1
|
Templeton HN, Ehrlich AT, Schwerdtfeger LA, Sheng JA, Tjalkens RB, Tobet SA. Sex Specific Effects of Environmental Toxin-Derived Alpha Synuclein on Enteric Neuronal-Epithelial Interactions. Neurogastroenterol Motil 2025; 37:e70046. [PMID: 40273372 PMCID: PMC12163210 DOI: 10.1111/nmo.70046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 02/04/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Parkinson's Disease (PD) is a neurodegenerative disorder with prodromal gastrointestinal (GI) issues often emerging decades before motor symptoms. Pathologically, PD can be driven by the accumulation of misfolded alpha synuclein (aSyn) protein in the brain and periphery, including the GI tract. Disease epidemiology differs by sex, with men twice as likely to develop PD. Women, however, experience faster disease progression, higher mortality, and more severe GI symptoms. Gut calcitonin gene-related peptide (CGRP) is a key regulator of intestinal contractions and visceral pain. The current study tests the hypothesis that sex differences in GI symptomatology in PD are the result of aSyn aggregation altering enteric CGRP signaling pathways. METHODS To facilitate peripheral aSyn aggregation, the pesticide rotenone was administered intraperitoneally once daily for 2 weeks to male and female mice. Mice were sacrificed 2 weeks after the last rotenone injection, and immunohistochemistry was performed on sections of proximal colon. KEY RESULTS Levels of aSyn were heightened in PGP9.5 immunoreactive myenteric plexus neurons, a subset of which were immunoreactive to CGRP and showed a similar increase in aSyn immunoreactivity in rotenone-treated mice. Female mice exhibited 153% more myenteric aSyn, 26% more apical CGRP immunoreactivity in the mucosa, and 66.7% more aSyn in apical CGRP+ fibers after rotenone when compared to males. Goblet cell numbers were diminished, but the individual cells were larger in the apical regions of crypts in the colons of rotenone-treated mice with no difference between males and females. CONCLUSIONS This study used a mouse model of PD to uncover sex-specific alterations in enteric neuronal and epithelial populations, underscoring the importance of considering sex as a biological variable while investigating prodromal GI symptoms.
Collapse
Affiliation(s)
- Hayley N. Templeton
- Department of Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Alexis T. Ehrlich
- Department of Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Luke A. Schwerdtfeger
- Ann Romney Center for Neurological DiseaseHarvard Medical School, Brigham and Women's HospitalBostonMassachusettsUSA
| | - Julietta A. Sheng
- Department of Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Ronald B. Tjalkens
- Department of Environmental and Radiological Health SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Stuart A. Tobet
- Department of Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
- School of Biomedical EngineeringColorado State UniversityFort CollinsColoradoUSA
| |
Collapse
|
2
|
Sun Y, Wu J, Li H, Zhong Y, Ye Z, Zhang J, Su M. Gut microbiota dysbiosis triggered by salinity stress enhances systemic inflammation in spotted scat (Scatophagus argus). FISH & SHELLFISH IMMUNOLOGY 2025; 162:110353. [PMID: 40254087 DOI: 10.1016/j.fsi.2025.110353] [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: 01/25/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
As an ecological disturbance, salinity changes substantially impact aquatic organism health. Gut microbiota plays a pivotal role in host health and exhibits heightened sensitivity to environmental salinity stress; however, the potential correlative mechanisms between gut microbiota dysbiosis triggered by salinity changes and host health remain unclear. The present study conducted a 4-week stress experiment to investigate the precise impact of gut microbiota on the inflammatory response in Scatophagus argus under different salinities (0 ‰ [hyposaline group, HO], 25 ‰ [control group, CT], and 40 ‰ [hypersaline group, HE]). Our results revealed that both HO and HE stress significantly changed the relative abundances of Gram-negative bacteria and the impairment of intestinal barrier function. Subsequently, the levels of lipopolysaccharide (LPS) in the serum exhibited a significant increase, and the expression levels of genes (tlrs, myd88, irak1, irak4, and traf6) involving TLRs/MyD88/NF-κB signaling pathway and pro-inflammatory cytokines (il-6, il-8, il-1β, and tnf-α) in the representative immune organs were significantly upregulated. Conversely, the abundance of the anti-inflammatory gene (tgf-β1) and its protein contents in serum were decreased. Transplantation of the gut microbiota from S. argus exposed to varying salinities into germ-free Oryzias latipes resulted in an enhanced inflammatory response. Our results suggested that both HO and HE stress increased the presence of Gram-negative bacteria and disrupted the intestinal barrier, leading to elevated serum LPS and subsequent systemic inflammation in fish. These findings provide innovative insights into the influence of salinity manipulation strategies on the health of aquatic organisms, contributing to the mariculture management in coastal areas.
Collapse
Affiliation(s)
- Yuan Sun
- Shenzhen Key Laboratory of Marine Bioresource & Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China; Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Jiajia Wu
- Shenzhen Key Laboratory of Marine Bioresource & Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Huixue Li
- Shenzhen Key Laboratory of Marine Bioresource & Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Youling Zhong
- Shenzhen Key Laboratory of Marine Bioresource & Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Zhiyin Ye
- Shenzhen Key Laboratory of Marine Bioresource & Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Junbin Zhang
- Shenzhen Key Laboratory of Marine Bioresource & Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China
| | - Maoliang Su
- Shenzhen Key Laboratory of Marine Bioresource & Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
3
|
Hu X, Wang R, Kille P, Maret W, Hogstrand C. Zinc amino acid chelate and the Aryl Hydrocarbon Receptor (AHR) cooperate in improving the barrier function of a Caco-2 cell intestinal epithelium. J Nutr Biochem 2025; 141:109909. [PMID: 40154643 DOI: 10.1016/j.jnutbio.2025.109909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
Zinc and several physiologically relevant ligands of the aryl hydrocarbon receptor (AHR) are nutrients that promote intestinal barrier function. We have identified that AHR activation upregulates the expression of zinc importers in the intestinal epithelium to increase intracellular zinc concentrations, which leads to improved epithelial barrier function. Here, we investigated if an amino acid chelate of zinc, in cooperation with AHR activation, can improve the barrier function of a differentiated Caco-2 cell epithelium. Functional assays of the Caco-2 cell epithelium demonstrate that both ZnSO4 and a lysine and glutamic acid chelate of Zn, in combination with the physiological AHR agonist 6-formylindolo[3,2-b]carbazole (FICZ), increase expression of tight junction proteins at the mRNA and protein levels. FICZ increases uptake of zinc into the epithelium in the presence of ZnSO4 or the amino acid Zn chelate in the medium to equal extents. We conclude that the lysine and glutamic acid chelate of Zn is as efficacious as ZnSO4 in reducing permeability of the Caco-2 cell epithelium in the presence of FICZ. The results suggest that dietary supplementation with bioavailable forms of zinc together with nutritional AHR agonists may be beneficial in improving gut barrier function and help prevent inflammatory bowel disease (IBD).
Collapse
Affiliation(s)
- Xiuchuan Hu
- Department of Nutritional Sciences, School of Life Course and Population Sciences, King's College London, London, UK
| | - Rui Wang
- Department of Nutritional Sciences, School of Life Course and Population Sciences, King's College London, London, UK
| | - Peter Kille
- School of Biosciences, Cardiff University, Cardiff, UK
| | - Wolfgang Maret
- Department of Nutritional Sciences, School of Life Course and Population Sciences, King's College London, London, UK
| | - Christer Hogstrand
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK.
| |
Collapse
|
4
|
Wang Y, Liu Z, Wang Z, Xia J, Fu G. Dietary Litsea cubeba essential oil enhances growth, immunity, and intestinal health in channel catfish. FISH & SHELLFISH IMMUNOLOGY 2025; 165:110487. [PMID: 40516798 DOI: 10.1016/j.fsi.2025.110487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 05/30/2025] [Accepted: 06/10/2025] [Indexed: 06/16/2025]
Abstract
This study investigated the effects of dietary supplementation with Litsea cubeba essential oil (LEO) on growth performance, immune function, intestinal health, and microbiota composition in channel catfish (Ictalurus punctatus). An eight-week feeding trial was conducted with 750 fish (60.00 ± 0.50 g) assigned to five diets: 0 (CON), 100 mg/kg (LEO100), 200 mg/kg (LEO200), 400 mg/kg (LEO400), and 800 mg/kg (LEO800). The LEO100 and LEO200 groups showed significant improvements in growth performance, immune response (C3, C4, and ACP levels), and digestive enzyme activities (amylase, lipase, and trypsin). Gene expression analysis revealed an upregulation of genes related to intestinal barrier integrity (zo-1, zo-2, and occludin), immunoregulatory cytokines (tgf-β1, tgf-β2, tgf-β3, and il-10), and key transcriptional regulators nf-κb and stat3). Conversely, pro-inflammatory mediators (il-8, il-1β, and tlr5) were significantly downregulated. In the LEO100 and LEO200 groups, the protein expression of TNF-α and NF-κB was elevated. Additionally, intestinal morphology (villus height, muscle thickness, and goblet cell count) and beneficial microbiota (Lactococcus and Plesiomonas) were enhanced in the LEO100 and LEO200 groups. These results demonstrate that dietary supplementation with LEO at 100-200 mg/kg improves growth performance, immune function, and intestinal health in channel catfish, making it a promising natural feed additive for aquaculture.
Collapse
Affiliation(s)
- Yan Wang
- Fisheries College, Hunan Agricultural University, Changsha, Hunan, 410128, PR China
| | - Zhimou Liu
- Hunan Nuoz Biological Technology Co., Ltd., Yiyang, 413001, PR China
| | - Zhenbin Wang
- Hunan Nuoz Biological Technology Co., Ltd., Yiyang, 413001, PR China
| | - Junhong Xia
- State Key Laboratory of Biocontrol, Institute of Aquatic Economic Animals and Guangdong Provincial Key Laboratory for Aquatic Economic Animals, College of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, PR China
| | - Guihong Fu
- Fisheries College, Hunan Agricultural University, Changsha, Hunan, 410128, PR China.
| |
Collapse
|
5
|
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
|
6
|
Sato S, Hasan AU, Obara M, Kondo Y, Taira E. Long-term consumption of moderate amounts of sucrose-sweetened drinks disrupts intestinal barrier function by impairing goblet cell differentiation. Cell Tissue Res 2025; 400:273-285. [PMID: 40072586 DOI: 10.1007/s00441-025-03961-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025]
Abstract
While the prolonged consumption of sucrose-containing beverages is known to impact many organs, their specific effects on the small intestine remain elusive. This study aimed to evaluate how regular intake of sucrose, in amounts typically consumed, affects goblet cells, which play a critical role in regulating the mucosal barrier and innate immune defenses in the small intestine. Ten-week-old male ddY mice, a model of diet-induced obesity, were given a regular diet with either plain water or 7% sucrose water. Caloric intake was monitored weekly through food and drink measurements. After 8 weeks, glucose and insulin responses were evaluated following an oral gavage of glucose or sucrose. At 14 weeks, plasma, whole small intestine, and liver samples were collected. Despite achieving an isocaloric state, mice drinking sucrose water showed approximately a 1.5-fold increase in body weight and impaired glucose tolerance. In the small intestine, genes involved in sucrose digestion and absorption (Sis, Sglt1, Glut2, and Glut5) were upregulated, while genes essential for maintaining the intestinal barrier and function (Epcam, Fabp2, Cldn1, Ocln, and Tjp1) were downregulated. Serum levels and mRNA expression of the inflammatory cytokine, interleukin-18 were elevated. Genes responsible for goblet cell differentiation and function (Hes1, Gfi1, Spdef, and Klf4) were downregulated, leading to an increase in immature goblet cells and a decrease in mucin-producing markers (Muc2, Muc4, and Muc13) in the jejunum. The findings underscore that besides obesity, long-term intake of sucrose-containing drinks provokes localized inflammation and disrupts small intestinal barrier function by impairing goblet cell differentiation and activity.
Collapse
Affiliation(s)
- Sachiko Sato
- Department of Pharmacology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Arif U Hasan
- Department of Pharmacology, School of Medicine, Iwate Medical University, Iwate, Japan.
| | - Mami Obara
- Department of Pharmacology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Yukiko Kondo
- Department of Pharmacology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Eiichi Taira
- Department of Pharmacology, School of Medicine, Iwate Medical University, Iwate, Japan
| |
Collapse
|
7
|
Jiang Y, Chen J, Du Y, Fan M, Shen L. Immune modulation for the patterns of epithelial cell death in inflammatory bowel disease. Int Immunopharmacol 2025; 154:114462. [PMID: 40186907 DOI: 10.1016/j.intimp.2025.114462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/23/2025] [Accepted: 03/08/2025] [Indexed: 04/07/2025]
Abstract
Inflammatory bowel disease (IBD) is an inflammatory disease of the intestine whose primary pathological presentation is the destruction of the intestinal epithelium. The intestinal epithelium, located between the lumen and lamina propria, transmits luminal microbial signals to the immune cells in the lamina propria, which also modulate the intestinal epithelium. In IBD patients, intestinal epithelial cells (IECs) die dysfunction and the mucosal barrier is disrupted, leading to the recruitment of immune cells and the release of cytokines. In this review, we describe the structure and functions of the intestinal epithelium and mucosal barrier in the physiological state and under IBD conditions, as well as the patterns of epithelial cell death and how immune cells modulate the intestinal epithelium providing a reference for clinical research and drug development of IBD. In addition, according to the targeting of epithelial apoptosis and necroptotic pathways and the regulation of immune cells, we summarized some new methods for the treatment of IBD, such as necroptosis inhibitors, microbiome regulation, which provide potential ideas for the treatment of IBD. This review also describes the potential for integrating AI-driven approaches into innovation in IBD treatments.
Collapse
Affiliation(s)
- Yuting Jiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Jie Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Yaoyao Du
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Minwei Fan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Lan Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
8
|
Livingston DBH, Sweet A, Chowdary M, Demissie MS, Rodrigue A, Gedara KP, Kishore L, Mahmoodianfard S, Power KA. Diet alters the effects of lipopolysaccharide on intestinal health and cecal microbiota composition in C57Bl/6 male mice. J Nutr Biochem 2025:109951. [PMID: 40345399 DOI: 10.1016/j.jnutbio.2025.109951] [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: 12/03/2024] [Revised: 02/13/2025] [Accepted: 05/02/2025] [Indexed: 05/11/2025]
Abstract
Lipopolysaccharide (LPS), a component of the gram-negative bacteria cell well, is established to induce an acute and transient inflammatory cascade upon exogenous administration in mice, while negatively impacting aspects of the microbiota gut-brain axis (mGBA). Dietary supplementation with flaxseed (FS) has been shown to partially attenuate these impairments across the mGBA, although the importance of intestinal and hepatic health has not been established in this context. In this study, we furthered our investigation of the impact of dietary supplementation with FS and FS oil (FO) in mitigating LPS-induced perturbations to intestinal health (i.e. cecal microbiota, barrier integrity, inflammation, short-chain fatty acid (SCFA) production), hepatic inflammation, and their relationships to LPS-induced systemic and neuroinflammation. We noted distinct, diet-dependent shifts in the microbiota composition twenty-four hours post-LPS injection, that may be partially driven by the LPS-induced anorexia. BD-fed mice challenged with LPS showed reduced Muribaculaceae, while FS-LPS mice had elevated Akkermansia, and both the FS-LPS and FO-LPS mice had reduced Bacteroides, each with unique correlations to diet intake. These LPS-driven shifts in the microbiota were coupled with elevated ileal mucous content in the FS- and FO-fed mice, and intestinal inflammation which was partially attenuated in the FS-fed mice. Collectively, we highlight that FS modulates aspects of the intestinal microenvironment, which may be related to the anti-inflammatory effects across the mGBA.
Collapse
Affiliation(s)
- Dawson B H Livingston
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada, K1H 8L1
| | - Allison Sweet
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada, K1H 8L1
| | - Maryam Chowdary
- Faculty of Science, Department of Biology, University of Ottawa, Ottawa, Canada, K1N 9A7
| | - Meron Samuel Demissie
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada, K1H 8L1
| | - Alexane Rodrigue
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada, K1H 8L1
| | - Kasuni Pillagawa Gedara
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada, K1H 8L1
| | - Lalit Kishore
- Faculty of Health Science, School of Nutrition Sciences, University of Ottawa, Ottawa, Canada, K1N 6N5
| | - Salma Mahmoodianfard
- Faculty of Health Science, School of Nutrition Sciences, University of Ottawa, Ottawa, Canada, K1N 6N5
| | - Krista A Power
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada, K1H 8L1; Faculty of Health Science, School of Nutrition Sciences, University of Ottawa, Ottawa, Canada, K1N 6N5.
| |
Collapse
|
9
|
Martínez‐Augustin O, Tena‐Garitaonaindia M, Ceacero‐Heras D, Jiménez‐Ortas Á, Enguix‐Huete JJ, Álvarez‐Mercado AI, Ruiz‐Henares G, Aranda CJ, Gámez‐Belmonte R, Sánchez de Medina F. Macronutrients as Regulators of Intestinal Epithelial Permeability: Where Do We Stand? Compr Rev Food Sci Food Saf 2025; 24:e70178. [PMID: 40421830 PMCID: PMC12108046 DOI: 10.1111/1541-4337.70178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/23/2025] [Accepted: 04/04/2025] [Indexed: 05/28/2025]
Abstract
The intestinal barrier function (IBF) is essential for intestinal homeostasis. Its alterations have been linked to intestinal and systemic disease. Regulation of intestinal permeability is key in the maintenance of the IBF, in which the intestinal epithelium and tight junctions, the mucus layer, sIgA, and antimicrobial peptides are important factors. This review addresses the concept of IBF, focusing on permeability, and summarizes state-of-the-art information on how starvation and macronutrients regulate it. Novel mechanisms regulate intestinal permeability, like its induction by the normal process of nutrient absorption, the contribution of starvation-induced autophagy, or the stimulation of sIgA production by high-protein diets in a T-cell-independent fashion. In addition, observations evidence that starvation and protein restriction increase intestinal permeability, compromising mucin, antimicrobial peptides, and/or intestinal sIgA production. Regarding specific macronutrients, substantial evidence indicates that casein (compared to other protein sources), specific protein-derived peptides and glutamine reinforce IBF. Dietary carbohydrates regulate intestinal permeability in a structure- and composition-dependent fashion; fructose, glucose, and sucrose increase it, while nondigestible oligosaccharides (NDOs) decrease it. Among NDOs, human milk oligosaccharides (HMOs) stand as a promising tool. NODs effects are mediated by intestinal microbiota modulation, production of short-chain fatty acids, and direct interactions with intestinal cells. Finally, evidence supports avoiding high-fat diets for their detrimental effects on IBF. Most studies have been carried out in vitro or in animal models. More information is needed from clinical studies to substantiate beneficial effects and the use of macronutrients in the treatment and prevention of IBF-related diseases.
Collapse
Affiliation(s)
- Olga Martínez‐Augustin
- Department of Biochemistry and Molecular Biology II, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Insituto de Nutrición y Tecnología de los alimentos José Mataix and Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Mireia Tena‐Garitaonaindia
- Department of Biochemistry and Molecular Biology II, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Insituto de Nutrición y Tecnología de los alimentos José Mataix and Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Diego Ceacero‐Heras
- Department of Biochemistry and Molecular Biology II, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Insituto de Nutrición y Tecnología de los alimentos José Mataix and Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Ángela Jiménez‐Ortas
- Department of Biochemistry and Molecular Biology II, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Insituto de Nutrición y Tecnología de los alimentos José Mataix and Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Juan J. Enguix‐Huete
- Department of Pharmacology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Ana I. Álvarez‐Mercado
- Department of Pharmacology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Guillermo Ruiz‐Henares
- Department of Pharmacology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| | - Carlos J. Aranda
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina‐ IBIMA Plataforma BIONANDRICORS “Enfermedades inflamatorias”MálagaSpain
| | - Reyes Gámez‐Belmonte
- Department of Pharmacology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
- Department of Medicine 1University of Erlangen‐NurembergErlangenGermany
| | - Fermín Sánchez de Medina
- Department of Pharmacology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), School of Pharmacy, Instituto de Investigación Biosanitaria ibs.GRANADAUniversity of GranadaGranadaSpain
| |
Collapse
|
10
|
Tanabe M, Kunisawa K, Saito I, Kosuge A, Tezuka H, Kawai T, Kon Y, Yoshidomi K, Kagami A, Hasegawa M, Kubota H, Ojika H, Fujii T, Tochio T, Hirooka Y, Saito K, Nabeshima T, Mouri A. Adolescent social isolation decreases colonic goblet cells and impairs spatial cognition through the reduction of cystine. Mol Psychiatry 2025; 30:2137-2151. [PMID: 39613916 PMCID: PMC12014494 DOI: 10.1038/s41380-024-02826-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 12/01/2024]
Abstract
Negative experiences during adolescence, such as social isolation (SI), bullying, and abuse, increase the risk of psychiatric diseases in adulthood. However, the pathogenesis of psychiatric diseases induced by these factors remain poorly understood. In adolescents, stress affects the intestinal homeostasis in the gut-brain axis. This study determined whether adolescent SI induces behavioral abnormalities by disrupting colonic function. Adolescent mice exposed to SI exhibit spatial cognitive deficits and microglial activation in the hippocampus (HIP). SI decreased the differentiation of mucin-producing goblet cells, which was accompanied by alterations in the composition of the gut microbiota, particularly the depletion of mucin-feeding bacteria. Treatment with rebamipide, which promotes goblet cell differentiation in the colon, attenuated SI-induced spatial cognitive deficits and microglial activation in the HIP and decreased cystine, a downstream metabolite of homocysteine. Treatment with cystine ameliorated SI-induced spatial cognitive deficits and increased microglial C-C motif chemokine ligand 7 (CCL7) levels in the HIP. Inhibition of CCL7 receptors by antagonists of CC motif chemokine receptors 2 (CCR2) and 3 (CCR3) in the HIP prevented spatial cognitive deficits induced by SI. Infusion of CCL7 into the HIP following microglial ablation with clodronate liposome induced spatial cognitive deficits. These findings suggest that adolescent SI decreases serum cystine levels by damaging the colonic goblet cells, resulting in spatial cognitive deficits by triggering microglial activation in the HIP. Our results indicate that increased CCL7 expression in hippocampal microglia may contribute to spatial cognitive deficits by activating CCR2 and CCR3.
Collapse
Affiliation(s)
- Moeka Tanabe
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
- Laboratory of Health and Medical Science Innovation, Fujita Health University Graduate School of Medical Science, Toyoake, Aichi, Japan
| | - Kazuo Kunisawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan.
- International Center for Brain Science (ICBS), Fujita Health University, Toyoake, Aichi, Japan.
| | - Imari Saito
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Aika Kosuge
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Hiroyuki Tezuka
- Department of Cellular Function Analysis, Research Promotion Headquarters, Fujita Health University, Toyoake, Aichi, Japan
| | - Tomoki Kawai
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Yuki Kon
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Koyo Yoshidomi
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Akari Kagami
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Masaya Hasegawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Hisayoshi Kubota
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
- Division of Behavioral Neuropharmacology, International Center for Brain Science (ICBS), Fujita Health University, Toyoake, Aichi, Japan
| | - Haruto Ojika
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Tadashi Fujii
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Toyoake, Aichi, Japan
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake, Aichi, Japan
| | - Takumi Tochio
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Toyoake, Aichi, Japan
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake, Aichi, Japan
| | - Yoshiki Hirooka
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Toyoake, Aichi, Japan
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake, Aichi, Japan
| | - Kuniaki Saito
- Laboratory of Health and Medical Science Innovation, Fujita Health University Graduate School of Medical Science, Toyoake, Aichi, Japan
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Medical Science, Toyoake, Aichi, Japan
| | - Toshitaka Nabeshima
- Laboratory of Health and Medical Science Innovation, Fujita Health University Graduate School of Medical Science, Toyoake, Aichi, Japan
- International Center for Brain Science (ICBS), Fujita Health University, Toyoake, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research (J-DO), Nagoya, Aichi, Japan
| | - Akihiro Mouri
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan.
- International Center for Brain Science (ICBS), Fujita Health University, Toyoake, Aichi, Japan.
- Japanese Drug Organization of Appropriate Use and Research (J-DO), Nagoya, Aichi, Japan.
| |
Collapse
|
11
|
Luo Y, Luo L, Xia M, Liu Q, Zhang G. Studies on the changes in rectal permeability and intestinal microbiota with developmental age in young rats. Front Microbiol 2025; 16:1551693. [PMID: 40336831 PMCID: PMC12058081 DOI: 10.3389/fmicb.2025.1551693] [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: 12/26/2024] [Accepted: 03/31/2025] [Indexed: 05/09/2025] Open
Abstract
Introduction The gut contains a diverse array of commensal microorganisms, forming a vital biological barrier within the intestine that contributes to the overall intestinal mucosal barrier. However, research on the rectal barrier during early development remains limited. This study aims to investigate the relationship between intestinal microbiota and rectal barrier function in young rats. Methods We evaluated the rectal barrier structure and function in rats at 2-, 4-, and 10-week-old. Methodology included histological analysis, Muc2 expression quantification, immunofluorescence localization of tight junction proteins (ZO-1, Occludin, Claudins), blood glucose monitoring after rectal insulin administration, and 16S rDNA sequencing of rectal microbiota. Spearman correlation analysis was used to explore mechanisms linking age-dependent changes in rectal permeability to microbiota dynamics. Results Physiological rectal permeability was significantly higher in 2-week-old rats compared to 4- and 10-week-old rats (p < 0.01), although systemic biomarkers (LPS, D-LA, and LBP) showed no significant differences. The rectal microbiota exhibited marked age-dependent shifts in composition, α/β-diversity, and metabolic pathways, with increased abundance of beneficial taxa (e.g., Muribaculaceae, Akkermansia) in older rats. Correlation analysis revealed strong associations between reduced permeability, elevated Occludin expression, and microbiota maturation (R = 0.65, p < 0.001). Conclusion This study demonstrates that age-dependent maturation of the rectal barrier is closely linked to microbiota composition and tight junction protein expression, providing insights into developmental mechanisms and potential strategies for pediatric rectal drug delivery.
Collapse
Affiliation(s)
- Yunfeng Luo
- Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Liangming Luo
- Yudu County Hospital of Traditional Chinese Medicine, Ganzhou, China
| | - Mengle Xia
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Qian Liu
- Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Guosong Zhang
- Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
12
|
Momoh TA, Odu-Onikosi SG, Amulejoye FD, Wilson J, Eynon B, Kühlwein H, Kuri V, Merrifield DL. Brewers' Yeast ( Saccharomyces cerevisiae) Purified Functional Feed Additives Mitigate Soybean Meal-Induced Enteritis in Atlantic Salmon ( Salmo salar) Parr. AQUACULTURE NUTRITION 2025; 2025:8555658. [PMID: 40255579 PMCID: PMC12006713 DOI: 10.1155/anu/8555658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/17/2025] [Indexed: 04/22/2025]
Abstract
Soybean meal (SBM) is commonly used in aquafeeds due to its wide availability, reasonable protein content, and cost-effectiveness. However, high SBM inclusion levels in the diets of carnivorous fish, such as Atlantic salmon (Salmo salar), can cause soybean meal-induced enteritis (SBMIE), resulting in compromised gut health, reduced nutrient absorption, and impaired growth. An 8-week study was conducted to evaluate the potential of brewers' yeast-derived functional feed additives (FFAs), specifically yeast cell wall β-glucans (PβG [purified β-glucan]) and yeast cytosolic extracts (YEs), to mitigate the adverse effects of SBMIE in Atlantic salmon parr. Fish were fed diets containing 30% SBM (30-SBM) with either 0.02% β-glucan (30-SBM+PβG) or YE at 1% (30-SBM+YE1) and 2.5% (30-SBM+YE2.5) inclusion levels and compared against a control diet without SBM (0-SBM). The study assessed growth performance, haematological parameters, distal intestinal morphology, and the distal intestinal gene expression levels of enteritis biomarkers (casp3b, pcna, and hsp70). The results showed that PβG and 1% YE supplementation significantly reduced the severity of SBMIE, with improvements in intestinal morphology, including reduced intraepithelial leukocytes (IELs) levels and goblet cell hyperplasia. Intestinal gene expression levels of casp3b and pcna were significantly downregulated in the PβG and YE fed fish relative to the 30-SBM fed fish, indicating reduced apoptosis and more controlled cell proliferation. However, the effects of 2.5% YE supplementation were less pronounced, indicating a dose-dependent response. These findings demonstrate that both PβG and YE from 100% Saccharomyces cerevisiae can alleviate SBMIE in juvenile Atlantic salmon by supporting gut health and modulating cellular recovery processes.
Collapse
Affiliation(s)
- Taofik A. Momoh
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth, UK
| | - Sheu Gbolahan Odu-Onikosi
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth, UK
| | - Folasade Damilola Amulejoye
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth, UK
- Department of Fisheries and Aquaculture Technology, Olusegun Agagu University of Science and Technology, Okitipupa, Nigeria
| | - Joshua Wilson
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth, UK
| | - Benjamin Eynon
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth, UK
| | | | - Victor Kuri
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth, UK
| | - Daniel L. Merrifield
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth, UK
| |
Collapse
|
13
|
Jia R, Han Y, Zhu Q, Zhang J, Zhang H, Ka M, Ma Y, Gamah M, Zhang W. Activation of notch signaling pathway is a potential mechanism for mucin2 reduction and intestinal mucosal barrier dysfunction in high-altitude hypoxia. Sci Rep 2025; 15:12154. [PMID: 40204779 PMCID: PMC11982276 DOI: 10.1038/s41598-025-96176-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 03/26/2025] [Indexed: 04/11/2025] Open
Abstract
High-altitude hypoxia can cause gastrointestinal issues and damage the intestinal mucosal barrier, which is crucial for digestion and nutrient absorption. The Notch signaling pathway affects this barrier's integrity. This study explores the Notch pathway's role in hypoxia-induced intestinal injury. C57BL/6 mice were used to model intestinal mucosal barrier injury through dextran sodium sulfate (DSS) and hypobaric hypoxia (simulating 5000 m altitude for 7 days). Mice were treated with Notch inhibitor Dibenzazepine (DBZ) and Mucin2 (MUC2) activator Prostaglandin E2 (PGE2). We evaluated weight, colon length, histology, Zonula occludens 1 (ZO-1) and Claudin-1 levels, MUC2 and Notch1 staining, serum diamine oxidase (DAO) and D-lactate (D-La), inflammatory markers, and Notch pathway proteins. DSS and hypoxia caused weight loss, colon shortening, ulcers, and inflammation, with fewer goblet cells and lower MUC2 levels. Elevated serum DAO, D-La, and inflammatory markers indicated severe intestinal damage. DBZ treatment post-DSS and hypoxia significantly reduced these symptoms. PGE2 activation of MUC2 also alleviated symptoms and mitigated intestinal damage. Hypoxia worsens DSS-induced mucosal barrier disruption by activating the Notch pathway, shifting stem cell differentiation towards absorptive cells instead of goblet cells, reducing MUC2 secretion, and intensifying damage. Targeting the Notch pathway and enhancing MUC2 expression could effectively treat hypoxia-induced intestinal injury.
Collapse
Affiliation(s)
- Ruhan Jia
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Ying Han
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Qinfang Zhu
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
- Qinghai Provincial People's Hospital, Xining, Qinghai, China
| | - Jingxuan Zhang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Huan Zhang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
- Department of Pathology, The Second Affiliated Hospital of Xi'an Jiaotong University, 710004, Xi'an, China
| | - Maojia Ka
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Yi Ma
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Mohammed Gamah
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China
| | - Wei Zhang
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining, China.
| |
Collapse
|
14
|
Kasem SM, Dkhil MA, Marey AM, Abdel-Gaber R, Thagfan FA, Elshanat S. Juglone as a Therapeutic Agent in Murine Coccidiosis: Enhancing Intestinal Recovery and Regulating Host Immune Responses. Microsc Res Tech 2025. [PMID: 40186391 DOI: 10.1002/jemt.24871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/18/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
Coccidiosis, caused by Eimeria spp., significantly impacts gastrointestinal health and leads to economic losses. This study investigated juglone, a phenolic compound, as a potential treatment for mouse coccidiosis caused by Eimeria papillata. Twenty-five male Swiss albino mice were categorized into five groups: Group 1: non-infected, Group 2: juglone control (JG), Group 3: infected with 1,000 sporulated oocysts, Group 4: infected-treated with juglone (Infected + JG), and Group 5: infected-treated with anticoccidial drug, amprolium (Infected + ACD). Following 5 days of treatment, the mice were euthanized, and numerous analyses were performed such as growth performance, oocyst shedding, oxidative stress, histology, goblet cell response, and inflammation. Results showed that E. papillata infection significantly decreased body weight and feed intake, increased oocyst shedding, and elevated oxidative stress markers while reducing antioxidant enzyme levels. Histopathological analysis indicated severe intestinal damage, with intracellular parasitic stages and reduced goblet cell density and MUC-2 expression. Infection also heightened gene expression of inflammatory cytokines including tumor necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ), interleukin 1β (IL1β), and interleukin 10 (IL10). Juglone treatment effectively mitigated these negative effects, improving body weight and feed consumption, lowering oocyst shedding, and restoring antioxidant enzyme activity. It also repaired intestinal structure, enhanced goblet cell density and MUC-2 expression, as well as diminished inflammatory cytokines. The findings indicated that juglone significantly mitigates the pathological effects induced by E. papillata by decreasing parasite shedding, oxidative stress, inflammation, and modulating immune responses while facilitating intestinal recovery and enhancing overall health in infected mice.
Collapse
Affiliation(s)
- Shaimaa M Kasem
- Zoology Department, Faculty of Science, Kafrelsheikh University, Kafr Elsheikh, Egypt
| | - Mohamed A Dkhil
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
- Applied Science Research Center, Applied Science Private University, Amman, Jordan
| | - Amal M Marey
- Chemistry Department (Biotechnology Program), Faculty of Science, Helwan University, Cairo, Egypt
| | - Rewaida Abdel-Gaber
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Felwa A Thagfan
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Sherif Elshanat
- Department of Parasitology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
15
|
Jeong S, Davis CK, Chokkalla AK, Kim B, Park S, Vemuganti R. Fecal microbiota transplantation fails to impart the benefits of circadian-dependent intermittent fasting following ischemic stroke. J Cereb Blood Flow Metab 2025; 45:779-789. [PMID: 39917846 PMCID: PMC11806450 DOI: 10.1177/0271678x251319636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/28/2024] [Accepted: 01/21/2025] [Indexed: 02/11/2025]
Abstract
Intermittent fasting (IF) is known to induce significant ischemic tolerance. Diet is a major proponent of gut microbiota, and gut microbial dysbiosis plays a role in post-stroke brain damage. Hence, we currently evaluated whether IF-mediated ischemic tolerance is mediated by gut microbiota. Additionally, circadian cycle is known to modulate post-ischemic outcomes, and thus we further evaluated if gut microbiota would be influenced by prophylactic IF during the inactive phase (fasting during daytime; IIF) or active phase (fasting during nighttime; AIF). The AIF, but not IIF, cohort showed a significantly decreased fecal Firmicutes/Bacteroidetes ratio compared with the ad libitum (AL) cohort. Moreover, the levels of gut microbiota-derived metabolites butyrate and propionate decreased in AL cohort following focal ischemia, whereas they increased in AIF cohort. However, fecal microbiota transplantation (FMT) from IIF or AIF cohort had no significant effects on post-ischemic motor and cognitive function recovery, anxiety-, and depression-like behaviors compared with FMT from AL cohort. Furthermore, FMT from IIF or AIF cohort did not influence the post-ischemic infarct volume, atrophy volume or white matter damage. Overall, the current findings indicate that the beneficial effects of IF after focal ischemia are not mediated by the gut microbiota.
Collapse
Affiliation(s)
- Soomin Jeong
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
| | - Charles K Davis
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Anil K Chokkalla
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Bori Kim
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Sena Park
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
- William S. Middleton Veterans Hospital, Madison, WI, USA
| |
Collapse
|
16
|
Freitas JA, Nehmi Filho V, Santamarina AB, Murata GM, Franco LAM, Fonseca JV, Martins RC, Souza GA, Benicio G, Sabbag IM, de Souza EA, Otoch JP, Pessoa AFM. Nutraceutical supplement slim reshaped colon histomorphology and reduces Mucispirillum schaedleri in obese mice. Front Microbiol 2025; 16:1494994. [PMID: 40236479 PMCID: PMC11997693 DOI: 10.3389/fmicb.2025.1494994] [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: 09/24/2024] [Accepted: 02/28/2025] [Indexed: 04/17/2025] Open
Abstract
Introduction Bioactive compounds and whole foods have emerged as promising interventions to address gut microbiota dysbiosis linked to obesity. Compounds such as berberine and coenzyme Q10 are well-recognized for their roles in managing metabolic syndrome and exerting antioxidant effects, while beet pulp, rich in fiber and antioxidants, enhances gut health through additional prebiotic benefits. Methods This study evaluated the effects of a nutraceutical supplement, Slim, on the modulation of gut microbiota in obese mice induced by a high-fat diet. Results Our results demonstrated that Slim supplementation significantly improved lipid metabolism, reshaped colon histomorphology, and decreased levels of Mucispirillum schaedleri, which were correlated with VLDL-c and triglycerides. Discussion We suggest these effects are driven by a duplibiotic effect, resulting from the synergistic action of the bioactive compounds.
Collapse
Affiliation(s)
- Jessica Alves Freitas
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
- Pesquisa e Desenvolvimento Efeom Nutrição S/A, São Paulo, SP, Brazil
| | - Victor Nehmi Filho
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
- Pesquisa e Desenvolvimento Efeom Nutrição S/A, São Paulo, SP, Brazil
| | - Aline Boveto Santamarina
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
- Pesquisa e Desenvolvimento Efeom Nutrição S/A, São Paulo, SP, Brazil
| | - Gilson Masahiro Murata
- Universidade de São Paulo Faculdade de Medicina da Universidade de São Paulo, Departamento de Clínica Médica, Laboratório de Nefrologia (LIM-29), São Paulo, SP, Brazil
| | - Lucas Augusto Moyses Franco
- Universidade de São Paulo Instituto de Medicina Tropical de São Paulo, Departamento de Doenças Infecciosas e Parasitárias, Laboratório de Parasitologia Médica (LIM-46), São Paulo, SP, Brazil
| | - Joyce Vanessa Fonseca
- Universidade de São Paulo Instituto de Medicina Tropical de São Paulo, Departamento de Doenças Infecciosas e Parasitárias, Laboratório de Investigação Médica em Protozoologia, Bacteriologia e Resistência Antimicrobiana (LIM-49), São Paulo, SP, Brazil
| | - Roberta Cristina Martins
- Universidade de São Paulo Instituto de Medicina Tropical de São Paulo, Departamento de Doenças Infecciosas e Parasitárias, Laboratório de Parasitologia Médica (LIM-46), São Paulo, SP, Brazil
| | - Gabriele Alves Souza
- Universidade de São Paulo Faculdade de Medicina da Universidade de São Paulo, Departamento de Patologia, Laboratório de Neurociência (LIM-01), São Paulo, SP, Brazil
| | - Gabriela Benicio
- Universidade de São Paulo Faculdade de Medicina da Universidade de São Paulo, Departamento de Patologia, Laboratório de Neurociência (LIM-01), São Paulo, SP, Brazil
| | - Isabella Mirandez Sabbag
- Universidade de São Paulo Faculdade de Medicina da Universidade de São Paulo, Departamento de Patologia, Laboratório de Neurociência (LIM-01), São Paulo, SP, Brazil
| | - Esther Alves de Souza
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
- Pesquisa e Desenvolvimento Efeom Nutrição S/A, São Paulo, SP, Brazil
| | - José Pinhata Otoch
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
- Pesquisa e Desenvolvimento Efeom Nutrição S/A, São Paulo, SP, Brazil
- Hospital Universitário da Universidade de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - Ana Flávia Marçal Pessoa
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
- Universidade de São Paulo Instituto de Medicina Tropical de São Paulo, Departamento de Doenças Infecciosas e Parasitárias, Laboratório de Parasitologia Médica (LIM-46), São Paulo, SP, Brazil
- Instituto Botânio, São Paulo, Brazil
| |
Collapse
|
17
|
Liu J, Dai Y, Yang W, Chen ZY. Role of Mushroom Polysaccharides in Modulation of GI Homeostasis and Protection of GI Barrier. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6416-6441. [PMID: 40063730 PMCID: PMC11926878 DOI: 10.1021/acs.jafc.5c00745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/25/2025] [Accepted: 02/28/2025] [Indexed: 03/20/2025]
Abstract
Edible and medicinal mushroom polysaccharides (EMMPs) have been widely studied for their various biological activities. It has been shown that EMMPs could modulate microbiota in the large intestine and improve intestinal health. However, the role of EMMPs in protecting the gastric barrier, regulating gastric microbiota, and improving gastric health cannot be ignored. Hence, this review will elucidate the effect of EMMPs on gastric and intestinal barriers, with emphasis on the interaction of EMMPs with microbiota in maintaining overall gastrointestinal health. Additionally, this review highlights the gastroprotective effects and underlying mechanisms of EMMPs against gastric mucosa injury, gastritis, gastric ulcer, and gastric cancer. Furthermore, the effects of EMMPs on intestinal diseases, including inflammatory bowel disease, colorectal cancer, and intestinal infection, are also summarized. This review will also discuss the future perspective and challenges in the use of EMMPs as a dietary supplement or a nutraceutical in preventing and treating gastrointestinal diseases.
Collapse
Affiliation(s)
- Jianhui Liu
- Collaborative
Innovation Center for Modern Grain Circulation and Safety, Jiangsu
Province Engineering Research Center of Edible Fungus Preservation
and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
- School
of Life Sciences, The Chinese University
of Hong Kong, Shatin, NT, Hong Kong 999077, China
| | - Yi Dai
- Collaborative
Innovation Center for Modern Grain Circulation and Safety, Jiangsu
Province Engineering Research Center of Edible Fungus Preservation
and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Wenjian Yang
- Collaborative
Innovation Center for Modern Grain Circulation and Safety, Jiangsu
Province Engineering Research Center of Edible Fungus Preservation
and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Zhen-Yu Chen
- School
of Life Sciences, The Chinese University
of Hong Kong, Shatin, NT, Hong Kong 999077, China
| |
Collapse
|
18
|
Tang MH, Ligthart I, Varga S, Lebeer S, van Overveld FJ, Rijkers GT. Mutual Interactions Between Microbiota and the Human Immune System During the First 1000 Days of Life. BIOLOGY 2025; 14:299. [PMID: 40136555 PMCID: PMC11940030 DOI: 10.3390/biology14030299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/25/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025]
Abstract
The development of the human immune system starts during the fetal period in a largely, but probably not completely, sterile environment. During and after birth, the immune system is exposed to an increasingly complex microbiota. The first microbiota encountered during passage through the birth canal colonize the infant gut and induce the tolerance of the immune system. Transplacentally derived maternal IgG as well as IgA from breast milk protect the infant from infections during the first 100 days, during which the immune system further develops and immunological memory is formed. The Weaning and introduction of solid food expose the immune system to novel (food) antigens and allow for other microbiota to colonize. The cells and molecules involved in the mutual and intricate interactions between microbiota and the developing immune system are now beginning to be recognized. These include bacterial components such as polysaccharide A from Bacteroides fragilis, as well as bacterial metabolites such as the short-chain fatty acid butyrate, indole-3-aldehyde, and indole-3-propionic acid. All these, and probably more, bacterial metabolites have specific immunoregulatory functions which shape the development of the human immune system during the first 1000 days of life.
Collapse
Affiliation(s)
- Muy Heang Tang
- Department of Science and Engineering, University College Roosevelt, 4331 CB Middelburg, The Netherlands; (M.H.T.); (I.L.); (S.V.); (F.J.v.O.)
| | - Ishbel Ligthart
- Department of Science and Engineering, University College Roosevelt, 4331 CB Middelburg, The Netherlands; (M.H.T.); (I.L.); (S.V.); (F.J.v.O.)
| | - Samuel Varga
- Department of Science and Engineering, University College Roosevelt, 4331 CB Middelburg, The Netherlands; (M.H.T.); (I.L.); (S.V.); (F.J.v.O.)
| | - Sarah Lebeer
- Lab of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, 2020 Antwerpen, Belgium;
| | - Frans J. van Overveld
- Department of Science and Engineering, University College Roosevelt, 4331 CB Middelburg, The Netherlands; (M.H.T.); (I.L.); (S.V.); (F.J.v.O.)
| | - Ger T. Rijkers
- Department of Science and Engineering, University College Roosevelt, 4331 CB Middelburg, The Netherlands; (M.H.T.); (I.L.); (S.V.); (F.J.v.O.)
| |
Collapse
|
19
|
Liu MC, Shu YA, Wang YC, Tseng HY, Li MJ, Yu YT, Cheng HC, Tsai PJ, Yang YJ. Faecalibacterium prausnitzii Colonization Attenuates Gut Inflammation and Epithelial Damage in a DSS-Induced Colitis Mice Model. Mediators Inflamm 2025; 2025:7280675. [PMID: 40224484 PMCID: PMC11986197 DOI: 10.1155/mi/7280675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 02/04/2025] [Indexed: 04/15/2025] Open
Abstract
Background: Reduction of Faecalibacterium prausnitzii abundance is related to inflammatory bowel diseases (IBDs), and supplement of it exists protective effects. Aim: This study aimed to establish a F. prausnitzii-colonized mouse model and investigate that the presence of F. prausnitzii in the gut can ameliorate the severity of dextran sulfate sodium (DSS)-induced colitis. Methods: A F. prausnitzii (ATCC 27768) strain was maintained on the PS-BHI agar plates and manipulated in a strictly anaerobic chamber. A F. prausnitzii-colonized C57BL/6 mice model was tested by a rectal enema with 1 × 109 bacteria/day for 3 days. The 5% DSS was added to drinking water for 3 days to induce colitis and diarrhea in experimental mice. The clinical, cytological, and histological severities were compared between groups. Results: The F. prausnitzii-colonized mice model was successfully established via rectal enema with the property of transfer to offspring. DSS treatment altered gut microbiota and significantly attenuated the abundance of F. prausnitzii in colonized mice. Mice with F. prausnitzii colonization had significantly improved weight loss, anal bleeding, stool consistency, cecum weight, colon length, and serum amyloid A (SAA) level than those without after DSS treatment. Furthermore, the F. prausnitzii-colonized mice significantly reduced the transcription levels of TNF-α, INF-γ and IL-18, and epithelial damage and PMN infiltration in the lamina propria and had better preservation of goblet cells than the control group. Conclusion: We have successfully established a mouse model colonized with F. prausnitzii via rectal enema administration and showed colonization of F. prausnitzii in the gut has a protective effect against DSS-induced colitis.
Collapse
Affiliation(s)
- Meng-Chuan Liu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-An Shu
- Department of Medical Laboratory Science and Biotechnology and Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Chin Wang
- Department of Medical Laboratory Science and Biotechnology and Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Hsiu-Ying Tseng
- Department of Medical Laboratory Science and Biotechnology and Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Meng-Jia Li
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Ting Yu
- Department of Pathology, Chung Shan Medical University Hospital, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hsiu-Chi Cheng
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Jane Tsai
- Department of Medical Laboratory Science and Biotechnology and Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yao-Jong Yang
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
20
|
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
|
21
|
Huang H, Liu S, Peng Z, Wang B, Zhan S, Huang S, Li W, Liu D, Yang X, Zhu Y, Xiao W. Comparative effects of different sugar substitutes: Mogroside V, stevioside, sucralose, and erythritol on intestinal health in a type 2 diabetes mellitus mouse. Food Funct 2025; 16:2108-2123. [PMID: 39969196 DOI: 10.1039/d4fo04446k] [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: 02/20/2025]
Abstract
Intestinal health disorders significantly contribute to the development of type 2 diabetes mellitus (T2DM). Sugar substitutes such as mogroside V (MOG), stevioside (ST), sucralose (TGS), and erythritol (ERT), are increasingly used in T2DM management as alternatives to sucrose (SUC). However, their effects on intestinal health in T2DM have not been fully compared. In the present study, we established a T2DM mouse model using a high-fat diet and streptozotocin injection. These mice were treated with equal doses of SUC, MOG, ST, TGS, or ERT for 4 weeks to evaluate the effects of these sugar substitutes on intestinal health in T2DM. T2DM mice exhibited increased intestinal permeability, reduced goblet cell numbers, elevated pro-inflammatory cytokine levels, and alterations in both gut microbiota and metabolite composition. After 4 weeks of treatment, MOG showed the most significant benefits. MOG activates the PI3K/AKT pathway, enhancing the expression of tight junction proteins, which improves intestinal barrier function and reduces permeability. This is accompanied by NF-κB inhibition, leading to reduced pro-inflammatory cytokine production and increased mucus secretion. These changes help maintain healthy gut microbiota and metabolites, preventing pathogenic bacteria from entering the bloodstream. ST downregulates NF-κB to alleviate intestinal inflammation and improves gut microbiota and metabolic homeostasis in T2DM. ERT has less beneficial effects. TGS and SUC reduce intestinal inflammation and have a better effect on the duodenum. However, TGS has a negative effect on the colon microbiota and metabolites, whereas SUC has a negative effect on the colon microbiota alone. MOG improved intestinal health in T2DM by modulating the PI3K/AKT and NF-κB pathways, whereas ST primarily modulated NF-κB to alleviate intestinal inflammation. Both treatments were effective, with MOG showing the best performance. Therefore, MOG can be considered a viable alternative to SUC for T2DM management.
Collapse
Affiliation(s)
- Huaxue Huang
- School of Pharmacy, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China.
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China.
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Sha Liu
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China.
| | - Zhi Peng
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Bin Wang
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China.
| | - Shuang Zhan
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Sirui Huang
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Wei Li
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Dai Liu
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Xiulian Yang
- Hunan Huacheng Biological Resources Co. Ltd, Changsha, Hunan, 410000, China
- Hunan Natural Sweetener Engineering Technology Research Center, Changsha, Hunan, 410000, China
| | - Yizhun Zhu
- School of Pharmacy, Macau University of Science and Technology, Taipa, Macao SAR, 999078, China.
| | - Wenjun Xiao
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha, Hunan 410128, China.
| |
Collapse
|
22
|
Zhao Y, Zeng Z, Zheng W, Zhang Z, Zhang H, Luo Y, Zhao K, Ding Y, Lu W, Hao F, Huang Y, Shen L. Cow Placenta Peptides Ameliorate D-Galactose-Induced Intestinal Barrier Damage by Regulating TLR/NF-κB Pathway. Vet Sci 2025; 12:229. [PMID: 40266951 PMCID: PMC11945863 DOI: 10.3390/vetsci12030229] [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: 01/18/2025] [Revised: 02/19/2025] [Accepted: 02/26/2025] [Indexed: 04/25/2025] Open
Abstract
This study investigated the protective effects and mechanisms of cow placenta peptides (CPP) on intestinal barrier damage in aging model mice. Forty-eight male ICR mice were assigned to four groups: a control group (N), an aging model group (M), a CPP treatment group (T), and a vitamin C treatment group (P). Groups T and P received oral administration of CPP (2000 mg/kg/day) and vitamin C (100 mg/kg/day), respectively, while groups M, T, and P were subjected to intraperitoneal injections of D-galactose (D-gal) (300 mg/kg/day). Group N received an equivalent volume of normal saline via intraperitoneal injection. Treatments were administered once daily for 8 weeks. The results demonstrated that CPP significantly alleviated D-galactose-induced intestinal structural damage, increasing the villus height-to-crypt depth ratio and reducing serum diamine oxidase (DAO) and lipopolysaccharide (LPS) levels. CPP notably alleviated intestinal oxidative stress and inflammation, restored tight junction expression, and enhanced intestinal barrier integrity. Transcriptome sequencing identified 1396 DEGs associated with CPP's effects, highlighting TLR4, IL-1β, and Mmp9 as core regulatory genes through protein-protein interaction network analysis. Kyoto Encyclopedia of Genes and Genomes and Gene Ontology enrichment analyses implicated the TLR4/NF-κB signaling pathway, which was further validated. Western blotting confirmed that CPP significantly down-regulated TLR4, IKKβ, and p-NF-κB p65 protein expression in the intestines of aging mice. In conclusion, CPP effectively alleviates D-gal-induced intestinal barrier damage in aging mice by enhancing antioxidant defense and inhibiting the TLR4/NF-κB signaling pathway, thereby diminishing inflammation and protecting intestinal barrier integrity.
Collapse
Affiliation(s)
- Yuquan Zhao
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.Z.); (Z.Z.); (W.Z.); (Z.Z.); (H.Z.); (Y.L.); (K.Z.); (Y.D.)
| | - Zhi Zeng
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.Z.); (Z.Z.); (W.Z.); (Z.Z.); (H.Z.); (Y.L.); (K.Z.); (Y.D.)
| | - Weijian Zheng
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.Z.); (Z.Z.); (W.Z.); (Z.Z.); (H.Z.); (Y.L.); (K.Z.); (Y.D.)
| | - Zeru Zhang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.Z.); (Z.Z.); (W.Z.); (Z.Z.); (H.Z.); (Y.L.); (K.Z.); (Y.D.)
| | - Hanwen Zhang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.Z.); (Z.Z.); (W.Z.); (Z.Z.); (H.Z.); (Y.L.); (K.Z.); (Y.D.)
| | - Yuxin Luo
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.Z.); (Z.Z.); (W.Z.); (Z.Z.); (H.Z.); (Y.L.); (K.Z.); (Y.D.)
| | - Kunshan Zhao
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.Z.); (Z.Z.); (W.Z.); (Z.Z.); (H.Z.); (Y.L.); (K.Z.); (Y.D.)
| | - Yuyan Ding
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.Z.); (Z.Z.); (W.Z.); (Z.Z.); (H.Z.); (Y.L.); (K.Z.); (Y.D.)
| | - Wei Lu
- Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (W.L.); (F.H.)
| | - Fuxing Hao
- Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (W.L.); (F.H.)
| | - Yixin Huang
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.Z.); (Z.Z.); (W.Z.); (Z.Z.); (H.Z.); (Y.L.); (K.Z.); (Y.D.)
| | - Liuhong Shen
- The Key Laboratory of Animal Disease and Human Health of Sichuan Province, The Medical Research Center for Cow Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.Z.); (Z.Z.); (W.Z.); (Z.Z.); (H.Z.); (Y.L.); (K.Z.); (Y.D.)
| |
Collapse
|
23
|
Jiang J, Xie H, Cao S, Xu X, Zhou J, Liu Q, Ding C, Liu M. Post-stroke depression: exploring gut microbiota-mediated barrier dysfunction through immune regulation. Front Immunol 2025; 16:1547365. [PMID: 40098959 PMCID: PMC11911333 DOI: 10.3389/fimmu.2025.1547365] [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: 12/18/2024] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Post-stroke depression (PSD) is one of the most common and devastating neuropsychiatric complications in stroke patients, affecting more than one-third of survivors of ischemic stroke (IS). Despite its high incidence, PSD is often overlooked or undertreated in clinical practice, and effective preventive measures and therapeutic interventions remain limited. Although the exact mechanisms of PSD are not fully understood, emerging evidence suggests that the gut microbiota plays a key role in regulating gut-brain communication. This has sparked great interest in the relationship between the microbiota-gut-brain axis (MGBA) and PSD, especially in the context of cerebral ischemia. In addition to the gut microbiota, another important factor is the gut barrier, which acts as a frontline sensor distinguishing between beneficial and harmful microbes, regulating inflammatory responses and immunomodulation. Based on this, this paper proposes a new approach, the microbiota-immune-barrier axis, which is not only closely related to the pathophysiology of IS but may also play a critical role in the occurrence and progression of PSD. This review aims to systematically analyze how the gut microbiota affects the integrity and function of the barrier after IS through inflammatory responses and immunomodulation, leading to the production or exacerbation of depressive symptoms in the context of cerebral ischemia. In addition, we will explore existing technologies that can assess the MGBA and potential therapeutic strategies for PSD, with the hope of providing new insights for future research and clinical interventions.
Collapse
Affiliation(s)
- Jia Jiang
- The Second Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, China
| | - Haihua Xie
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Sihui Cao
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Xuan Xu
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Jingying Zhou
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Qianyan Liu
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Changsong Ding
- School of Information Science and Engineering, Hunan University of Chinese Medicine, Changsha, China
| | - Mi Liu
- School of Acupuncture & Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
24
|
Tao W, Zhang Y, Wang B, Nie S, Fang L, Xiao J, Wu Y. Advances in molecular mechanisms and therapeutic strategies for central nervous system diseases based on gut microbiota imbalance. J Adv Res 2025; 69:261-278. [PMID: 38579985 PMCID: PMC11954836 DOI: 10.1016/j.jare.2024.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/12/2024] [Accepted: 03/29/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUD Central nervous system (CNS) diseases pose a serious threat to human health, but the regulatory mechanisms and therapeutic strategies of CNS diseases need to be further explored. It has been demonstrated that the gut microbiota (GM) is closely related to CNS disease. GM structure disorders, abnormal microbial metabolites, intestinal barrier destruction and elevated inflammation exist in patients with CNS diseases and promote the development of CNS diseases. More importantly, GM remodeling alleviates CNS pathology to some extent. AIM OF REVIEW Here, we have summarized the regulatory mechanism of the GM in CNS diseases and the potential treatment strategies for CNS repair based on GM regulation, aiming to provide safer and more effective strategies for CNS repair from the perspective of GM regulation. KEY SCIENTIFIC CONCEPTS OF REVIEW The abundance and composition of GM is closely associated with the CNS diseases. On the basis of in-depth analysis of GM changes in mice with CNS disease, as well as the changes in its metabolites, therapeutic strategies, such as probiotics, prebiotics, and FMT, may be used to regulate GM balance and affect its microbial metabolites, thereby promoting the recovery of CNS diseases.
Collapse
Affiliation(s)
- Wei Tao
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Yanren Zhang
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Bingbin Wang
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Saiqun Nie
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Li Fang
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China
| | - Jian Xiao
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Yanqing Wu
- The Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
25
|
Di Mattia M, Sallese M, Lopetuso LR. The interplay between gut microbiota and the unfolded protein response: Implications for intestinal homeostasis preservation and dysbiosis-related diseases. Microb Pathog 2025; 200:107279. [PMID: 39761770 DOI: 10.1016/j.micpath.2025.107279] [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: 03/08/2024] [Revised: 11/28/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025]
Abstract
The unfolded protein response (UPR) is a complex intracellular signal transduction system that orchestrates the cellular response during Endoplasmic Reticulum (ER) stress conditions to reestablish cellular proteostasis. If, on one side, prolonged ER stress conditions can lead to programmed cell death and autophagy as a cytoprotective mechanism, on the other, unresolved ER stress and improper UPR activation represent a perilous condition able to trigger or exacerbate inflammatory responses. Notably, intestinal and immune cells experience ER stress physiologically due to their high protein secretory rate. Indeed, there is evidence of UPR's involvement in both physiological and pathological intestinal conditions, while less is known about its bidirectional interaction with gut microbiota. However, gut microbes and their metabolites can influence ER stress and UPR pathways, and, in turn, ER stress conditions can shape gut microbiota composition, with important implications for overall intestinal health. Thus, targeting UPR components is an intriguing strategy for treating ER stress-linked dysbiosis and diseases, particularly intestinal inflammation.
Collapse
Affiliation(s)
- Miriam Di Mattia
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy.
| | - Michele Sallese
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Loris Riccardo Lopetuso
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell'Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| |
Collapse
|
26
|
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
|
27
|
Zhou C, Zou Y, Huang H, Zhao F, Fan X, Bai L, Zhang X, Ye K. Virulence expression difference to intestinal cells of different pathogenic Listeria monocytogenes contaminating sausages after simulated digestive tract. Int J Food Microbiol 2025; 430:111067. [PMID: 39813952 DOI: 10.1016/j.ijfoodmicro.2025.111067] [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: 11/12/2024] [Revised: 01/08/2025] [Accepted: 01/12/2025] [Indexed: 01/18/2025]
Abstract
This study investigated the difference in survival among Listeria monocytogenes (LM) 10403S (highly pathogenic strain) and M7 (low pathogenic strain) in sausage under a simulated digestive environment, and established intestinal organoids and macrophages co-culture model to further explore the virulence expression difference to intestinal cells between LM 10403S and M7 after in vitro gastrointestinal digestion. Results showed that, compared with LM M7, LM 10403S exhibited a high survival rate during in vitro digestion, which may be due to the increased expression of stress response-related genes. In addition, the expression of virulence genes in LM 10403S was significantly higher than in LM M7 under the gastrointestinal environment. Furthermore, in the intestinal organoids and macrophages co-culture model infected by LM 10403S and M7 after in vitro gastrointestinal digestion, results showed that, compared with the LM M7 group, the LM 10403S group had significantly lower budding rate and significantly higher mortality of organoids. Also, the significantly increased LDH release and inflammatory factors (TNF-α and IL-1β) in the LM 10403S group were observed, and the main virulence genes (iap, inlA, inlB, actA, hly, plcA, and plcB) of 10403S were significantly highly expressed than LM M7 during the cell infection. These results reflected that the reason for the different pathogenicity between LM 10403S and M7 may be due to the high tolerance and the expression of virulence genes than LM M7 during gastrointestinal digestion and cell infection, which would be expected to provide a better understanding of the infection mechanisms among different pathogenic strains of L. monocytogenes in food.
Collapse
Affiliation(s)
- Cong Zhou
- China National Center for Food Safety Risk Assessment Key Laboratory of Food Safety Risk Assessment, Beijing, PR China; State Key Laboratory of Meat Quality Control and Cultured Meat Development, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Yafang Zou
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Haorui Huang
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Fanwen Zhao
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Xia Fan
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Li Bai
- China National Center for Food Safety Risk Assessment Key Laboratory of Food Safety Risk Assessment, Beijing, PR China
| | - Xinhao Zhang
- China National Center for Food Safety Risk Assessment Key Laboratory of Food Safety Risk Assessment, Beijing, PR China
| | - Keping Ye
- State Key Laboratory of Meat Quality Control and Cultured Meat Development, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, PR China.
| |
Collapse
|
28
|
Di Mattia M, Sallese M, Lopetuso LR. Unfolded protein response: An essential element of intestinal homeostasis and a potential therapeutic target for inflammatory bowel disease. Acta Physiol (Oxf) 2025; 241:e14284. [PMID: 39822064 DOI: 10.1111/apha.14284] [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: 06/11/2024] [Revised: 12/23/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025]
Abstract
Different physiological and pathological situations can produce alterations in the cell's endoplasmic reticulum (ER), leading to a condition known as ER stress, which can trigger an intricate intracellular signal transduction system known as the unfolded protein response (UPR). UPR is primarily tailored to restore proteostasis and ER equilibrium; otherwise, if ER stress persists, it can cause programmed cell death as a cytoprotective mechanism and drive inflammatory processes. Therefore, since intestinal cells strongly rely on UPR for their biological functions and unbalanced UPR has been linked to inflammatory, metabolic, and immune disorders, here we discussed the role of the UPR within the intestinal tract, focusing on the UPR contribution to inflammatory bowel disease development. Importantly, we also highlighted the promising potential of UPR components as therapeutic targets for intestinal inflammatory diseases.
Collapse
Affiliation(s)
- Miriam Di Mattia
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Michele Sallese
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Loris Riccardo Lopetuso
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell'Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| |
Collapse
|
29
|
Zhang L, Li J, Wan Q, Bu C, Jin W, Yuan F, Zhou W. Intestinal stem cell-derived extracellular vesicles ameliorate necrotizing enterocolitis injury. Mol Cell Probes 2025; 79:101997. [PMID: 39645054 DOI: 10.1016/j.mcp.2024.101997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 12/09/2024]
Abstract
The therapeutic potential of intestinal stem cell-derived extracellular vesicles (ISCs-EVs) in necrotizing enterocolitis (NEC) remains largely unexplored. This research aims to investigate the therapeutic effects of ISCs-EVs on NEC. Lgr5-positive ISCs were screened from the small intestine of mice by flow cytometry, and ISCs-EVs were isolated by density gradient centrifugation. Subsequently, ISCs-EVs were identified through transmission electron microscopy, nanoparticle tracking analysis, and western blotting. Subsequently, we evaluated the efficacy of ISCs-EVs in a mouse model of NEC and found that they enhanced survival (more than 20 %), reduced intestinal damage (restore the number of intestinal crypts and decrease the expression of MPO and cleaved-caspase 3 in intestinal tissues), promoted angiogenesis (the mRNA expression of VEGF was increased by approximately 35 %), and mitigated inflammation (decreased the level of MUC1, p-NF-κB, IL-6 and TNF-α). Furthermore, in vitro assessments demonstrated that ISCs-EVs reduced apoptosis (P < 0.01) and stimulated proliferation (P < 0.05) of IEC-6 cells, while enhancing mucin secretion in LS174T cells. In summary, our study provides a comprehensive assessment of the therapeutic effects of ISCs-EVs on NEC, using both animal and cell models. This highlights their potential for use in NEC treatment.
Collapse
Affiliation(s)
- Le Zhang
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University, Wuxi Children's Hospital, 214023, Wuxi, Jiangsu, China; Key Laboratory of Birth Defects, Children's Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jiahong Li
- Department of Pediatric Laboratory, Affiliated Children's Hospital of Jiangnan University, Wuxi Children's Hospital, 214023, Wuxi, Jiangsu, China
| | - Qiwen Wan
- Department of Pediatric Surgery, Affiliated Children's Hospital of Jiangnan University, Wuxi Children's Hospital, 214023, Wuxi, Jiangsu, China
| | - Chaozhi Bu
- State Key Laboratory of Reproductive Medicine, Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, 214002, Wuxi, Jiangsu, China
| | - Weilai Jin
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University, Wuxi Children's Hospital, 214023, Wuxi, Jiangsu, China.
| | - Fuqiang Yuan
- Department of Neonatology, Affiliated Children's Hospital of Jiangnan University, Wuxi Children's Hospital, 214023, Wuxi, Jiangsu, China.
| | - Wenhao Zhou
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China; Key Laboratory of Birth Defects, Children's Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
30
|
Zhang Z, Zeng Z, Wang L, Xiong B, Zheng B, Zhang Y, Pan L. Dictyophora indusiata polysaccharide attenuated LPS-induced intestinal inflammation of mice via the TLR4/JNK signaling pathway. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:974-981. [PMID: 39268777 DOI: 10.1002/jsfa.13888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/06/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Dictyophora indusiata polysaccharide is an important bioactive component of D. indusiata, playing an important role in alleviating inflammation. The present study aimed to investigate the anti-inflammatory effect and mechanism of D. indusiata polysaccharide on lipopolysaccharide (LPS)-induced intestinal inflammation in mice. RESULTS Our results indicated that D. indusiata polysaccharide ameliorated intestinal inflammation of mice by increasing the body weight, the number of goblet cells and decreasing inflammatory cell infiltration. In addition, D. indusiata polysaccharide significantly up-regulated expression of ZO-1, Occuldin mRNA, which were 2.55-fold and 2.28-fold higher than the LPS group, respectively. In particular, D. indusiata polysaccharide effectively inhibited the Toll-like receptor 4 (TLR4)/ c-Jun NH2-terminal kinase (JNK) signalling pathway which was 0.34-fold and 0.49-fold of gene expression and 0.41-fold and 0.39-fold of protein expression in the LPS group, respectively. CONCLUSION The results of the present study suggested that D. indusiata polysaccharide exerted anti-inflammatory and intestinal protective effects by inhibiting the TLR4/JNK signaling pathway, which will provide a basis for the potential value of D. indusiata polysaccharide as prebiotics in food applications. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Zihao Zhang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Zhikun Zeng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Integrated Scientific Research Base of Edible Fungi Processing and Comprehensive Utilization Technology, Ministry of Agriculture and Rural Affairs, Fuzhou, China
| | - Lin Wang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Integrated Scientific Research Base of Edible Fungi Processing and Comprehensive Utilization Technology, Ministry of Agriculture and Rural Affairs, Fuzhou, China
| | - Bin Xiong
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Baodong Zheng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Integrated Scientific Research Base of Edible Fungi Processing and Comprehensive Utilization Technology, Ministry of Agriculture and Rural Affairs, Fuzhou, China
| | - Yi Zhang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Integrated Scientific Research Base of Edible Fungi Processing and Comprehensive Utilization Technology, Ministry of Agriculture and Rural Affairs, Fuzhou, China
| | - Lei Pan
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Integrated Scientific Research Base of Edible Fungi Processing and Comprehensive Utilization Technology, Ministry of Agriculture and Rural Affairs, Fuzhou, China
| |
Collapse
|
31
|
Odu-Onikosi SG, Momoh TA, Abarra ST, Wood NE, Amulejoye FD, Emery M, Harper GM, Eynon B, Kuri V, Kühlwein H, Merrifield DL. Impact of Autolysed Brewer's Yeast and Soluble Dried Yeast Extract on Growth Performance and Mucosal Health of Atlantic Salmon ( Salmo salar) Parr. Animals (Basel) 2025; 15:323. [PMID: 39943093 PMCID: PMC11816051 DOI: 10.3390/ani15030323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Yeast-based feed additives have emerged as promising functional feed additives (FFAs) to promote sustainable aquaculture development through enhanced gut health and immune modulation in fish. The present study evaluated the impact of autolysed brewer's yeast (ABY) and soluble dried yeast extract (SDYE) in improving the intestinal and skin histology, immune response, and intestinal microbiome of Atlantic salmon parr (Salmo salar) over a 9-week feeding trial. Three experimental diets were produced: a control diet, a diet supplemented with ABY at 2.5 g/kg, and a diet supplemented with SDYE at 2.5 g/kg. These diets were administered to triplicate tanks of Atlantic salmon. The yeast-supplemented diets, especially ABY, improved intestinal mucosal fold length, lamina propria width, microvilli density, and intestinal goblet cell counts, as well as skin goblet cell counts. The yeast additives had no detrimental effects on the fish haematology, with no significant differences in haemoglobin concentration, red blood cell counts, and white blood cell counts among the treatment groups. Gene expression analysis revealed upregulation of il-1β and muc-2 in fish fed the ABY diet, indicating enhanced immune function and potentially mucosal protection. Intestinal microbiota analysis revealed Firmicutes as the most dominant phylum in all groups, followed by Actinobacteriota. Distinct bacterial community shifts were observed between the treatment groups, with a significant increase in the relative abundance of taxa such as Staphylococcus in yeast-supplemented diets and a significant decrease in Streptococcus and Weissella. Collectively, these findings suggest that the yeast additives, especially ABY, enhance gut health and immune function without compromising growth performance.
Collapse
Affiliation(s)
- Sheu G. Odu-Onikosi
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (T.A.M.); (S.T.A.); (N.E.W.); (M.E.); (G.M.H.); (B.E.); (V.K.)
- Department of Fisheries and Aquaculture, Lagos State University of Science and Technology, Ikorodu 104101, Nigeria
| | - Taofik A. Momoh
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (T.A.M.); (S.T.A.); (N.E.W.); (M.E.); (G.M.H.); (B.E.); (V.K.)
| | - Sherilyn T. Abarra
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (T.A.M.); (S.T.A.); (N.E.W.); (M.E.); (G.M.H.); (B.E.); (V.K.)
| | - Noah E. Wood
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (T.A.M.); (S.T.A.); (N.E.W.); (M.E.); (G.M.H.); (B.E.); (V.K.)
| | - Folasade D. Amulejoye
- Department of Fisheries and Aquaculture Technology, Olusegun Agagu University of Science and Technology, P.M.B. 353, Okitipupa 350105, Nigeria;
| | - Matthew Emery
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (T.A.M.); (S.T.A.); (N.E.W.); (M.E.); (G.M.H.); (B.E.); (V.K.)
| | - Glenn M. Harper
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (T.A.M.); (S.T.A.); (N.E.W.); (M.E.); (G.M.H.); (B.E.); (V.K.)
| | - Benjamin Eynon
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (T.A.M.); (S.T.A.); (N.E.W.); (M.E.); (G.M.H.); (B.E.); (V.K.)
| | - Victor Kuri
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (T.A.M.); (S.T.A.); (N.E.W.); (M.E.); (G.M.H.); (B.E.); (V.K.)
| | | | - Daniel L. Merrifield
- Fish Health and Nutrition Research Group, School of Biological and Marine Sciences, University of Plymouth, Plymouth PL4 8AA, UK; (T.A.M.); (S.T.A.); (N.E.W.); (M.E.); (G.M.H.); (B.E.); (V.K.)
| |
Collapse
|
32
|
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
|
33
|
Thonapan N, Wongdee K, Aksornthong S, Teerapornpuntakit J, Tiyasatkulkovit W, Panupinthu N, Charoenphandhu N. Long-term excessive salt consumption alters villous and crypt morphology and the protein expression of uroguanylin, TRPV6 and PMCA1b in the rat small intestine. PLoS One 2025; 20:e0317415. [PMID: 39820616 PMCID: PMC11737712 DOI: 10.1371/journal.pone.0317415] [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: 07/31/2024] [Accepted: 12/27/2024] [Indexed: 01/19/2025] Open
Abstract
Although long-term high dietary sodium consumption often aggravates hypertension and bone loss, sodium in the intestinal lumen has been known to promote absorption of nutrients and other ions, e.g., glucose and calcium. However, whether high-salt diet (HSD) altered mucosal morphology, villous cell turnover and calcium transporter expression remained elusive. Herein, rats were treated with HSD containing 8% wt/wt NaCl for up to 5 months. HSD rats exhibited a marked increase in sodium intake with high fecal and urinary sodium excretion, as compared to the control group treated with normal diet. Intestinal histomorphometry revealed increasing of crypt depth and villous height in 3- and 4-month HSD groups, respectively, consistent with larger mucosal-to-serosal amplification ratio that reflected an increased surface area for nutrient absorption. The signals of Ki-67-positive cells was enhanced in the crypts as visualized by multiphoton fluorescence microscopy, whereas the TUNEL-positive cells were decreased in the villi of HSD, suggesting greater crypt cell proliferation and a reduction of villous cell apoptosis. Confocal microscopy showed higher expression of TRPV6 protein in the villous tip of HSD, while PMCA1 expression was increased in villous tip and crypt areas. The percentage of cells with highly expressed uroguanylin-an endogenous intestinal natriuretic peptide-was significantly higher in HSD group. In conclusion, HSD profoundly changed the intestinal morphology and turnover of epithelial cell, increased the expression of calcium transporters and uroguanylin. Our findings reflect pathophysiological adaptations in the intestine, which might be another target organ for drug discovery against HSD-induced osteopathy in the future.
Collapse
Affiliation(s)
- Natchayaporn Thonapan
- Graduate Program in Molecular Medicine, Faculty of Science, Mahidol University, Bangkok, Thailand
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Kannikar Wongdee
- Faculty of Allied Health Sciences, Burapha University, Saen Suk, Chonburi, Thailand
| | - Sirion Aksornthong
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Jarinthorn Teerapornpuntakit
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
- Physiology Division, Preclinical Sciences, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| | - Wacharaporn Tiyasatkulkovit
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
- Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Nattapon Panupinthu
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Narattaphol Charoenphandhu
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
- The Academy of Science, The Royal Society of Thailand, Dusit, Bangkok, Thailand
| |
Collapse
|
34
|
Du M, Liu X, Ji X, Wang Y, Liu X, Zhao C, Jin E, Gu Y, Wang H, Zhang F. Berberine alleviates enterotoxigenic Escherichia coli-induced intestinal mucosal barrier function damage in a piglet model by modulation of the intestinal microbiome. Front Nutr 2025; 11:1494348. [PMID: 39877539 PMCID: PMC11772193 DOI: 10.3389/fnut.2024.1494348] [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: 09/10/2024] [Accepted: 12/13/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction Enterotoxic Escherichia coli (ETEC) is the main pathogen that causes diarrhea, especially in young children. This disease can lead to substantial morbidity and mortality and is a major global health concern. Managing ETEC infections is challenging owing to the increasing prevalence of antibiotic resistance. Berberine, categorized as a substance with similarities in "medicine and food," has been used in China for hundreds of years to treat gastrointestinal disorders and bacteria-induced diarrhea. This study investigated the preventive effect of dietary berberine on the intestinal mucosal barrier induced by ETEC and the microbial community within the intestines of weaned piglets. Methods Twenty-four piglets were randomly divided into four groups. Piglets were administered either a standard diet or a standard diet supplemented with berberine at concentrations of 0.05 and 0.1%. and orally administered ETEC or saline. Results Dietary supplementation with berberine reduced diamine oxidase, d-lactate, and endotoxin levels in piglets infected with ETEC (P < 0.05). Berberine increased jejunal villus height, villus/crypt ratio, mucosal thickness (P < 0.05), and goblet cell numbers in the villi and crypts (P < 0.05). Furthermore, berberine increased the optical density of mucin 2 and the mucin 2, P-glycoprotein, and CYP3A4 mRNA expression levels (P < 0.05). Berberine increased the expressions of zonula occludins-1 (ZO-1), zonula occludins-2 (ZO-2), Claudin-1, Occludin, and E-cadherin in the ileum (P < 0.05). Moreover, berberine increased the expression of BCL2, reduced intestinal epithelial cell apoptosis (P < 0.05) and decreased the expression of BAX and BAK in the duodenum and jejunum, as well as that of CASP3 and CASP9 in the duodenum and ileum (P < 0.05). Berberine decreased the expression of IL-1β, IL-6, IL-8, TNF-α, and IFN-γ (P < 0.05) and elevated total volatile fatty acids, acetic acid, propionic acid, valeric acid, and isovaleric acid concentrations (P < 0.05). Notably, berberine enhanced the abundance of beneficial bacteria including Enterococcus, Holdemanella, Weissella, Pediococcus, Muribaculum, Colidextribacter, Agathobacter, Roseburia, Clostridium, Fusicatenibacter, and Bifidobacterium. Simultaneously, the relative abundance of harmful and pathogenic bacteria, such as Prevotella, Paraprevotella, Corynebacterium, Catenisphaera, Streptococcus, Enterobacter, and Collinsella, decreased (P < 0.05). Discussion Berberine alleviated ETEC-induced intestinal mucosal barrier damage in weaned piglets models. This is associated with enhancement of the physical, chemical, and immune barrier functions of piglets by enhancing intestinal microbiota homeostasis.
Collapse
Affiliation(s)
- Min Du
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Xinran Liu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Xu Ji
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, China
| | - Yue Wang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Xiaodan Liu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Chunfang Zhao
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou, China
| | - Erhui Jin
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou, China
| | - Youfang Gu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou, China
| | - Hongyu Wang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Feng Zhang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou, China
| |
Collapse
|
35
|
Liu JQ, Hao WA, Liu YL, Yang D, Wang HL, Zhao L, Chen H, Li L, Jiang CL, Zhou X, Fu J, Li Z. The efficacy and active compounds of Chaihuang Qingyi Huoxue granule to Ameliorate intestinal mucosal barrier injury in rats with severe acute pancreatitis by suppressing the HMGB1/TLR4/NF-κB signaling pathway. Int Immunopharmacol 2025; 144:113632. [PMID: 39566389 DOI: 10.1016/j.intimp.2024.113632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 10/30/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024]
Abstract
Intestinal mucosal barrier injury represents a critical complication of severe acute pancreatitis (SAP) without effective treatment. This study investigated the efficacy, underlying mechanism, and responsible active compounds of the traditional Chinese medicinal prescription Chaihuang Qingyi Huoxue granule (CHQY) in treating SAP-induced intestinal mucosal barrier injury. SAP was established in Sprague-Dawley rats via intra-pancreaticobiliary duct infusion of sodium taurocholate, followed by oral CHQY administration (3.15 g/kg every 6 h for 12 and 24 h). Blood and tissues were harvested to assess the severity of pancreatitis, intestinal mucosal barrier integrity, and extent of inflammatory injury. Intestine-absorbing compounds were identified using ultra-high-performance liquid chromatography coupled with high-resolution mass spectrometry (UHPLC-HRMS). Our results showed that CHQY treatment effectively mitigated SAP-induced intestinal mucosal injury, as evidenced by improved intestinal epithelial structure, decreased serum levels of intestinal injury markers (d-lactic acid, diamine oxidase, I-FABP, and Zonulin), restored expression of the tight junction protein ZO-1, and reduced serum endotoxin levels. Furthermore, CHQY administration suppressed the expression of proinflammatory mediator HMGB1, its receptor TLR4, and downstream NF-κB signaling in the intestine, leading to downregulated intestinal IL-1β expression and reduced circulating TNF-α and IL-6. UHPLC-HRMS analysis identified 15 intestine-absorbing compounds in CHQY, of which paeoniflorin sulfite and chrysin-7-O-glucuronide independently inhibited TNF-α-induced tight junction loss in IEC-6 cells and mitigated intestinal mucosal barrier injury in SAP rats through suppressing NF-κB signaling. In summary, CHQY ameliorates SAP-induced intestinal mucosal barrier injury by downregulating the proinflammatory HMGB1/TLR4/NF-κB signaling, with efficacy partially attributed to its active compounds paeoniflorin sulfite and chrysin-7-O-glucuronide.
Collapse
Affiliation(s)
- Jian-Qin Liu
- Research Center of Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University Luzhou, 646000, China; College of Integrated Chinese and Western Medicine and the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Wei-An Hao
- Research Center of Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University Luzhou, 646000, China; College of Integrated Chinese and Western Medicine and the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Ya-Li Liu
- Research Center of Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University Luzhou, 646000, China; College of Integrated Chinese and Western Medicine and the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Dan Yang
- College of Integrated Chinese and Western Medicine and the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Hong-Lian Wang
- Research Center of Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University Luzhou, 646000, China; College of Integrated Chinese and Western Medicine and the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Long Zhao
- College of Integrated Chinese and Western Medicine and the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China; Department of Spleen & Stomach, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China; The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou city, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Lu zhou, 646000, China
| | - Hui Chen
- College of Integrated Chinese and Western Medicine and the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China; Department of Spleen & Stomach, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China; The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou city, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Lu zhou, 646000, China
| | - Li Li
- College of Integrated Chinese and Western Medicine and the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China; Department of Spleen & Stomach, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China; The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou city, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Lu zhou, 646000, China
| | - Chao-Li Jiang
- College of Integrated Chinese and Western Medicine and the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China; Department of Spleen & Stomach, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Xin Zhou
- Research Center of Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University Luzhou, 646000, China; College of Integrated Chinese and Western Medicine and the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China; Department of Spleen & Stomach, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China; The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou city, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Lu zhou, 646000, China.
| | - Juan Fu
- College of Integrated Chinese and Western Medicine and the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China; Department of Spleen & Stomach, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China.
| | - Zhi Li
- Research Center of Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University Luzhou, 646000, China; School of Integrated Traditional Chinese and Western Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, China; College of Integrated Chinese and Western Medicine and the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China; The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou city, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Lu zhou, 646000, China.
| |
Collapse
|
36
|
Saleh SMM, Abdel-Zaher S, Mohamed MS, Sayed AEDH. Microplastics induced ileum damage: Morphological and immunohistochemical study. Microsc Res Tech 2025; 88:251-269. [PMID: 39315562 DOI: 10.1002/jemt.24696] [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: 07/16/2024] [Revised: 08/08/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024]
Abstract
Microplastics (MPs) are small pieces of plastic that are widely distributed in the environment and accumulate within living organisms, so they are the most common types of pollutants at the present time. One of the most widespread types of MP in the environment is polyethylene (PE) MPs. There have been many published studies on the effect of PE MPs combined with other pollutants or chemicals such as benzoanthracene, emamectin benzoate, heavy metals and 4-nonylphenol, on some marine, amphibian, and mouse models. However, research has rarely been conducted on how single-use PE MPs affect the ileum of mammals. The current study is focused on the impact of PE MP exposure with different concentration (6, 60, 600 μg/mL PE/MPs) for 15 days, followed by 15 days of recovery on small intestine(ileum) of C57BL/6 murine model with precision and detail at the cell level by using different technique (histology, histochemistry, immunohistochemistry, and transmission electron microscope). Results demonstrated that the intestinal tissue exhibited nuclear pyknosis, villus deformation, shortness of villi, degeneration of lamina propria, hyperplasia of goblet cells, increase of goblet cells secretion, Alcian blue and Periodic acid-Schiff stain positivity of intact goblet cells, highly significance of P53 immunoreaction expression specially in high concentrations (600 μg/day of PE/MPs) and Ki-67 immunoreaction expression. RESEARCH HIGHLIGHTS: Different doses of microplastics (MPs) induced sever morphological alternations and clinical observations. MPs were deposits in cells and were observed in ultrastructure study. Recovery period able to ameliorate to the most extent the alternations caused by MPs administration.
Collapse
Affiliation(s)
- Shaimaa M M Saleh
- Department of Zoology, Faculty of Science, Assiut University, Assiut, Egypt
| | - Souzan Abdel-Zaher
- Department of Molecular Biology, Molecular Biology Research & Studies Institute, Assiut University, Assiut, Egypt
| | - Mahmoud S Mohamed
- Department of Zoology, Faculty of Science, Assiut University, Assiut, Egypt
| | - Alaa El-Din H Sayed
- Department of Zoology, Faculty of Science, Assiut University, Assiut, Egypt
- Department of Molecular Biology, Molecular Biology Research & Studies Institute, Assiut University, Assiut, Egypt
| |
Collapse
|
37
|
Nicolò S, Faggiani I, Errico C, D'Amico F, Parigi TL, Danese S, Ungaro F. Translational characterization of immune pathways in inflammatory bowel disease: insights for targeted treatments. Expert Rev Clin Immunol 2025; 21:55-72. [PMID: 39313992 DOI: 10.1080/1744666x.2024.2400300] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 08/30/2024] [Indexed: 09/25/2024]
Abstract
INTRODUCTION The pathogenesis of inflammatory bowel disease (IBD) involves the dysregulation of multiple inflammatory pathways. The understanding of these mechanisms allows their selective targeting for therapeutic purposes. The discovery of Tumor Necrosis Factor-alpha's (TNF-α) role in mucosal inflammation ushered an exciting new era of drug development which now comprises agents targeting multiple pro-inflammatory signaling pathways, integrins, and leukocyte trafficking regulators. AREA COVERED This review provides an overview of the main molecular players of IBD, their translation into therapeutic targets and the successful development of the advanced agents modulating them. We combine basic science with clinical trials data to present a critical review of both the successful and failed drug development programs. A PubMed literature search was conducted to delve into the available literature and clinical trials. EXPERT OPINION The treatment landscape for IBD has rapidly expanded, particularly with the development of biologics targeting TNF-α, integrins, and S1P modulators, as well as newer agents such as IL-12/IL-23 inhibitors and JAK inhibitors, offering robust efficacy and safety profiles. However, challenges persist in understanding and effectively treating difficult-to-treat IBD, highlighting the need for continued research to uncover novel therapeutic targets and optimize patient outcomes.
Collapse
Affiliation(s)
- Sabrina Nicolò
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Ilaria Faggiani
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Carmela Errico
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Ferdinando D'Amico
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Tommaso Lorenzo Parigi
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Silvio Danese
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| | - Federica Ungaro
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
- Faculty of Medicine, Università Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
38
|
Elkhaiat IA, El-Kassas S, Abdo SE, El-Naggar K, Shalaby HK, Nofal RY, Farag MR, Azzam MM, Lestingi A. Leverage of lysozyme dietary supplementation on gut health, hematological, antioxidant, and immune parameters in different plumage-colors Japanese quails. Poult Sci 2025; 104:104474. [PMID: 39571202 PMCID: PMC11617721 DOI: 10.1016/j.psj.2024.104474] [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: 08/21/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 12/08/2024] Open
Abstract
The current study was conducted on two different feather-colored Japanese quail varieties (brown and white) to examine the impact of lysozyme (LZ) dietary supplementation on growth performance, hematological profile, serum lysozyme, phagocytic and antioxidant activities, along with the gut status and the relative expression of some antioxidant- and immune-related genes. Two forms of LZ; extracted from egg white (natural LZ (NLZ)), and the commercial LZ (CLZ) were included in this experiment. For each quail variety, 240 birds were randomly allocated into four groups with four replicates per group. The first group (control) ate the basal diet (BD) only. The other groups ate the BD supplemented with commercial lysozyme (CLZ, at 100 mg/kg diet), NLZ at 100 (NLZ1) and 200 (NLZ2) mg/kg diet. Different LZ treatments differentially modulated the quail's growth performance with significant increases in the final body weight of white-feathered quails fed the NLZ1 compared to other treatments. The NLZ2 and CLZ noticeably increased the total antioxidant activity (TA) in the white- and brown-feathered quails, respectively. Also, all LZ groups displayed distinct increases in the serum lysozyme and phagocytic activities. For gut status, both varieties exhibited increases in intestinal villi length and goblet cell count with significant reductions in the total lactobacillus, total coliform, and total bacterial counts. These effects were linked with marked modulations of SOD, CAT, GPX, andIL-1βgene expression levels in both quail varieties. Therefore, the LZ could differentially impact quail growth, immune and antioxidant status as well as gut health.
Collapse
Affiliation(s)
- Ibrahim A Elkhaiat
- Department of Poultry Production, Faculty of Agriculture, Kafrelsheikh University, 33516, Egypt
| | - Seham El-Kassas
- Animal, Poultry and Fish Breeding and Production, Department of Animal Wealth Development, Faculty of Veterinary Medicine, Kafrelsheikh University, Egypt
| | - Safaa E Abdo
- Genetics and Genetic Engineering, Department of Animal Wealth Development, Faculty of Veterinary Medicine, Kafrelsheikh University, Egypt
| | - Karima El-Naggar
- Department of Nutrition and Veterinary Clinical Nutrition, Faculty of Veterinary Medicine, Alexandria University, Alexandria, 21944, Egypt
| | - Haitham K Shalaby
- Department of Poultry Production, Faculty of Agriculture, Kafrelsheikh University, 33516, Egypt
| | - Reyad Y Nofal
- Department of Poultry Production, Faculty of Agriculture, Kafrelsheikh University, 33516, Egypt
| | - Mayada R Farag
- Forensic Medicine and Toxicology Department, Veterinary Medicine Faculty, Zagazig University, Zagazig 44519, Egypt.
| | - Mahmoud M Azzam
- Department of Animal Production, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Antonia Lestingi
- Department of Veterinary Medicine, University of Bari Aldo Moro, S.P. per Casamassima km 3, 70010, Valenzano, BA, Italy
| |
Collapse
|
39
|
Nakamura K, Baba R, Kokubu K, Harada M, Morimoto H. Alterations in Ileal Secretory Cells of The DSS-Induced Colitis Model Mice. Acta Histochem Cytochem 2024; 57:199-209. [PMID: 39776935 PMCID: PMC11703563 DOI: 10.1267/ahc.24-00049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 10/18/2024] [Indexed: 01/11/2025] Open
Abstract
Inflammatory bowel disease is triggered by abnormalities in epithelial barrier function and immunological responses, although its pathogenesis is poorly understood. The dextran sodium sulphate (DSS)-induced colitis model has been used to examine inflammation in the colon. Damage to mucosa primality occurs in the large intestine and scarcely in the small intestine. To evaluate the effect on the ileum, we histologically analyzed the inflammatory and recovery phases in DSS model mice, and 40 kDa FITC-dextran was used to investigate barrier function. In the inflammatory phase, histological damage was insignificant. However, expanded crypts, hypertrophic goblet and Paneth cells, increased mucus production and secretion were observed. The cellular morphology was restored to that of the control in the recovery phase. According to in situ hybridization and lectin histochemistry, the expression of intestinal stem cell markers, secretory cell differentiation factors, and glycosylation of secretory granules in Paneth cells differed in the DSS model. DSS-treatment did not influence the barrier function in the ileum, and FITC-dextran did not diffuse via the paracellular pathway into the mucosa. However, cells incorporating FITC appeared even under normal conditions. The number of FITC-positive Paneth cells was lower in the DSS group than the control group. Our results showed morphological and functional alterations in ileal epithelial cells, especially secretory cells, in the DSS colitis model.
Collapse
Affiliation(s)
- Kenta Nakamura
- Third Department of Internal Medicine, University of Occupational and Environmental Health, 1–1, Iseigaoka, Yahatanishi, Kitakyushu, Fukuoka 807–8555, Japan
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, 1–1, Iseigaoka, Yahatanishi, Kitakyushu, Fukuoka 807–8555, Japan
| | - Ryoko Baba
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, 1–1, Iseigaoka, Yahatanishi, Kitakyushu, Fukuoka 807–8555, Japan
| | - Keiji Kokubu
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, 1–1, Iseigaoka, Yahatanishi, Kitakyushu, Fukuoka 807–8555, Japan
| | - Masaru Harada
- Third Department of Internal Medicine, University of Occupational and Environmental Health, 1–1, Iseigaoka, Yahatanishi, Kitakyushu, Fukuoka 807–8555, Japan
| | - Hiroyuki Morimoto
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, 1–1, Iseigaoka, Yahatanishi, Kitakyushu, Fukuoka 807–8555, Japan
| |
Collapse
|
40
|
Shen JW, Pan PK, Chen YY, Nan FH, Wu YS. Characteristics of Gracilariopsis lemaneiformis hydrocolloids and their effects on intestine PPAR signaling and liver lipid metabolism in Oreochromis niloticus: A multiomics analysis. Heliyon 2024; 10:e40416. [PMID: 39669144 PMCID: PMC11635660 DOI: 10.1016/j.heliyon.2024.e40416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 12/14/2024] Open
Abstract
This study evaluated the effects of Gracilariopsis lemaneiformis hydrocolloids on Nile tilapia (Oreochromis niloticus) using an advanced multiomics approach (transcriptome and proteome) linked with genomic isoform structure to elucidate the biofunctions of G. lemaneiformis hydrocolloids. The results showed that G. lemaneiformis hydrocolloids did not affect growth, as indicated by the nonsignificant differences in growth and blood biochemical indicators. Regarding the response, both intestine and liver tissues were assessed. These findings indicate that 20 % G. lemaneiformis hydrocolloids enhanced cytokine expression, which may contribute to a biological function in the intestine and liver of O. niloticus. Genome and proteome profiles indicated that G. lemaneiformis hydrocolloids upregulated the intestine and liver peroxisome proliferator-activated receptor (PPAR) signaling pathway, nucleocytoplasmic transport, steroid biosynthesis, and histidine metabolism. In contrast, co-factor biosynthesis, nucleocytoplasmic transport, tryptophan metabolism, arginine and proline metabolism, arginine biosynthesis, and ribosome activity were downregulated. These findings indicate that G. lemaneiformis hydrocolloids significantly affect liver lipid and carbohydrate metabolism. Proteomics analysis revealed that G. lemaneiformis hydrocolloids upregulated the PPAR signaling pathway, playing a crucial role in lipid metabolism. In summary, 20 % G. lemaneiformis hydrocolloids are primarily involved in modulating the intestine and liver PPAR signaling pathway to regulate lipid metabolism.
Collapse
Affiliation(s)
- Jia-Wei Shen
- Department of Aquaculture, National Pingtung University of Science and Technology, Pingtung, 912301, Taiwan
| | - Po-Kai Pan
- Department of Aquaculture, National Pingtung University of Science and Technology, Pingtung, 912301, Taiwan
| | - Yin-Yu Chen
- Department of Aquaculture, National Taiwan Ocean University, Keelung, 202301, Taiwan
| | - Fan-Hua Nan
- Department of Aquaculture, National Taiwan Ocean University, Keelung, 202301, Taiwan
| | - Yu-Sheng Wu
- Department of Aquaculture, National Pingtung University of Science and Technology, Pingtung, 912301, Taiwan
| |
Collapse
|
41
|
Templeton HN, Ehrlich AT, Schwerdtfeger LA, Sheng JA, Tjalkens RB, Tobet SA. Sex specific effects of environmental toxin-derived alpha synuclein on enteric neuronal-epithelial interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.27.625701. [PMID: 39677613 PMCID: PMC11642731 DOI: 10.1101/2024.11.27.625701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Background Parkinson's Disease (PD) is a neurodegenerative disorder with prodromal gastrointestinal (GI) issues often emerging decades before motor symptoms. Pathologically, PD can be driven by accumulation of misfolded alpha synuclein (aSyn) protein in the brain and periphery, including the GI tract. Disease epidemiology differs by sex, with men twice as likely to develop PD. Women, however, experience faster disease progression, higher mortality, and more severe GI symptoms. Gut calcitonin gene related peptide (CGRP) is a key regulator of intestinal contractions and visceral pain. The current study tests the hypothesis that sex differences in GI symptomology in PD are the result of aSyn aggregation altering enteric CGRP signaling pathways. Methods To facilitate peripheral aSyn aggregation, the pesticide rotenone was administered intraperitoneally once daily for two weeks to male and female mice. Mice were sacrificed two weeks after the last rotenone injection and immunohistochemistry was performed on sections of proximal colon. Key Results Levels of aSyn were heightened in myenteric plexus neurons and a subset of neurons immunoreactive to CGRP in rotenone treated mice. Female mice exhibited 153% more myenteric aSyn, 26% more apical CGRP immunoreactivity, and 66.7% more aSyn in apical CGRP + fibers after rotenone when compared to males. Goblet cell numbers were diminished but the individual cells were larger in the apical regions of crypts in the colons of rotenone treated mice. Conclusions This study used a mouse model of PD to uncover sex specific alterations in enteric neuronal and epithelial populations, underscoring the importance of considering sex as a biological variable while investigating prodromal GI symptoms. KEY POINTS Mouse model of Parkinson's Disease (PD) was used to investigate sex specific impact of enteric alpha synuclein (aSyn) on colonic goblet cells and CGRP + neurons and fibers. Sex specific alterations in intestinal neuronal and epithelial signaling pathways in response to aSyn provides insight into sex differences in PD etiology and prodromal gastrointestinal symptoms.
Collapse
|
42
|
Ren W, Li W, Cha X, Wang S, Cai B, Wang T, Li F, Li T, Xie Y, Xu Z, Wang Z, Liu H, Yu Y. Single-cell transcriptomic atlas of taste papilla aging. Aging Cell 2024; 23:e14308. [PMID: 39169434 PMCID: PMC11634696 DOI: 10.1111/acel.14308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 07/02/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024] Open
Abstract
Taste perception is one of the important senses in mammals. Taste dysfunction causes significant inconvenience in daily life, leading to subhealth and even life-threatening condition. Aging is a major cause to taste dysfunction, while the underlying feature related to gustatory aging is still not known. Using single-cell RNA Sequencing, differentially expressed genes between aged and young taste papillae are identified, including upregulated mt-Nd4l and Xist, as well as downregulated Hsp90ab1 and Tmem59. In the Tmem59-/- circumvallate papillae (CVP), taste mature cell generation is impaired by reduction in the numbers of PLCβ2+ and Car4+ cells, as well as decreases in expression levels of taste transduction genes. Tmem59-/- mice showed deficits in sensitivities to tastants. Through screening by GenAge and DisGeNET databases, aging-dependent genes and oral disease-associated genes are identified in taste papillae. In the CVP, aging promotes intercellular communication reciprocally between (cycling) basal cell and mature taste cell by upregulated Crlf1/Lifr and Adam15/Itga5 signaling. By transcriptional network analysis, ribosome proteins, Anxa1, Prdx5, and Hmgb1/2 are identified as transcriptional hubs in the aged taste papillae. Chronological aging-associated transcriptional changes throughout taste cell maturation are revealed. Aged taste papillae contain more Muc5b+ cells that are not localized in gustatory gland. Collectively, this study shows molecular and cellular features associated with taste papilla aging.
Collapse
Affiliation(s)
- Wenwen Ren
- Department of OtolaryngologyThe Second Affiliated Hospital of the Naval Medical University (Shanghai Changzheng Hospital)ShanghaiChina
| | - Weihao Li
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan UniversityShanghaiChina
- Olfactory Disorder Diagnosis and Treatment CenterEye & ENT Hospital, Fudan UniversityShanghaiChina
| | - Xudong Cha
- Department of OtolaryngologyThe Second Affiliated Hospital of the Naval Medical University (Shanghai Changzheng Hospital)ShanghaiChina
| | - Shenglei Wang
- Department of OtolaryngologyThe Second Affiliated Hospital of the Naval Medical University (Shanghai Changzheng Hospital)ShanghaiChina
| | - Boyu Cai
- Department of OtolaryngologyThe Second Affiliated Hospital of the Naval Medical University (Shanghai Changzheng Hospital)ShanghaiChina
| | - Tianyu Wang
- Department of OtolaryngologyThe Second Affiliated Hospital of the Naval Medical University (Shanghai Changzheng Hospital)ShanghaiChina
| | - Fengzhen Li
- Department of OtolaryngologyThe Second Affiliated Hospital of the Naval Medical University (Shanghai Changzheng Hospital)ShanghaiChina
| | - Tengfei Li
- Department of OtolaryngologyThe Second Affiliated Hospital of the Naval Medical University (Shanghai Changzheng Hospital)ShanghaiChina
| | - Yingqi Xie
- Department of OtolaryngologyThe Second Affiliated Hospital of the Naval Medical University (Shanghai Changzheng Hospital)ShanghaiChina
| | - Zengyi Xu
- Department of OtolaryngologyThe Second Affiliated Hospital of the Naval Medical University (Shanghai Changzheng Hospital)ShanghaiChina
| | - Zhe Wang
- Department of OtolaryngologyThe Second Affiliated Hospital of the Naval Medical University (Shanghai Changzheng Hospital)ShanghaiChina
| | - Huanhai Liu
- Department of OtolaryngologyThe Second Affiliated Hospital of the Naval Medical University (Shanghai Changzheng Hospital)ShanghaiChina
| | - Yiqun Yu
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan UniversityShanghaiChina
- Olfactory Disorder Diagnosis and Treatment CenterEye & ENT Hospital, Fudan UniversityShanghaiChina
| |
Collapse
|
43
|
Shah DD, Chorawala MR, Pandya AJ, Kothari N, Prajapati BG, Parekh PS. Advancing the Battle against Cystic Fibrosis: Stem Cell and Gene Therapy Insights. Curr Med Sci 2024; 44:1155-1174. [PMID: 39676146 DOI: 10.1007/s11596-024-2936-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 09/03/2024] [Indexed: 12/17/2024]
Abstract
Cystic fibrosis (CF) is a hereditary disorder characterized by mutations in the CFTR gene, leading to impaired chloride ion transport and subsequent thickening of mucus in various organs, particularly the lungs. Despite significant progress in CF management, current treatments focus mainly on symptom relief and do not address the underlying genetic defects. Stem cell and gene therapies present promising avenues for tackling CF at its root cause. Stem cells, including embryonic, induced pluripotent, mesenchymal, hematopoietic, and lung progenitor cells, offer regenerative potential by differentiating into specialized cells and modulating immune responses. Similarly, gene therapy aims to correct CFTR gene mutations by delivering functional copies of the gene into affected cells. Various approaches, such as viral and nonviral vectors, gene editing with CRISPR-Cas9, small interfering RNA (siRNA) therapy, and mRNA therapy, are being explored to achieve gene correction. Despite their potential, challenges such as safety concerns, ethical considerations, delivery system optimization, and long-term efficacy remain. This review provides a comprehensive overview of the current understanding of CF pathophysiology, the rationale for exploring stem cell and gene therapies, the types of therapies available, their mechanisms of action, and the challenges and future directions in the field. By addressing these challenges, stem cell and gene therapies hold promise for transforming CF management and improving the quality of life of affected individuals.
Collapse
Affiliation(s)
- Disha D Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, 380009, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, 380009, India
| | - Aanshi J Pandya
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, 380009, India
| | - Nirjari Kothari
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, 380009, India
| | - Bhupendra G Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Shree S. K. Patel College of Pharmaceutical Education & Research, Ganpat University, Mehsana, 384012, India.
- Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand.
| | | |
Collapse
|
44
|
Oliveira ICCS, Marinsek GP, Gonçalves ARN, Lopes BS, Correia LVB, Da Silva RCB, Castro IB, Mari RB. Investigating tributyltin's toxic effects: Intestinal barrier and neuroenteric disruption in rat's jejunum. Neurotoxicology 2024; 105:208-215. [PMID: 39396746 DOI: 10.1016/j.neuro.2024.10.004] [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: 06/20/2024] [Revised: 09/24/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Abstract
The expansion of economic activities in coastal areas has significantly increased chemical contamination, leading to major environmental challenges. Contaminants enter the human body through the food chain, particularly via seafood and water consumption, triggering biomagnification and bioaccumulation processes. The gastrointestinal tract (GIT) acts as a selective barrier, protecting against chemical pollutants and maintaining homeostasis through a complex network of cells and immune responses. This study assessed impact of tributyltin (TBT), a highly toxic organometallic compound used in antifouling coatings for ships, on the GIT and myenteric neural plasticity in young rats. TBT exposure leads to histopathological changes, including epithelial detachment and inflammatory foci, especially at lower environmental doses. The study found that TBT causes significant reductions in villi height, increases in goblet cells and intraepithelial lymphocytes, and disrupts the myenteric plexus, with higher densities of extraganglionic neurons in exposed animals.
Collapse
Affiliation(s)
- I C C S Oliveira
- UNESP, São Paulo State University, Institute of Biosciences, Paulista Coast Campus (CLP), São Vicente, SP, Brazil.
| | - G P Marinsek
- UNESP, São Paulo State University, Institute of Biosciences, Paulista Coast Campus (CLP), São Vicente, SP, Brazil
| | - A R N Gonçalves
- UNESP, São Paulo State University, Institute of Biosciences, Paulista Coast Campus (CLP), São Vicente, SP, Brazil
| | - B S Lopes
- UNESP, São Paulo State University, Institute of Biosciences, Paulista Coast Campus (CLP), São Vicente, SP, Brazil
| | - L V B Correia
- UNIFESP, Federal University of São Paulo, Institute of Health and Society, Baixada Santista Campus, Santos, SP, Brazil
| | - R C B Da Silva
- UNIFESP, Federal University of São Paulo, Institute of Health and Society, Baixada Santista Campus, Santos, SP, Brazil
| | - I B Castro
- UNIFESP, Federal University of São Paulo, Institute of Marine Science, Baixada Santista Campus, Santos, SP, Brazil
| | - R B Mari
- UNESP, São Paulo State University, Institute of Biosciences, Paulista Coast Campus (CLP), São Vicente, SP, Brazil
| |
Collapse
|
45
|
Luo J, Gao J, Song H, Mo Z, Hong B, Zhu L, Song W, Qian G, Li C. Low temperature alleviated the adverse effects of simulated transport stress on the intestinal health in Chinese soft-shelled turtle Pelodiscus sinensis. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109936. [PMID: 39357630 DOI: 10.1016/j.fsi.2024.109936] [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: 07/20/2024] [Revised: 09/15/2024] [Accepted: 09/30/2024] [Indexed: 10/04/2024]
Abstract
Transport stress always poses a threat to aquatic animals. Transportation under low temperatures was often used to relieve transport stress in practical production of Chinese soft-shelled turtle Pelodiscus sinensis, but their effect on the turtle's intestinal barrier remains unclear. In this study, P. sinensis (initial weight 200 ± 20 g) were exposed to simulated transport stress for 12 h at control (30 °C) and low (20 °C) temperature, and then recovery for 24 h, and each treatment had 4 replicates with each replicate containing 4 turtles. The results showed that transportation induced obvious morphological and histological damages in intestinal villus, with a down-regulated expression of the tight junction related genes. Besides turtles in transport group showed an oxidative stress in intestine, which stimulated a physiological detoxification response together with apoptosis. Low temperature transport plays a mitigative effect on the transport stress of turtle intestine via relieved stress response. Specifically, the intestinal villus/crypt (V/C) ratio and the expression of tight junction genes in the low-temperature group were significantly higher compared to the control temperature group, while stress response parameters such as intestinal cortisol levels and hsp expression were significantly lower in the low-temperature group. Additionally, low temperature alleviated oxidative damage and apoptosis caused by transport stress relative to the control temperature group. However, the protective effect of low temperature on P. sinensis intestine was limited, especially after the temperature recovery stage. Overall, the findings of the present study demonstrated that transport stress would induce the disruption of intestinal integrity and oxidative damage, also activated the mucosal immunity and antioxidant enzyme system response of turtles. It was also suggested that low temperature could alleviate the adverse effects of transport stress on intestinal integrity through modulation of oxidative status and apoptosis, whereas much less impact after temperature recovery.
Collapse
Affiliation(s)
- Jiaxiang Luo
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100, China
| | - Jintao Gao
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100, China
| | - Haoran Song
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100, China
| | - Zhiyin Mo
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100, China
| | - Binquan Hong
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100, China
| | - Leyan Zhu
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100, China
| | - Wei Song
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100, China
| | - Guoying Qian
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100, China.
| | - Caiyan Li
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100, China.
| |
Collapse
|
46
|
Provitera L, Tomaselli A, Algieri F, Tripodi M, Raffaeli G, Amodeo I, Raymo L, Bronzoni CV, Fumagalli M, Garrido F, Cavallaro G. Gut Microbiota-Derived Metabolites and Their Role in the Pathogenesis of Necrotizing Enterocolitis in Preterm Infants: A Narrative Review. Metabolites 2024; 14:570. [PMID: 39590806 PMCID: PMC11596930 DOI: 10.3390/metabo14110570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease that occurs predominantly in premature infants and is characterized by the inflammation and necrosis of the intestine, showing high morbidity and mortality rates. Despite decades of research efforts, a specific treatment is currently lacking, and preventive strategies are the mainstays of care. This review aims to help understand the complex interplay between gut microbiota and their metabolites in NEC pathogenesis. In particular, we focused on how these factors can influence gut health, immune responses, and intestinal barrier integrity. Discussion: Current research has increasingly focused on the role of the gut microbiota and their metabolites in NEC pathogenesis, thanks to their involvement in modulating gut health, immune responses, and intestinal barrier integrity. Conclusions: A deeper understanding of the interplay between gut microbiota and their metabolites is essential for developing personalized strategies to prevent NEC. By targeting these microbial interactions, new therapeutic approaches may emerge that offer improved outcomes for preterm infants at a high risk of NEC.
Collapse
Affiliation(s)
- Livia Provitera
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
| | - Andrea Tomaselli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Francesca Algieri
- Research and Development Unit, Postbiotica S.R.L., 20123 Milan, Italy;
| | - Matteo Tripodi
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
| | - Genny Raffaeli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
| | - Ilaria Amodeo
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
| | - Ludovica Raymo
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Carolina Vittoria Bronzoni
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Monica Fumagalli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Felipe Garrido
- Department of Pediatrics, Clínica Universidad de Navarra, 28027 Madrid, Spain;
| | - Giacomo Cavallaro
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (A.T.); (M.T.); (G.R.); (I.A.); (L.R.); (C.V.B.); (M.F.); (G.C.)
| |
Collapse
|
47
|
Gao F, Wu S, Zhang K, Xu Z, Zhang X, Zhu Z, Quan F. Goat Milk Exosomes Ameliorate Ulcerative Colitis in Mice through Modulation of the Intestinal Barrier, Gut Microbiota, and Metabolites. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23196-23210. [PMID: 39390385 DOI: 10.1021/acs.jafc.4c03212] [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: 10/12/2024]
Abstract
Goat milk is rich in a variety of nutrients that are important for intestinal health and disease prevention. However, the role of exosomes in goat milk remains to be elucidated. This study investigated for the first time the therapeutic efficacy and molecular underlying mechanisms of mature milk exosomes (M-exo) and goat colostrum exosomes (C-exo) on dextran sodium sulfate-induced ulcerative colitis (UC) in mice. The findings demonstrate that M-exo and C-exo significantly improved physiological indices, suppressed the secretion of proinflammatory cytokines, and diminished oxidative stress and apoptosis in UC mice. Moreover, C-exo and M-exo restored the intestinal barrier function, remodeled the gut microbiota, and improved metabolite composition in the feces of colitis mice. In conclusion, goat milk exosomes ameliorate UC in mice, which provides a basis for the development of functional food applications for the prevention and treatment of inflammatory bowel disease.
Collapse
Affiliation(s)
- Feng Gao
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Shenghui Wu
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Kang Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Zhiming Xu
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Xin Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Zhengjin Zhu
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Fusheng Quan
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
48
|
Way R, Templeton H, Ball D, Cheng MH, Tobet SA, Chen T. A microphysiological system for studying barrier health of live tissues in real time. COMMUNICATIONS ENGINEERING 2024; 3:142. [PMID: 39396075 PMCID: PMC11470921 DOI: 10.1038/s44172-024-00285-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 09/30/2024] [Indexed: 10/14/2024]
Abstract
Epithelial cells create barriers that protect many different components in the body from their external environment. Increased gut barrier permeability (leaky gut) has been linked to several chronic inflammatory diseases. Understanding the cause of leaky gut and effective interventions are elusive due to the lack of tools that maintain tissue's physiological environment while elucidating cellular functions under various stimuli ex vivo. Here we present a microphysiological system that records real-time barrier permeability of mouse colon in a physiological environment over extended durations. The system includes a microfluidic chamber; media composition that preserves microbiome and creates necessary oxygen gradients across the barrier; and integrated sensor electrodes for acquiring transepithelial electrical resistance (TEER). Our results demonstrate that the system can maintain tissue viability for up to 72 h. The TEER sensors can distinguish levels of barrier permeability when treated with collagenase and low pH media and detect different thickness in the tissue explant.
Collapse
Affiliation(s)
- Ryan Way
- Department of Electrical & Computer Engineering, Colorado State University, Fort Collins, CO, USA
| | - Hayley Templeton
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Daniel Ball
- Department of Electrical & Computer Engineering, Colorado State University, Fort Collins, CO, USA
| | - Ming-Hao Cheng
- Department of Electrical & Computer Engineering, Colorado State University, Fort Collins, CO, USA
| | - Stuart A Tobet
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Thomas Chen
- Department of Electrical & Computer Engineering, Colorado State University, Fort Collins, CO, USA.
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
49
|
Nie HY, Ge J, Huang GX, Liu KG, Yue Y, Li H, Lin HG, Zhang T, Yan HF, Xu BX, Sun HW, Yang JW, Si SY, Zhou JL, Cui Y. New insights into the intestinal barrier through "gut-organ" axes and a glimpse of the microgravity's effects on intestinal barrier. Front Physiol 2024; 15:1465649. [PMID: 39450142 PMCID: PMC11499591 DOI: 10.3389/fphys.2024.1465649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/02/2024] [Indexed: 10/26/2024] Open
Abstract
Gut serves as the largest interface between humans and the environment, playing a crucial role in nutrient absorption and protection against harmful substances. The intestinal barrier acts as the initial defense mechanism against non-specific infections, with its integrity directly impacting the homeostasis and health of the human body. The primary factor attributed to the impairment of the intestinal barrier in previous studies has always centered on the gastrointestinal tract itself. In recent years, the concept of the "gut-organ" axis has gained significant popularity, revealing a profound interconnection between the gut and other organs. It speculates that disruption of these axes plays a crucial role in the pathogenesis and progression of intestinal barrier damage. The evaluation of intestinal barrier function and detection of enterogenic endotoxins can serve as "detecting agents" for identifying early functional alterations in the heart, kidney, and liver, thereby facilitating timely intervention in the disorders. Simultaneously, consolidating intestinal barrier integrity may also present a potential therapeutic approach to attenuate damage in other organs. Studies have demonstrated that diverse signaling pathways and their corresponding key molecules are extensively involved in the pathophysiological regulation of the intestinal barrier. Aberrant activation of these signaling pathways and dysregulated expression of key molecules play a pivotal role in the process of intestinal barrier impairment. Microgravity, being the predominant characteristic of space, can potentially exert a significant influence on diverse intestinal barriers. We will discuss the interaction between the "gut-organ" axes and intestinal barrier damage, further elucidate the signaling pathways underlying intestinal barrier damage, and summarize alterations in various components of the intestinal barrier under microgravity. This review aims to offer a novel perspective for comprehending the etiology and molecular mechanisms of intestinal barrier injury as well as the prevention and management of intestinal barrier injury under microgravity environment.
Collapse
Affiliation(s)
- Hong-Yun Nie
- Department of General Surgery, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Jun Ge
- Clinical laboratory, The Ninth Medical Center of the PLA General Hospital, Beijing, China
| | - Guo-Xing Huang
- 306th Clinical College of PLA, The Fifth Clinical College, Anhui Medical University, Beijing, China
| | - Kai-Ge Liu
- Department of General Surgery, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Yuan Yue
- Department of Disease Control and Prevention, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Hao Li
- Department of General Surgery, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Hai-Guan Lin
- Department of General Surgery, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Tao Zhang
- Department of General Surgery, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Hong-Feng Yan
- Department of General Surgery, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Bing-Xin Xu
- Special Medical Laboratory Center, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Hong-Wei Sun
- Department of General Surgery, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Jian-Wu Yang
- Department of General Surgery, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Shao-Yan Si
- Special Medical Laboratory Center, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Jin-Lian Zhou
- Department of Pathology, The Ninth Medical Center of PLA General Hospital, Beijing, China
| | - Yan Cui
- Department of General Surgery, The Ninth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
50
|
Dewanti R, Martien R, Ariyadi B, Zuprizal. Nano-emulsified black soldier fly oil concerning performance traits, health, and immunity of broilers. Poult Sci 2024; 103:104116. [PMID: 39128390 PMCID: PMC11367124 DOI: 10.1016/j.psj.2024.104116] [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: 07/16/2024] [Accepted: 07/24/2024] [Indexed: 08/13/2024] Open
Abstract
In the antibiotic-free era, traditional antibiotics have been suggested as alternatives to antibiotic-based growth promoters. Among the various methods, self-nano-emulsifying drug delivery systems (SNEDDS) are increasingly utilized to improve the bioavailability of oils containing essential substances. In this study, we evaluated the effects of black soldier fly oil (BSFO) SNEDDS in chicken drinking water on growth performance, small intestine histomorphology, and poultry health status. We divided 225 male Indian River strain broiler chickens into five treatment groups, each consisting of 5 replicates. The chickens were reared from to 0 to 35 d of age in a controlled environmental housing system. The BSFO SNEDDS was administered via drinking water. Treatments included ordinary drinking water (P1), bacitracin (P2), and 10 mL/L (P3), 20 mL/L (P4), and 30 mL/L (P5) BSFO SNEDDS. The observed parameters included growth performance, carcass yield, blood hematology, intestinal histomorphology, digestive microflora, and immunoglobulin (Ig) levels. Data were analyzed using analysis of variance (ANOVA) and Duncan's test. The results indicated that administering BSFO SNEDDS via drinking water improved feed conversion (P < 0.05), enhanced the performance index (P < 0.05), increased carcass percentage (P < 0.001), and increased the weight of the carcass parts. Additionally, it increased villus height (P < 0.01), lowered jejunal pH (P < 0.001), reduced pathogenic bacteria in the jejunum, and decreased the leukocyte count. BSFO SNEDDS in drinking water is expected to be used as a traditional antibiotic with the potential to replace synthetic antibiotic growth promoters in broiler chickens.
Collapse
Affiliation(s)
- Ratih Dewanti
- Department of Animal Science, Faculty of Animal Science, Universitas Sebelas Maret, Surakarta, Indonesia; Department of Animal Nutrition and Feed Science, Faculty of Animal Science, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| | - Ronny Martien
- Department of Pharmaceutics, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Bambang Ariyadi
- Department of Animal Production, Faculty of Animal Science, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Zuprizal
- Department of Animal Nutrition and Feed Science, Faculty of Animal Science, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|