oxidative Stress and MASLD. Antioxidants (Basel) 2025; 14:56. [PMID: 39857390 PMCID: PMC11759774 DOI: 10.3390/antiox14010056] [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: 12/13/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent chronic liver condition marked by excessive lipid accumulation in hepatic tissue. This disorder can lead to a range of pathological outcomes, including metabolic dysfunction-associated steatohepatitis (MASH) and cirrhosis. Despite extensive research, the molecular mechanisms driving MASLD initiation and progression remain incompletely understood. Oxidative stress and lipid peroxidation are pivotal in the "multiple parallel hit model", contributing to hepatic cell death and tissue damage. Gut microbiota plays a substantial role in modulating hepatic oxidative stress through multiple pathways: impairing the intestinal barrier, which results in bacterial translocation and chronic hepatic inflammation; modifying bile acid structure, which impacts signaling cascades involved in lipidic metabolism; influencing hepatocytes' ferroptosis, a form of programmed cell death; regulating trimethylamine N-oxide (TMAO) metabolism; and activating platelet function, both recently identified as pathogenetic factors in MASH progression. Moreover, various exogenous factors impact gut microbiota and its involvement in MASLD-related oxidative stress, such as air pollution, physical activity, cigarette smoke, alcohol, and dietary patterns. This manuscript aims to provide a state-of-the-art overview focused on the intricate interplay between gut microbiota, lipid peroxidation, and MASLD pathogenesis, offering insights into potential strategies to prevent disease progression and its associated complications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Luca Miele
- CEMAD Digestive Diseases Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy (S.A.)
| |
Collapse
|
47
|
Zhang Q, Zhu Q, Xiao Y, Yu Q, Shi S. Co-housing with Tibetan chickens improved the resistance of Arbor Acres chickens to Salmonella enterica serovar Enteritidis infection by altering their gut microbiota composition. J Anim Sci Biotechnol 2025; 16:2. [PMID: 39748400 PMCID: PMC11697627 DOI: 10.1186/s40104-024-01132-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/26/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Salmonella enterica serovar Enteritidis (S. Enteritidis) is a global foodborne pathogen that poses a significant threat to human health, with poultry being the primary reservoir host. Therefore, addressing S. Enteritidis infections in poultry is crucial to protect human health and the poultry industry. In this study, we investigated the effect of co-housing Arbor Acres (AA) chickens, a commercial breed susceptible to S. Enteritidis, with Tibetan chickens, a local breed resistant to S. Enteritidis infection, on the resistance of the latter to the pathogen. RESULTS Ninety-six 1-day-old Tibetan chickens and 96 1-day-old AA chickens were divided into a Tibetan chicken housed alone group (n = 48), an AA chicken housed alone group (n = 48), and a co-housed group (48 birds from each breed for 2 cages). All birds were provided the same diet, and the experimental period lasted 14 d. At d 7, all chickens were infected with S. Enteritidis, and samples were collected at 1-, 3-, and 7-day-post-infection. We found that the body weight of AA chickens significantly increased when co-housed with Tibetan chickens at 1- and 3-day-post-infection (P < 0.05). In addition, the cecal S. Enteritidis load in AA chickens was significantly reduced at 1-, 3-, and 7-day-post-infection (P < 0.05). Furthermore, the inflammatory response in AA chickens decreased, as evidenced by the decreased expression of pro-inflammatory cytokines NOS2, TNF-α, IL-8, IL-1β, and IFN-γ in their cecal tonsils (P < 0.05). Co-housing with Tibetan chickens significantly increased the height of villi and number of goblet cells (P < 0.05), as well as the expression of claudin-1 (P < 0.05), a tight junction protein, in the jejunum of AA chickens. Further analysis revealed that co-housing altered the gut microbiota composition in AA chickens; specifically, the relative abundances of harmful microbes, such as Intestinimonas, Oscillibacter, Tuzzerella, Anaerotruncus, Paludicola, and Anaerofilum were reduced (P < 0.05). CONCLUSIONS Our findings indicate that co-housing with Tibetan chickens enhanced the resistance of AA chickens to S. Enteritidis infection without compromising the resistance of Tibetan chickens. This study provides a novel approach for Salmonella control in practical poultry production.
Collapse
Affiliation(s)
- Qianyun Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Institute of Poultry Sciences, Yangzhou, China
| | - Qidong Zhu
- Jiangsu Institute of Poultry Sciences, Yangzhou, China
| | - Yunqi Xiao
- Jiangsu Institute of Poultry Sciences, Yangzhou, China
| | - Qinghua Yu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.
| | - Shourong Shi
- Jiangsu Institute of Poultry Sciences, Yangzhou, China.
| |
Collapse
|
48
|
Azhar Ud Din M, Lin Y, Lyu C, Yi C, Fang A, Mao F. Advancing therapeutic strategies for graft-versus-host disease by targeting gut microbiome dynamics in allogeneic hematopoietic stem cell transplantation: current evidence and future directions. Mol Med 2025; 31:2. [PMID: 39754054 PMCID: PMC11699782 DOI: 10.1186/s10020-024-01060-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 12/27/2024] [Indexed: 01/06/2025] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is a highly effective therapy for malignant blood illnesses that pose a high risk, as well as diseases that are at risk due to other variables, such as genetics. However, the prevalence of graft-versus-host disease (GVHD) has impeded its widespread use. Ensuring the stability of microbial varieties and associated metabolites is crucial for supporting metabolic processes, preventing pathogen intrusion, and modulating the immune system. Consequently, it significantly affects the overall well-being and susceptibility of the host to disease. Patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) may experience a disruption in the balance between the immune system and gut bacteria when treated with medicines and foreign cells. This can lead to secondary intestinal inflammation and GVHD. Thus, GM is both a reliable indicator of post-transplant mortality and a means of enhancing GVHD prevention and treatment after allo-HSCT. This can be achieved through various strategies, including nutritional support, probiotics, selective use of antibiotics, and fecal microbiota transplantation (FMT) to target gut microbes. This review examines research advancements and the practical use of intestinal bacteria in GVHD following allo-HSCT. These findings may offer novel insights into the prevention and treatment of GVHD after allo-HSCT.
Collapse
Affiliation(s)
- Muhammad Azhar Ud Din
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, No. 8 Dianli Road, Zhenjiang, 212002, Jiangsu, People's Republic of China
- Institute of Hematology, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Yan Lin
- The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, 212399, Jiangsu, People's Republic of China
| | - Changkun Lyu
- School of Medical Technology, Shangqiu Medical College Shangqiu, Shangqiu, 476100, Henan, People's Republic of China
| | - Chengxue Yi
- School of Medical Technology, Zhenjiang College, Zhenjiang, 212028, Jiangsu, People's Republic of China
| | - Anning Fang
- Basic Medical School, Anhui Medical College, 632 Furong Road, Economic and Technological Development Zone, Hefei, 230061, Anhui, People's Republic of China.
| | - Fei Mao
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, No. 8 Dianli Road, Zhenjiang, 212002, Jiangsu, People's Republic of China.
- Institute of Hematology, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China.
| |
Collapse
|
49
|
Wang N, Xin Y. Review: Gut microbiota: Therapeutic targets of ginseng polysaccharides against multiple disorders. Int J Biol Macromol 2025; 287:138527. [PMID: 39662561 DOI: 10.1016/j.ijbiomac.2024.138527] [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: 03/21/2024] [Revised: 11/25/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
As biological macromolecules, ginseng polysaccharides (GP) are often difficult to be directly absorbed through the intestinal cell membrane. It has been found that it can regulate gut microbiota by acting as a prebiotic, and then play a therapeutic role in some diseases, such as diarrhea, tumour, diabetic, dementia, obesity. With the deepening of research, we found that the role played by GP as a prebiotic cannot be ignored. Not only that, it can also affect the immunity and the metabolism and absorption of ginsenosides to play a synergistic role. Overall, GP can regulate the diversity of gut microbiota, which in turn affects the synthesis of secondary metabolites. GP also promotes the transformation of ginsenosides, leading to improved absorptivity of these compounds. This review aims to provide a deeper understanding of how GP interacts with the gut microbiota in various disorders and the transformation of ginsenosides. By exploring these interactions, we can gain valuable insights into the potential benefits of GP in managing different health conditions and enhancing the bioavailability of ginsenosides.
Collapse
Affiliation(s)
- Na Wang
- Department of Pharmacy, The Affliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Yizhou Xin
- Department of Pharmacy, The Affliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| |
Collapse
|
50
|
Liu H, Wang D, Feng X, Liu L, Liu B, Zhu L, Sun J, Zuo X, Chen S, Xian J, Zhang C, Yang W. Sishen Pill & Tongxieyaofang ameliorated ulcerative colitis through the activation of HIF-1α acetylation by gut microbiota-derived propionate and butyrate. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156264. [PMID: 39612887 DOI: 10.1016/j.phymed.2024.156264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic inflammatory bowel disease closely related to gut microbiota dysbiosis and intestinal homeostasis imbalance. Sishen Pill&Tongxieyaofang (SSP-TXYF) has a long history of application in traditional Chinese medicine and is widely used in UC clinics. However, its mechanism of action is still unclear. PURPOSE This study aimed to explore the potential regulatory role of SSP-TXYF in protecting against UC through metabolites produced by the intestinal microbiota, and elucidate its underlying molecular mechanism. STUDY DESIGN AND METHODS 16S rRNA and UPLC-QE-Orbitrap-MS were used to assess the microbiota and short-chain fatty acids (SCFAs). A rat model of 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced gut microbiota dysbiosis was used to study the effects of SSP-TXYF on UC in vivo. Intestinal epithelial cells-6 (IEC-6) were treated with lipopolysaccharide (LPS). The intestinal mucosal barrier (IMB) functions were investigated by alcian blue staining and western blot analysis. The mechanism of SSP-TXYF influenced the HIF-1α acetylation pathway was examined by real-time fluorescence quantitative PCR (qPCR), Western blotting, and Co-immunoprecipitation. RESULTS Using 16S rRNA gene-based microbiota analysis, we found that SSP-TXYF ameliorated TNBS-induced gut microbiota dysbiosis. We found that SSP-TXYF significantly inhibited the decreased abundance of Firmicutes in UC rats, in addition, the abundance of Actinobacteria was also improved. The mechanism of SSP-TXYF-treated TNBS-induced UC resulted from improved IMB functions via the activation of hypoxia-inducible factor-1 (HIF-1α) acetylation. Notably, SSP-TXYF Enriched microbiota-derived metabolites propionate and butyrate, which could activate HIF-1α acetylation in IEC. Furthermore, exogenous treatment of propionate and butyrate reproduced similar protective effects as SSP-TXYF to UC through improving HIF-1α-dependent IMB functions. CONCLUSIONS Overall, our findings suggest that the gut microbiota-propionate/butyrate-HIF-1α-IMB axis plays an important role in SSP-TXYF-maintaining intestinal homeostasis, which may represent a novel approach for UC prevention via the intervention of any link in this axis.
Collapse
Affiliation(s)
- Haifan Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dunfang Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xue Feng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bin Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lin Zhu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jingwei Sun
- Beijing University of Chinese Medicine, Beijing, China
| | - Xingbo Zuo
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Siyuan Chen
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Junying Xian
- Nanning Hospital of Traditional Chinese Medicine, Nanning, China
| | - Caijuan Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Weipeng Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|