1
|
Liu Z, Zhang H, Wang J, Yao Y, Wang X, Liu Y, Fang W, Liu X, Zheng Y. Clca1 deficiency exacerbates colitis susceptibility via impairment of mucus barrier integrity and gut microbiota homeostasis. Microbiol Res 2025; 297:128191. [PMID: 40300372 DOI: 10.1016/j.micres.2025.128191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/21/2025] [Accepted: 04/21/2025] [Indexed: 05/01/2025]
Abstract
The intestinal mucus barrier has emerged as a promising therapeutic target for inflammatory bowel disease. Understanding its regulatory mechanisms is critical for elucidating ulcerative colitis (UC) pathogenesis, improving diagnostics, guiding treatments, and preventing relapse. Chloride Channel Accessory 1 (Clca1), a constituent of the mucus layer, remains understudied in colitis. Here, we investigated Clca1's role in mucosal immunity and intestinal homeostasis using experimental colitis models. Clca1-deficient (Clca1-/-) mice displayed compromised mucus layer integrity, reduced neutrophil infiltration, and gut microbiota dysbiosis. Notably, Clca1-/- mice exhibited exacerbated colitis severity following dextran sulfate sodium (DSS) challenge, accompanied by a diminished goblet cell populations. Fecal microbiota transplantation (FMT) studies revealed that gut microbiota critically modulates divergent phenotypic outcomes between genotypes. Our findings establish Clca1 as a multifunctional regulator of mucus barrier integrity through mechanisms involving goblet cell maintenance, neutrophil-mediated immunity, and host-microbiota crosstalk. These results advance the understanding of UC pathogenesis and identify Clca1-associated pathways as potential targets for barrier restoration therapies.
Collapse
Affiliation(s)
- Zhi Liu
- Department of Microbiology, State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Hong Zhang
- Department of Microbiology, State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jingjing Wang
- Department of Microbiology, State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yutong Yao
- Department of Microbiology, State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiaoyi Wang
- Core Facility Center, The First Afliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yang Liu
- Department of Microbiology, State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Weijia Fang
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Xingyin Liu
- Department of Microbiology, State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China; Department of Biochemistry, SUSTech Homeostatic Medicine Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China.
| | - Yi Zheng
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.
| |
Collapse
|
2
|
Neurath MF, Artis D, Becker C. The intestinal barrier: a pivotal role in health, inflammation, and cancer. Lancet Gastroenterol Hepatol 2025; 10:573-592. [PMID: 40086468 DOI: 10.1016/s2468-1253(24)00390-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/08/2024] [Accepted: 11/15/2024] [Indexed: 03/16/2025]
Abstract
The intestinal barrier serves as a boundary between the mucosal immune system in the lamina propria and the external environment of the intestinal lumen, which contains a diverse array of microorganisms and ingested environmental factors, including pathogens, food antigens, toxins, and other foreign substances. This barrier has a central role in regulating the controlled interaction between luminal factors and the intestinal immune system. Disruptions of intestinal epithelial cells, which serve as a physical barrier, or the antimicrobial peptides and mucins they produce, which act as a chemical barrier, can lead to a leaky gut. In this state, the intestinal wall is unable to efficiently separate the intestinal flora and luminal contents from the intestinal immune system. The subsequent activation of the immune system has an important role in the pathogenesis of inflammatory bowel disease, as well as in metabolic dysfunction-associated steatohepatitis, primary sclerosing cholangitis, and colorectal cancer. Dysregulated intestinal barrier integrity has also been described in patients with chronic inflammatory diseases outside the gastrointestinal tract, including rheumatoid arthritis and neurodegenerative disorders. Mechanistic studies of barrier dysfunction have revealed that the subsequent local activation and systemic circulation of activated immune cells and the cytokines they secrete, as well as extracellular vesicles, promote proinflammatory processes within and outside the gastrointestinal tract. In this Review, we summarise these findings and highlight several new therapeutic concepts currently being developed that attempt to control inflammatory processes via direct or indirect modulation of intestinal barrier function.
Collapse
Affiliation(s)
- Markus F Neurath
- Medical Clinic 1, Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA; Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA; Joan and Sanford I Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Allen Discovery Center for Neuroimmune Interactions, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Christoph Becker
- Medical Clinic 1, Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
3
|
Fan F, Guo R, Pan K, Xu H, Chu X. Mucus and mucin: changes in the mucus barrier in disease states. Tissue Barriers 2025:2499752. [PMID: 40338015 DOI: 10.1080/21688370.2025.2499752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 05/09/2025] Open
Abstract
In this review we discuss mucus, the viscoelastic secretion from goblet or mucous producing cells that covers and protects all non-keratinized wet epithelial surfaces. In addition to the surface of organs directly contacting with the external environment such as the eyes, this layer provides protection to the underlying gastrointestinal, respiratory and female reproductive tracts by trapping pathogens, irritants, environmental fine particles and potentially harmful foreign substances. Mucins, the primary structural components of mucus, form structurally different mucus layers at different sites in a process regulated by a variety of factors. Currently, more and more studies have shown that the mucus barrier is not only closely related to various intestinal mucus diseases, but also involved in the occurrence and development of various airway diseases and mucus-related diseases, thus it may become a new target for the treatment of various related diseases in the future. Since the dysfunction of the mucous layer is closely related to various pathological processes, in-depth understanding of its molecular mechanism and physiological role is of great theoretical and practical significance for disease prevention and treatment. Here, we discuss different aspects of the mucus layer by focusing on its chemical composition, synthetic pathways, and some of the characteristics of the mucus layer in physiological and pathological situations.
Collapse
Affiliation(s)
- Fangfang Fan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Ruihan Guo
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Kun Pan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Hongye Xu
- Quality Assurance department, Tongling Institutes for Food and Drug Control, Tongling, China
| | - Xiaoqin Chu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China
- Engineering Technology Research Center of Modern Pharmaceutical Preparation, Hefei, Anhui Province, China
| |
Collapse
|
4
|
van der Werf J, Fleming NI. Are single nucleotide polymorphisms underutilized for guiding treatment of inflammatory bowel disease? Immunol Cell Biol 2025. [PMID: 40313162 DOI: 10.1111/imcb.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 05/03/2025]
Abstract
Inflammatory bowel disease (IBD), encompassing Crohn's disease (CD), ulcerative colitis (UC) and IBD unclassified (IBDU), significantly impacts quality of life. Despite significant advances in the management of the conditions, responses to treatments vary greatly, and this is due partly to our natural genetic variation. Here we will review the evidence for whether single nucleotide polymorphisms (SNPs) have the potential to guide treatment decisions for people with IBD. We will first consider SNPs that exhibit strong associations with IBD pathogenesis and their relevance to epithelial barrier integrity, cytokine production, and immune system function. Then, we will cover those SNPs implicated in altering response to our various current IBD therapeutics, including the recently implemented drugs ustekinumab and tofacitinib. Finally, we will explore lesser-known SNPs that exhibit complex relationships with the disease and which may be undervalued as pharmacogenetic tools. Overall, it will be demonstrated that SNPs associated with IBD pathology are largely distinct from those predicting response to treatments and that new discoveries of clinically useful tools can be expected from therapy-focused investigations. Given the growing list of treatments available, we argue that beneficial personalization of treatments based on SNPs is still underutilized.
Collapse
Affiliation(s)
| | - Nicholas Ian Fleming
- Department of Pathology, University of Otago, Dunedin, New Zealand
- The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
5
|
He J, Zhang Y, Li M, Xing H, Lü L, Zhao X, Han Z. Polysaccharide extracted from peony seed meal preventive effect of on loperamide-induced constipation in rats. Int J Biol Macromol 2025; 308:142391. [PMID: 40139599 DOI: 10.1016/j.ijbiomac.2025.142391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/26/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
In the present study, one polysaccharide (named PSP-1) was purified from peony (Paeonia suffruticosa Andr.) seed meal, and its structural characteristics, as well as preventive constipation activity, were investigated. The weight average molecular weight of PSP-1 was 15.3 kDa and was composed of arabinose, galactose, glucose, xylose, and mannose in the molar ratio of 1:4.8:5.1:0.4:0.2. The primary structure of PSP-1 was elucidated through Fourier-transform infrared spectroscopy (FT-IR) and methylation analysis, revealing its glycosidic linkages and branching pattern. In the loperamide-induced constipated rat, PSP-1 significantly increased fecal parameters and accelerated intestinal transit (69 min reduction). PSP-1 enhanced total short-chain fatty acid (SCFAs) production by 9.1 %, particularly propionic and butyric acids. Furthermore, PSP-1 had a bidirectional effect on gastrointestinal hormones: it increased the serum levels of motilin, gastric inhibitory polypeptide, and substance P by 12.8 %, 13.0 %, and 13.4 %, respectively, while decreasing the levels of secretin and vasoactive intestinal polypeptide by 21.7 % and 17.6 %, respectively. Additionally, 16S rRNA sequencing indicated that PSP-1-mediated gut microbiota rebalances, suppressing pathogens and enriching beneficial genera. These findings suggested that PSP-1, a natural plant extract, maybe a dietary supplement that helps prevent constipation, providing a theoretical reference for its application in functional foods.
Collapse
Affiliation(s)
- Jinxing He
- College of Food Science and Engineering, Shandong Key Laboratory of Healthy Food Resources Exploration and Creation, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, PR China
| | - Yingyu Zhang
- College of Food Science and Engineering, Shandong Key Laboratory of Healthy Food Resources Exploration and Creation, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, PR China
| | - Mengyi Li
- College of Food Science and Engineering, Shandong Key Laboratory of Healthy Food Resources Exploration and Creation, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, PR China
| | - Hanzhu Xing
- College of Food Science and Engineering, Shandong Key Laboratory of Healthy Food Resources Exploration and Creation, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, PR China
| | - Lei Lü
- College of Food Science and Engineering, Shandong Key Laboratory of Healthy Food Resources Exploration and Creation, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, PR China
| | - Xiaolei Zhao
- College of Food Science and Engineering, Shandong Key Laboratory of Healthy Food Resources Exploration and Creation, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, PR China
| | - Zhonghui Han
- College of Food Science and Engineering, Shandong Key Laboratory of Healthy Food Resources Exploration and Creation, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, PR China; Shandong Engineering Research Center of Food Nutrition and Active Health, Shandong Xiwang Foodstuffs Co., Ltd., Binzhou 256200, PR China.
| |
Collapse
|
6
|
Fu Y, Guzior DV, Okros M, Bridges C, Rosset SL, González CT, Martin C, Karunarathne H, Watson VE, Quinn RA. Balance between bile acid conjugation and hydrolysis activity can alter outcomes of gut inflammation. Nat Commun 2025; 16:3434. [PMID: 40210868 PMCID: PMC11985902 DOI: 10.1038/s41467-025-58649-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 03/27/2025] [Indexed: 04/12/2025] Open
Abstract
Conjugated bile acids (BAs) are multi-functional detergents in the gastrointestinal (GI) tract produced by the liver enzyme bile acid-CoA:amino acid N-acyltransferase (BAAT) and by the microbiome from the acyltransferase activity of bile salt hydrolase (BSH). Humans with inflammatory bowel disease (IBD) have an enrichment in both host and microbially conjugated BAs (MCBAs), but their impacts on GI inflammation are not well understood. We investigated the role of host-conjugated BAs in a mouse model of colitis using a BAAT knockout background. Baat-/- KO mice have severe phenotypes in the colitis model that were rescued by supplementation with taurocholate (TCA). Gene expression and histology showed that this rescue was due to an improved epithelial barrier integrity and goblet cell function. However, metabolomics also showed that TCA supplementation resulted in extensive metabolism to secondary BAs. We therefore investigated the BSH activity of diverse gut bacteria on a panel of conjugated BAs and found broad hydrolytic capacity depending on the bacterium and the amino acid conjugate. The complexity of this microbial BA hydrolysis led to the exploration of bsh genes in metagenomic data from human IBD patients. Certain bsh sequences were enriched in people with Crohn's disease particularly that from Ruminococcus gnavus. This study shows that both host and microbially conjugated BAs may provide benefits to those with IBD, but this is dictated by a delicate balance between BA conjugation/deconjugation based on the bsh genes present.
Collapse
Affiliation(s)
- Yousi Fu
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Douglas V Guzior
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
- Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, MI, USA
| | - Maxwell Okros
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Christopher Bridges
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Sabrina L Rosset
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Cely T González
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Christian Martin
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
- Mass Spectrometry and Metabolomics Core, Michigan State University, East Lansing, MI, USA
| | - Hansani Karunarathne
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Victoria E Watson
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Robert A Quinn
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA.
- Department of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
7
|
Cin M, Gündoğar Ö, Yarıkkaya E, Cin S. The importance of MUC6 immunohistochemistry staining in the histopathologic examination of Crohn's disease: can we enhance our diagnostic power? Ann Diagn Pathol 2025; 78:152482. [PMID: 40239438 DOI: 10.1016/j.anndiagpath.2025.152482] [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: 03/08/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Crohn's disease (CD) is a chronic inflammatory bowel disease characterized by transmural inflammation and histopathologic variability, making diagnosis challenging. Pseudopyloric metaplasia (PPM) is a key histological feature of chronicity in CD. However, its identification on hematoxylin & eosin (H&E)-stained slides is subject to interobserver variability. MUC6 immunohistochemistry (IHC) has been suggested as a useful marker for pyloric glands. This study aims to evaluate the diagnostic utility of MUC6 staining in detecting PPM and to assess interobserver agreement compared to H&E staining. In this retrospective study, 38 terminal ileum biopsies from CD patients were analyzed. H&E-stained and MUC6-stained slides were evaluated independently by four pathologists for the presence and gland count of PPM. Intraobserver and interobserver agreements were assessed using Intraclass Correlation Coefficient (ICC). The mean PPM count was significantly higher with MUC6 staining than by H&E alone. Intraobserver agreement between H&E and MUC6 staining was "moderate" (ICC = 0.577, 0.734, 0.738) for three pathologists and "poor" (ICC = 0.439) for one. Interobserver agreement was classified as "good" for H&E slides (ICC = 0.849) and "excellent" for MUC6-stained slides (ICC = 0.993). PPM is an important finding that is not specific for CD but indicates chronic mucosal damage in the gastrointestinal tract. In our study, MUC6 IHC staining in CD improved the detection of PPM and increased interobserver agreement in the detection of PPM. The increase in the number of detectable metaplastic glands in all observers demonstrates the potential of MUC6 staining as a reliable marker. MUC6 IHC may provide a more standardized and objective evaluation, reducing diagnostic variability.
Collapse
Affiliation(s)
- Merve Cin
- Department of Pathology, University of Health Science, Istanbul Training and Research Hospital, Istanbul, Turkey.
| | - Özgecan Gündoğar
- Department of Pathology, University of Health Science, Gaziosmanpasa Training and Research Hospital, Istanbul, Turkey
| | - Enver Yarıkkaya
- Department of Pathology, University of Health Science, Istanbul Training and Research Hospital, Istanbul, Turkey
| | - Selçuk Cin
- Department of Pathology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
8
|
Chieppa M, De Santis S, Verna G. Winnie Mice: A Chronic and Progressive Model of Ulcerative Colitis. Inflamm Bowel Dis 2025; 31:1158-1167. [PMID: 39912845 PMCID: PMC11985403 DOI: 10.1093/ibd/izaf006] [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: 12/23/2024] [Indexed: 02/07/2025]
Abstract
Recent trends show a continuous worldwide rise in the incidence of ulcerative colitis (UC), leading to increased interest in its etiology and pathogenesis, which is currently unknown. To gain a better mechanistic understanding of this disease, many mouse models have been developed over the last several years, with variations of dextran sodium sulfate administration representing the most widely employed. The Winnie mouse strain was created through elicited random mutations in Muc2, resulting in a progressive, chronic intestinal inflammation localized to the colon that worsens over time. Moreover, Winnie mice display immunologic and microbiota features that are similar to those that can be found in UC patients. Phenotypically, the presence, albeit rare, of rectal prolapse and other complications impacting quality of life can be observed in Winnie mice, as well as extraintestinal manifestations that are often associated with UC. While Winnie mice are currently less studied compared to other more established models of colitis, much has been discovered in the initial years of its use as a UC-like model. In summary, the use of Winnie mice adds to the growing armamentarium that is required to develop precision-based medicine for its future application in treating complex multifactorial diseases, such as UC.
Collapse
Affiliation(s)
- Marcello Chieppa
- Department of Experimental Medicine, University of Salento, 73100 Lecce, Italy
| | - Stefania De Santis
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Giulio Verna
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
9
|
Lin Q, Zhang S, Zhang J, Jin Y, Chen T, Lin R, Lv J, Xu W, Wu T, Tian S, Ying L, Li X, Huang Z, Niu J. Colonic epithelial-derived FGF1 drives intestinal stem cell commitment toward goblet cells to suppress inflammatory bowel disease. Nat Commun 2025; 16:3264. [PMID: 40188210 PMCID: PMC11972292 DOI: 10.1038/s41467-025-58644-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/31/2025] [Indexed: 04/07/2025] Open
Abstract
Understanding the molecular mechanisms that regulate intestinal epithelial cell (IEC) renewal provides potential targets for inflammatory bowel disease (IBD). Growing evidence has highlighted the importance of epithelial signals in regulating intestinal stem cell (ISC) differentiation. However, it remains unclear which IEC-derived cytokines can precisely regulate ISC commitment toward specific mature cells. Here we systematically analyze all fibroblast growth factors (FGFs) expression and find that colonic FGF1 levels are inversely correlated with the severity of IBD in mouse models and patients. IEC-specific Fgf1 deletion leads to impaired goblet cell differentiation and exacerbated colitis, while pharmacological administration of recombinant FGF1 (rFGF1) alleviates colitis by enhancing goblet cell differentiation and improving colonic epithelial integrity. Mechanistic studies reveal that rFGF1 directs ISC differentiation toward goblet cells via FGFR2-TCF4-ATOH1 signaling axis. In conclusion, our study identifies an epithelial niche-derived FGF1 that regulates ISC commitment toward goblet cells, shedding light on strategies for treating IBD.
Collapse
Affiliation(s)
- Qian Lin
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Sudan Zhang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jiaren Zhang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yi Jin
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Taoli Chen
- Department of Pharmacy, The Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang, China
| | - Ruoyu Lin
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Jiaxuan Lv
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wenjing Xu
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Tianzhen Wu
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Shenyu Tian
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Lei Ying
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiaokun Li
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Zhifeng Huang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Jianlou Niu
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
10
|
Valerio de Mello Braga LL, Silva Schiebel C, Simão G, Sauruk da Silva K, dos Santos Maia MH, Vieira Ulysséa Fernardes AC, do Nascimento GE, Cordeiro LMC, Adel Issa T, Gois MB, Fernandes Soares E, Maria-Ferreira D. Type I Arabinogalactan and Methyl-Esterified Homogalacturonan Polysaccharides from Tamarillo ( Solanum betaceum cav.) Fruit Pulp Ameliorate DSS-Induced Ulcerative Colitis. Pharmaceuticals (Basel) 2025; 18:461. [PMID: 40283898 PMCID: PMC12030512 DOI: 10.3390/ph18040461] [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: 01/29/2025] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Inflammatory bowel diseases, such as ulcerative colitis and Crohn's disease, affect the gastrointestinal tract. Treatment aims to induce remission and relieve symptoms but may fail or cause side effects. Recent studies suggest that natural polysaccharides can reduce inflammation and promote healing. The polysaccharides of the pulp of tamarillo (Solanum betaceum cav.) have shown beneficial effects, but their potential in colitis is still unexplored. Objective: To investigate the effect of polysaccharides from tamarillo pulp in an animal model of ulcerative colitis. Methods: Polysaccharides from tamarillo pulp (STWA) were extracted and tested in female mice (BALB/c) to investigate their effect on dextran sodium sulfate (DSS)-induced ulcerative colitis. Different doses of the polysaccharides were tested (10 mg/kg, 30 mg/kg, and 100 mg/kg). The course of the disease and the weight of the animals were monitored daily. At the end of the experimental protocol, the large intestine was removed and measured. Markers of oxidative stress and inflammation were then analyzed. Histological analysis was performed to assess microscopic changes. Results: Treatment with STWA (100 mg/kg) prevented weight loss in mice with DSS-induced colitis and reduced the disease activity index. The colon length was preserved, and occult blood in the feces was reduced. Treatment with STWA controlled oxidative stress. Glutathione S-transferase (GST) levels increased, while lipid peroxidation decreased. The inflammatory process was reduced, as indicated by the decrease in myeloperoxidase (MPO), N-acetylglucosamine (NAG), and tumor necrosis factor alpha (TNF-α) levels and the increase in interleukin 10 (IL-10) levels. STWA also improved the colon histology, while preserving the colonic epithelium. Conclusions: The results suggest that STWA has protective potential and reduces inflammation in an experimental model of ulcerative colitis in mice.
Collapse
Affiliation(s)
- Lara Luisa Valerio de Mello Braga
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (L.L.V.d.M.B.); (C.S.S.); (G.S.); (K.S.d.S.); (M.H.d.S.M.); (E.F.S.)
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Carolina Silva Schiebel
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (L.L.V.d.M.B.); (C.S.S.); (G.S.); (K.S.d.S.); (M.H.d.S.M.); (E.F.S.)
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Gisele Simão
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (L.L.V.d.M.B.); (C.S.S.); (G.S.); (K.S.d.S.); (M.H.d.S.M.); (E.F.S.)
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Karien Sauruk da Silva
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (L.L.V.d.M.B.); (C.S.S.); (G.S.); (K.S.d.S.); (M.H.d.S.M.); (E.F.S.)
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Mateus Henrique dos Santos Maia
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (L.L.V.d.M.B.); (C.S.S.); (G.S.); (K.S.d.S.); (M.H.d.S.M.); (E.F.S.)
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Ana Carolina Vieira Ulysséa Fernardes
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (L.L.V.d.M.B.); (C.S.S.); (G.S.); (K.S.d.S.); (M.H.d.S.M.); (E.F.S.)
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Georgia E. do Nascimento
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná, Curitiba 81531-980, PR, Brazil (L.M.C.C.)
| | - Lucimara Mach Côrtes Cordeiro
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná, Curitiba 81531-980, PR, Brazil (L.M.C.C.)
| | - Tufik Adel Issa
- Faculdade de Ciências da Saúde, Universidade Federal de Rondonópolis, Rondonópolis 78736-900, MT, Brazil (M.B.G.)
| | - Marcelo Biondaro Gois
- Faculdade de Ciências da Saúde, Universidade Federal de Rondonópolis, Rondonópolis 78736-900, MT, Brazil (M.B.G.)
| | - Elizabeth Fernandes Soares
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (L.L.V.d.M.B.); (C.S.S.); (G.S.); (K.S.d.S.); (M.H.d.S.M.); (E.F.S.)
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Daniele Maria-Ferreira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (L.L.V.d.M.B.); (C.S.S.); (G.S.); (K.S.d.S.); (M.H.d.S.M.); (E.F.S.)
- Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| |
Collapse
|
11
|
Zhu L, Luo Y, Liu Y, Sun S, Yuan J, Zhang L, Zhong W, Ma S, Yu Z, Zhou J, Chen X, Zhao J. Clostridium butyricum ameliorates indomethacin-induced enteropathy by promoting MUC2 secretion via suppressing the Notch pathway. Front Microbiol 2025; 16:1509876. [PMID: 40177488 PMCID: PMC11961966 DOI: 10.3389/fmicb.2025.1509876] [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: 10/11/2024] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
Nonsteroidal anti-inflammatory drug (NSAID) enteropathy is a serious clinical complication with no effective treatments available. Modulating the intestinal microbiota through dietary and nutritional targets is a promising strategy for preventing NSAID enteropathy. This study aimed to investigate the protective effect and underlying mechanisms of the probiotic Clostridium butyricum (CB) on indomethacin (IND)-induced enteropathy. C57BL/6J mice received CB treatment for 14 days along with concurrent IND gavage for the final 7 days. Caco2 cells were stimulated with IND to evaluate the effect of CB supernatant (CBS) on the intestinal barrier function, and LS174T cells were used to validate the modulatory action of CBS on the Notch signaling pathway. Our findings revealed that CB treatment prevented anorexia and weight loss, reduced the severity of enteropathy, and decreased the inflammatory response of the small intestine. CB also increased the expression of tight junction proteins and reduced permeability in mice and Caco2 cells. Additionally, CB suppressed apoptosis and promoted proliferation in the small intestine. Further research found that CB increased the number of goblet cells and MUC2 secretion. Mechanistically, CB may promote MUC2 secretion by suppressing the Notch signaling pathway, consistent with the results of intervention in LS174T cells with CBS. In conclusion, CB might prevent NSAID enteropathy by increasing MUC2 secretion through the inhibition of the Notch pathway. Our study identified the potential efficacy of CB as a preventive strategy against NSAID enteropathy and showed promising prospects for CB as a food supplement.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingwen Zhao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
12
|
Yin Q, Li X, Xiong Y, Jiang Y, Ma S, Qian G. Bletilla oligosaccharides improved 5-fluorouracil-induced intestinal mucositis in mice by activating NF-κB signalling pathway and regulating intestinal microbiota. Front Pharmacol 2025; 16:1526274. [PMID: 40183076 PMCID: PMC11965902 DOI: 10.3389/fphar.2025.1526274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/12/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction The Bletilla oligosaccharides (BO) are active compounds extracted from Bletilla striata and have the strong protective effect on the gastrointestinal tract. Chemotherapeutic intestinal mucositis (CIM) is one of the toxic side effects of chemotherapeutic agents on the gastrointestinal tract. The aim of this study was to identify the structure of BO and evaluate the therapeutic effect of BO on 5-fluorouracil-induced intestinal mucosal inflammation. Methods BO were purified from DEAE52 cellulose. The structure of BO were characterised by HPGPC, GC-MS and NMR. In vivo, the mouse model of intestinal mucositis was established by intraperitoneal injection of 5-FU. The effect of BO on intestinal mucositis in mice was detected by assessing the levels of intestinal flora, ZO-1, occludin, and MUC-2, and inflammatory cytokines (IL-1β, IL-6, IL-10, and TNF-α). Results Structural characterisation showed that BO were the neutral polysaccharide composed mainly of glucose and mannose. The backbone of BO consisted of→4)-β-Manp-(1→, →4)-β-Glcp-(1→ and small →3,4)-α-Manp-(1→. The results of the in vivo experiment showed that the symptoms of diarrhoea, haematochezia and colonic mucosal lesions improved after administration of BO. Further experiments showed that BO not only reduced the levels of pro-inflammatory factors such as IL-1β, IL-6 and TNF-α, but also improved the expression of intestinal barrier protein and intestinal microbial community after BO treatment. Conclusion BO can relieve the progress of intestinal mucositis by relieving inflammation, protecting the intestinal epithelial barrier and regulating the intestinal microbiota. These data provide experimental evidence for the application of BO in chemotherapeutic intestinal mucositis.
Collapse
Affiliation(s)
- Qiuxiong Yin
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xinran Li
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yanli Xiong
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yupeng Jiang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shengsuo Ma
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Guoqiang Qian
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
13
|
Chen D, Bi X, Feng Q, Sun Y. Supplementation with Lentil ( Lens culinaris) Hull Soluble Dietary Fiber Ameliorates Sodium Dextran Sulfate-Induced Colitis and Behavioral Deficits via the Gut-Brain Axis. Foods 2025; 14:870. [PMID: 40077572 PMCID: PMC11898428 DOI: 10.3390/foods14050870] [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: 02/08/2025] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
In this study, the impact of lentil hull soluble dietary fibers (SDFs) on colitis and behavioral deficits in mice was assessed. Structural characterizations of SDFs confirmed that cellulase-modified soluble dietary fiber exhibited better physicochemical properties: more porous microstructure; similar polysaccharide structure; more stable particle size distribution; higher crystallinity; better adsorption capacity; and lower viscosity. Additionally, we explored its potential cognitive benefits via the gut-brain axis by behavioral tests, histopathology, 16S rRNA sequencing, gas chromatography and metabolomics analysis. The results showed that SDFs significantly improved inflammatory symptoms in colon and brain and cognitive behaviors. LSDF had better efficacy than HSDF. LSDF intervention decreased the harmful bacteria abundance (Bacteroides, Flexispira and Escherichia, etc.) and increased beneficial bacteria abundance (Aggregatibacter and Helicobacter, etc.). LSDF also affected brain metabolites through the sphingolipid metabolism. Spearman correlation analysis showed that there was a positive correlation between harmful bacteria with inflammatory factors (LPS, IL-1β, IL-6, and TNF-α, etc.) and sphingolipid metabolites, while beneficial bacteria were positively correlated with brain-derived neurotrophic factor (BDNF), IL-10, and cognitive behavior. This study highlights the value of SDFs in future diet-based therapeutic strategies targeting gut-brain interactions.
Collapse
Affiliation(s)
- Dongying Chen
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China;
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China;
| | - Xin Bi
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China;
| | - Qian Feng
- The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China;
| | - Yong Sun
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China;
| |
Collapse
|
14
|
Lim HJ, Park IS, Kim MJ, Seo JW, Ha G, Yang HJ, Jeong DY, Kim SY, Jung CH. Protective Effect of Ganjang, a Traditional Fermented Soy Sauce, on Colitis-Associated Colorectal Cancer in Mice. Foods 2025; 14:632. [PMID: 40002076 PMCID: PMC11854912 DOI: 10.3390/foods14040632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/31/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent causes of cancer-related fatalities globally, and its development is closely associated with dietary and microbial factors. The aim of this study was to investigate the potential of ganjang, a traditional fermented soy sauce, in alleviating colitis-associated colorectal cancer (CAC) in a mouse model induced by azoxymethane/dextran sulfate sodium (AOM/DSS). The bacterial composition of ganjang samples from different regions primarily included Lactobacillus spp. and Bacillus spp. Administration of ganjang to AOM/DSS-induced mice significantly improved CAC-related symptoms, including increased body weight, restored colon length, and reduced spleen size. Additionally, ganjang administration led to a decrease in tumor size and number, the modulation of apoptotic and proliferative markers, decreased inflammatory cytokine levels, and the restoration of the intestinal epithelial barrier. Furthermore, ganjang samples altered the gut microbiota composition by increasing the relative abundance of Lactobacillus spp. These findings suggest that ganjang has potential as a functional food for CRC prevention or mitigation, primarily through the modulation of CAC symptoms, tumor growth, inflammatory responses, intestinal epithelial barrier integrity, and gut microbiota composition.
Collapse
Affiliation(s)
- Hyeon-Ji Lim
- Jeonju AgroBio-Materials Institute, Wonjangdong-gil 111-27, Jeonju 54810, Republic of Korea; (H.-J.L.); (I.-S.P.); (M.J.K.); (S.-Y.K.)
| | - In-Sun Park
- Jeonju AgroBio-Materials Institute, Wonjangdong-gil 111-27, Jeonju 54810, Republic of Korea; (H.-J.L.); (I.-S.P.); (M.J.K.); (S.-Y.K.)
| | - Min Ju Kim
- Jeonju AgroBio-Materials Institute, Wonjangdong-gil 111-27, Jeonju 54810, Republic of Korea; (H.-J.L.); (I.-S.P.); (M.J.K.); (S.-Y.K.)
| | - Ji Won Seo
- Microbial Institute for Fermentation Industry, Sunchang 56048, Republic of Korea; (J.W.S.); (G.H.); (H.-J.Y.); (D.-Y.J.)
| | - Gwangsu Ha
- Microbial Institute for Fermentation Industry, Sunchang 56048, Republic of Korea; (J.W.S.); (G.H.); (H.-J.Y.); (D.-Y.J.)
| | - Hee-Jong Yang
- Microbial Institute for Fermentation Industry, Sunchang 56048, Republic of Korea; (J.W.S.); (G.H.); (H.-J.Y.); (D.-Y.J.)
| | - Do-Youn Jeong
- Microbial Institute for Fermentation Industry, Sunchang 56048, Republic of Korea; (J.W.S.); (G.H.); (H.-J.Y.); (D.-Y.J.)
| | - Seon-Young Kim
- Jeonju AgroBio-Materials Institute, Wonjangdong-gil 111-27, Jeonju 54810, Republic of Korea; (H.-J.L.); (I.-S.P.); (M.J.K.); (S.-Y.K.)
| | - Chan-Hun Jung
- Jeonju AgroBio-Materials Institute, Wonjangdong-gil 111-27, Jeonju 54810, Republic of Korea; (H.-J.L.); (I.-S.P.); (M.J.K.); (S.-Y.K.)
| |
Collapse
|
15
|
van Wijngaarden EW, Arias SL, Rhee M, Silberstein MN, Brito IL. The role of human intestinal mucus in the prevention of microplastic uptake and cell damage. Biomater Sci 2025; 13:1010-1020. [PMID: 39810566 DOI: 10.1039/d4bm01574f] [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: 01/16/2025]
Abstract
An increase in plastic waste and its release into the environment has led to health concerns over microplastics (MPs) in the environment. The intestinal mucosal layer is a key defense mechanism against ingested MPs, preventing the migration of particles to other parts of the body. MP migration through intestinal mucus is challenging to study due to difficulties in obtaining intact mucus layers for testing and numerous formulations, shapes, and sizes of microplastics. Previous studies have primarily used mucus from animals, hydrogel models, and mucus samples from other parts of the body as substitutes. This study examines how different MP compositions, sizes (40-500 nm), and surface functionalizations alter MP migration through human intestinal mucus; how the mucus layer protects cells from MP uptake, toxicity, and inflammation; and how the intestinal mucus prevents the migration of other environmental toxins via MP particles. The presence of a mucus layer also provides critical protection against cytotoxicity, reactive oxygen species production, and uptake for all particles tested, although certain functionalizations, such as streptavidin, are particularly harmful to cells with high toxicity and inflammation. Understanding the properties that assist of impede the diffusion of MPs through mucus is relevant to the overall bioaccumulation and health effects of MPs as well as drug delivery purposes.
Collapse
Affiliation(s)
- Ellen W van Wijngaarden
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY 14850, USA
- Engineered Living Materials Institute, Cornell University, Ithaca, NY 14850, USA.
| | - Sandra L Arias
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Matthew Rhee
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Meredith N Silberstein
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY 14850, USA
- Engineered Living Materials Institute, Cornell University, Ithaca, NY 14850, USA.
| | - Ilana L Brito
- Engineered Living Materials Institute, Cornell University, Ithaca, NY 14850, USA.
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
16
|
Tu W, Wang H, Zhang Y, Huang J, Diao Y, Zhou J, Tan Y, Li X. Investigation of the Molecular Mechanism of Asthma in Meishan Pigs Using Multi-Omics Analysis. Animals (Basel) 2025; 15:200. [PMID: 39858200 PMCID: PMC11759154 DOI: 10.3390/ani15020200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Asthma has been extensively studied in humans and animals, but the molecular mechanisms underlying asthma in Meishan pigs, a breed with distinct genetic and physiological characteristics, remain elusive. Understanding these mechanisms could provide insights into veterinary medicine and human asthma research. We investigated asthma pathogenesis in Meishan pigs through transcriptomic and metabolomic analyses of blood samples taken during autumn and winter. Asthma in Meishan pigs is related to inflammation, mitochondrial oxidative phosphorylation, and tricarboxylic acid (TCA) cycle disorders. Related genes include CXCL10, CCL8, CCL22, CCL21, OLR1, and ACKR1, while metabolites include succinic acid, riboflavin-5-phosphate, and fumaric acid. Transcriptomic sequencing was performed on panting and normal Meishan pigs, and differentially expressed genes underwent functional enrichment screening. Metabolomic analysis revealed differential metabolites and pathways between groups. Combined analyses indicated that lung inflammation is influenced by genetic, allergenic, and environmental factors disrupting oxidative phosphorylation in lung mitochondria, affecting the TCA cycle. Mitochondrial reactive oxygen species, glutathione S-transferases, arginase 1 and RORC in immune regulation, the Notch pathway, YPEL4 in cell proliferation, and MARCKS in airway mucus secretion play roles in asthma pathogenesis. This study highlights that many cytokines and signaling pathways contribute to asthma. Further studies are needed to elucidate their complex interactions.
Collapse
Affiliation(s)
- Weilong Tu
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (W.T.); (H.W.); (Y.Z.); (J.H.); (Y.D.); (J.Z.)
- Key Laboratory of Livestock and Poultry Resources (Pig) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
- Shanghai Engineering Research Center of Breeding Pig, Shanghai 201106, China
| | - Hongyang Wang
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (W.T.); (H.W.); (Y.Z.); (J.H.); (Y.D.); (J.Z.)
- Key Laboratory of Livestock and Poultry Resources (Pig) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
- Shanghai Engineering Research Center of Breeding Pig, Shanghai 201106, China
| | - Yingying Zhang
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (W.T.); (H.W.); (Y.Z.); (J.H.); (Y.D.); (J.Z.)
- Key Laboratory of Livestock and Poultry Resources (Pig) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
- Shanghai Engineering Research Center of Breeding Pig, Shanghai 201106, China
| | - Ji Huang
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (W.T.); (H.W.); (Y.Z.); (J.H.); (Y.D.); (J.Z.)
- Key Laboratory of Livestock and Poultry Resources (Pig) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
- Shanghai Engineering Research Center of Breeding Pig, Shanghai 201106, China
| | - Yuduan Diao
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (W.T.); (H.W.); (Y.Z.); (J.H.); (Y.D.); (J.Z.)
- Key Laboratory of Livestock and Poultry Resources (Pig) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
- Shanghai Engineering Research Center of Breeding Pig, Shanghai 201106, China
| | - Jieke Zhou
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (W.T.); (H.W.); (Y.Z.); (J.H.); (Y.D.); (J.Z.)
- Key Laboratory of Livestock and Poultry Resources (Pig) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
- Shanghai Engineering Research Center of Breeding Pig, Shanghai 201106, China
| | - Yongsong Tan
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (W.T.); (H.W.); (Y.Z.); (J.H.); (Y.D.); (J.Z.)
- Key Laboratory of Livestock and Poultry Resources (Pig) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Shanghai 201106, China
- Shanghai Engineering Research Center of Breeding Pig, Shanghai 201106, China
| | - Xin Li
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai 201106, China; (W.T.); (H.W.); (Y.Z.); (J.H.); (Y.D.); (J.Z.)
| |
Collapse
|
17
|
Mansouri P, Mansouri P, Behmard E, Najafipour S, Kouhpayeh A, Farjadfar A. Novel targets for mucosal healing in inflammatory bowel disease therapy. Int Immunopharmacol 2025; 144:113544. [PMID: 39571265 DOI: 10.1016/j.intimp.2024.113544] [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: 06/01/2024] [Revised: 10/13/2024] [Accepted: 10/28/2024] [Indexed: 12/15/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic condition affecting the gastrointestinal tract, primarily manifesting as ulcerative colitis (UC) or Crohn's disease (CD). Both inflammation and disruption of the intestinal epithelial barrier are key factors in IBD pathogenesis. Substantial evidence has revealed a significant association between aberrant immune responses and impairment of the intestinal epithelial barrier in IBD pathogenesis. The components of the intestinal epithelium, particularly goblet cells and Paneth cells, are crucial to gut homeostasis, as they secrete mucin, antimicrobial peptides (AMPs), and cytokines. Furthermore, impairment of epithelial integrity, which is regulated by tight junctions, is a hallmark of IBD pathology. While common treatments for IBD, such as anti-inflammatory drugs, target various signaling pathways with varying efficacies, therapeutic approaches focused on mucosal and epithelial barrier healing have been largely neglected. Moreover, high costs, side effects, and insufficient or inconsistent therapeutic outcomes remain major drawbacks of conventional anti-IBD drugs. Recent studies on epithelial barrier regeneration and permeability reduction have introduced promising therapeutic targets, including farnesoid X receptor (FXR), urokinase-type plasminogen activator (uPA)-urokinase-type plasminogen activator receptor (uPAR) interaction, fecal microbiota transplantation (FMT), and insulin receptor (INSR). Notably, the simultaneous targeting of intestinal inflammation and promotion of epithelial barrier healing shows promise for efficient IBD treatment. Future research should explore targeted therapies and combination treatments, including natural remedies, microbiota colonization, stem cell approaches, and computer-aided drug design. It is also crucial to focus on accurate prognosis and developing a thorough understanding of IBD development mechanisms.
Collapse
Affiliation(s)
- Pardis Mansouri
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran; Department of Medical Biotechnology, Fasa University of Medical Sciences, Fasa, Iran
| | - Pegah Mansouri
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran; Department of Medical Biotechnology, Fasa University of Medical Sciences, Fasa, Iran
| | - Esmaeil Behmard
- School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran; Zarrin Avaye Kowsar Salamat (ZAX Company), Fasa, Iran
| | - Sohrab Najafipour
- School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran; Zarrin Avaye Kowsar Salamat (ZAX Company), Fasa, Iran
| | - Amin Kouhpayeh
- Department of Pharmacology, Faculty of Medicine, Fasa University of Medical Sciences, Fasa, Iran; Zarrin Avaye Kowsar Salamat (ZAX Company), Fasa, Iran.
| | - Akbar Farjadfar
- Department of Medical Biotechnology, Fasa University of Medical Sciences, Fasa, Iran; Zarrin Avaye Kowsar Salamat (ZAX Company), Fasa, Iran.
| |
Collapse
|
18
|
Kanika, Ahmad A, Kumar A, Rahul, Mishra RK, Ali N, Navik U, Parvez S, Khan R. Leveraging thiol-functionalized biomucoadhesive hybrid nanoliposome for local therapy of ulcerative colitis. Biomaterials 2025; 312:122747. [PMID: 39142219 DOI: 10.1016/j.biomaterials.2024.122747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/06/2024] [Accepted: 08/04/2024] [Indexed: 08/16/2024]
Abstract
Directly administering medication to inflamed intestinal sites for treating ulcerative colitis (UC), poses significant challenges like retention time, absorption variability, side effects, drug stability, and non-specific delivery. Recent advancements in therapy to treat colitis aim to improve local drug availability that is enema therapy at the site of inflammation, thereby reducing systemic adverse effects. Nevertheless, a key limitation lies in enemas' inability to sustain medication in the colon due to rapid peristaltic movement, diarrhea, and poor local adherence. Therefore, in this work, we have developed site-specific thiolated mucoadhesive anionic nanoliposomes to overcome the limitations of conventional enema therapy. The thiolated delivery system allows prolonged residence of the delivery system at the inflamed site in the colon, confirmed by the adhesion potential of thiolated nanoliposomes using in-vitro and in-vivo models. To further provide therapeutic efficacy thiolated nanoliposomes were loaded with gallic acid (GA), a natural compound known for its antibacterial, antioxidant, and potent anti-inflammatory properties. Consequently, Gallic Acid-loaded Thiolated 2,6 DALP DMPG (GATh@APDL) demonstrates the potential for targeted adhesion to the inflamed colon, facilitated by their small size 100 nm and anionic nature. Therapeutic studies indicate that this formulation offers protective effects by mitigating colonic inflammation, downregulating the expression of NF-κB, HIF-1α, and MMP-9, and demonstrating superior efficacy compared to the free GA enema. The encapsulated GA inhibits the NF-κB expression, leading to enhanced expression of MUC2 protein, thereby promoting mucosal healing in the colon. Furthermore, GATh@APDL effectively reduces neutrophil infiltration and regulates immune cell quantification in colonic lamina propria. Our findings suggest that GATh@APDL holds promise for alleviating UC and addressing the limitations of conventional enema therapy.
Collapse
Affiliation(s)
- Kanika
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali, Punjab, 140306, India
| | - Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N4N1, Canada
| | - Ajay Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali, Punjab, 140306, India
| | - Rahul
- Department of Chemistry, Malaviya National Institute of Technology, Jaipur, Rajasthan, 302017, India
| | - Rakesh Kumar Mishra
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, India
| | - Nemat Ali
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda, Ghudda, Punjab, 151401, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali, Punjab, 140306, India.
| |
Collapse
|
19
|
Bingöl FG, Ağagündüz D, Budán F. Probiotic Bacterium-Derived p40, p75, and HM0539 Proteins as Novel Postbiotics and Gut-Associated Immune System (GAIS) Modulation: Postbiotic-Gut-Health Axis. Microorganisms 2024; 13:23. [PMID: 39858791 PMCID: PMC11767761 DOI: 10.3390/microorganisms13010023] [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: 12/08/2024] [Revised: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
It is known that probiotics have direct and indirect effects on many systems in the body, especially the gastrointestinal system. Interest in using probiotic strain-derived cell components and metabolites has also increased as a result of the significant benefits of probiotics. Although many terminologies and definitions are used for these components and metabolites, the International Scientific Association of Probiotics and Prebiotics (ISAPP) recommended the use of the term postbiotic in 2021, which is defined as "a preparation of inanimate microorganisms and/or their components that confers a health benefit on the host". Postbiotics are bioactive metabolites such as organic acids, peptides/proteins, cell wall components, functional enzymes, short-chain fatty acids, vitamins, and phenols. These molecules mediate many positive effects such as immunomodulatory, antimicrobial, and antioxidant effects. These positive effects on maintaining health have enabled the identification of many new postbiotic proteins such as p40, p75, and HM0539. In this review, the postbiotic proteins p40, p75, and HM0539 derived from lactobacilli and their functional effects are systematically summarized. The p40 protein, in particular, has been shown to support gut barrier activity and reduce inflammation, potentially through mechanisms involving epidermal growth factor receptor-dependent signaling. Additionally, p40 and p75 proteins exhibit protective effects on intestinal epithelial tight junctions, suggesting their therapeutic potential in preventing intestinal damage and diseases such as colitis. HM0539 enhances intestinal barrier integrity, exhibits antiinflammatory properties, and protects against bacterial infection, suggesting its possible as a therapeutic for inflammatory bowel disease. This review may contribute to future studies on the therapeutic use of p40, p75, and HM0539 postbiotic proteins in inflammatory gastrointestinal system diseases.
Collapse
Affiliation(s)
- Feray Gençer Bingöl
- Department of Nutrition and Dietetics, Faculty of Health Science, Burdur Mehmet Akif Ersoy University, 15200 Burdur, Türkiye;
| | - Duygu Ağagündüz
- Department of Nutrition and Dietetics, Faculty of Health Science, Gazi University, 06490 Ankara, Türkiye;
| | - Ferenc Budán
- Institute of Physiology, Medical School, University of Pécs, H-7624 Pécs, Hungary
| |
Collapse
|
20
|
Zhong Y, Chang X, Zhao Z, Zheng L, Kuang G, Li P, Liu C, Fan Y, Liang Z, Zhuang K, Xie Q, Liu Y. Bacteroides fragilis capsular polysaccharide A ameliorates ulcerative colitis in rat by recovering intestinal barrier integrity and restoring gut microbiota. Front Pharmacol 2024; 15:1402465. [PMID: 39776580 PMCID: PMC11703662 DOI: 10.3389/fphar.2024.1402465] [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: 03/17/2024] [Accepted: 10/02/2024] [Indexed: 01/11/2025] Open
Abstract
Bacteroides fragilis (B. fragilis) is a Gram-negative, obligate anaerobic, commensal bacterium residing in the human gut and holds therapeutic potential for ulcerative colitis (UC). Previous studies have indicated that capsular polysaccharide A (PSA) of B. fragilis is a crucial component for its effectiveness, possessing various biological activities such as anti-inflammatory, anti-tumor, and immune-modulating effects. We previously isolated and characterized the B. fragilis strain ZY-312 from the feces of a healthy breastfed infant, and extracted its PSA, named TP2. In this study, we explored the impact of TP2 on colonic inflammation and delved into its potential mechanisms. Initially, we used 2,4,6-trinitrobenzenesulfonic acid (TNBS) to induce colitis in rats and found that TP2 treatment significantly ameliorated TNBS-induced weight loss, increased clinical scores, extensive ulcers, and intestinal epithelial damage in UC rats. Further analysis revealed that TP2 effectively restored the intestinal barrier integrity in UC rats by regulating the expression of Muc-2, tight junction proteins (ZO-1, occludin, claudin-1, and claudin-2), as well as apoptosis-related proteins Bcl-2, BAX, and Cleaved-Caspase-3. Additionally, TP2 suppressed the expression of pro-inflammatory cytokines TNF-α, IL-1β, IL-6, and IL23, while promoting the secretion of anti-inflammatory cytokines IL-10 and IL-22, thereby inhibiting the occurrence of inflammation. TP2 also downregulated the phosphorylation levels of AKT and PI3K, effectively inhibiting the abnormal activation of the PI3K/AKT signaling pathway. More interestingly, 16S rRNA sequencing results showed that TP2 restored the ecological imbalance of the rat intestinal microbiota, with an increase in beneficial bacteria such as Lactobacillus and Limosilactobacillus observed in the treatment group. In conclusion, TP2 through the regulation of intestinal barrier-related cells and proteins, inhibition of apoptosis, modulation of inflammation-related cytokine levels, and control of abnormal activation of the PI3K/AKT signaling pathway, restores intestinal barrier integrity. Additionally, by reshaping the ecological imbalance of the gut microbiota, TP2 ultimately alleviates ulcerative colitis in rats.
Collapse
Affiliation(s)
- Yijia Zhong
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xiujuan Chang
- Guangzhou ZhiYi Biotechnology Co. Ltd., Guangzhou, China
| | - Zihan Zhao
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Lijun Zheng
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Gaobo Kuang
- Guangzhou ZhiYi Biotechnology Co. Ltd., Guangzhou, China
| | - Ping Li
- Guangzhou ZhiYi Biotechnology Co. Ltd., Guangzhou, China
| | | | - Yuqin Fan
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zhixuan Liang
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Ke Zhuang
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Qiuling Xie
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yangyang Liu
- Guangzhou ZhiYi Biotechnology Co. Ltd., Guangzhou, China
| |
Collapse
|
21
|
Shin HY, Jeong WB, Joung MY, Shin KS, Yu KW. Effects of Centella asiatica-isolated pectic polysaccharide on dextran sulfate sodium-induced colitis. Int J Biol Macromol 2024; 285:138237. [PMID: 39622374 DOI: 10.1016/j.ijbiomac.2024.138237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/15/2024] [Accepted: 11/29/2024] [Indexed: 12/08/2024]
Abstract
Dietary supplementation of polysaccharides demonstrates strong therapeutic actions on inflammatory bowel disease (IBD). Centella asiatica (CA) has traditionally been used in Ayurveda and Chinese medicine. However, the effects of CA-isolated pectic polysaccharides on IBD remain unknown. This study determined the effect of a CA-isolated pectic polysaccharide on IBD. The crude polysaccharide (CA-CP), isolated from a hot-water extract of CA, is a typical pectic polysaccharide composed mainly of galacturonic acid (40.6 %), galactose (27.6 %), arabinose (13.5 %), and rhamnose (8.5 %). CA-CP improved clinical symptoms in a DSS-induced colitis murine model, including weight change (9.9-12.0 %), disease activity index (31.6-51.9 %), colon length (13.2-21.5 %), and spleen weight (21.8-26.3 %). CA-CP effectively regulated the levels of inflammatory and junctional factors by mediating the MAPK and NF-κB pathways. CA-CP partially alleviated DSS-induced crypt destruction, submucosal edema, inflammatory infiltration, and mucin secretion. The content of total short-chain fatty acids increased substantially (cecum 29.8-53.7 %, feces 75.4-109.3 %) with oral CA-CP administration compared to the DSS group. Cecal microbial community analysis revealed that CA-CP administration regulated DSS-induced colitis by reducing the abundance of Escherichia (5.0-10.9 %) and Clostridium (0.3-0.4 %), while increasing the abundance of Bacteroidetes (6.8-8.5 %), Ligilactobacillus (2.7-6.0 %), and Bilophilia (0.3-0.6 %). The findings provide fundamental data for developing novel functional therapeutic agents for the prevention and treatment of colitis, using CA-isolated pectic polysaccharides. Furthermore, to the best of our knowledge, our study is the first to demonstrate the effects of pectic polysaccharides isolated from CA on IBD.
Collapse
Affiliation(s)
- Hyun Young Shin
- Transdisciplinary Major in Learning Health Systems, Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea.
| | - Won Bi Jeong
- Major in Food & Nutrition, Korea National University of Transportation, Chungbuk 27909, Republic of Korea.
| | - Mi Yeun Joung
- Corporation ChamSunJin Green Juice, Jincheon 27865, Republic of Korea.
| | - Kwang-Soon Shin
- Department of Food Science and Biotechnology, Kyonggi University, Suwon 16227, Republic of Korea.
| | - Kwang-Won Yu
- Major in Food & Nutrition, Korea National University of Transportation, Chungbuk 27909, Republic of Korea.
| |
Collapse
|
22
|
Zhao C, Wang M, Li T, Li D, Feng Y, Wang Y, Qu L, Barcenas AR, Serrano BR, Shen M, Zhao W. Effects of 1-Deoxynojirimycin Extracts of Mulberry Leaves on Oxidative Stress and the Function of the Intestinal Tract in Broilers Induced by H 2O 2. Animals (Basel) 2024; 14:3319. [PMID: 39595371 PMCID: PMC11591279 DOI: 10.3390/ani14223319] [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: 10/16/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
The poultry industry struggles with oxidative stress affecting gut health and productivity. This study examined using 1-Deoxynojirimycin (DNJ) extracts from mulberry leaves as an antioxidant in broilers feed to combat this issue. We divided 240 broilers, aged 16 days, into six groups, including a control and groups exposed to oxidative stress through H2O2 injections, with different supplement levels of DNJ-E (40, 80, 120, and 160 mg/kg of the basal diet) lasting until the broilers reached 42 days old. We evaluated intestinal morphology, ultrastructure, oxidative stress markers, the tight junction, and inflammatory cytokines. Adding 40 mg/kg DNJ-E improved villus height, the villus-to-crypt ratio, and cellular ultrastructure, and increased SOD levels in the jejunum and ileum, as well as CAT levels in the duodenum and jejunum (p < 0.05), compared to the H2O2 group. The addition of DNJ had differential effects on oxidative stress, the intestinal barrier, and immune-related genes. Importantly, the dosages of 40 mg/kg and 80 mg/kg resulted in an upregulation of MUC2 mRNA expression (p < 0.05). These findings suggest that DNJ-E holds potential as a beneficial feed additive for enhancing broiler health, particularly at supplementation levels below 80 mg/kg, as higher concentrations may negatively influence intestinal health. Future investigations should aim to elucidate the underlying mechanisms through which DNJ-E operates within the avian gastrointestinal system.
Collapse
Affiliation(s)
- Chengfeng Zhao
- Jiangsu Key Laboratory of Sericultural Sericulture and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (C.Z.); (M.W.); (T.L.); (D.L.); (Y.F.); (Y.W.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Mingzhu Wang
- Jiangsu Key Laboratory of Sericultural Sericulture and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (C.Z.); (M.W.); (T.L.); (D.L.); (Y.F.); (Y.W.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Tao Li
- Jiangsu Key Laboratory of Sericultural Sericulture and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (C.Z.); (M.W.); (T.L.); (D.L.); (Y.F.); (Y.W.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Dehui Li
- Jiangsu Key Laboratory of Sericultural Sericulture and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (C.Z.); (M.W.); (T.L.); (D.L.); (Y.F.); (Y.W.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Yuan Feng
- Jiangsu Key Laboratory of Sericultural Sericulture and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (C.Z.); (M.W.); (T.L.); (D.L.); (Y.F.); (Y.W.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Yuhua Wang
- Jiangsu Key Laboratory of Sericultural Sericulture and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (C.Z.); (M.W.); (T.L.); (D.L.); (Y.F.); (Y.W.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Liang Qu
- Jiangsu Institute of Poultry Science, Chinese Academy of Agricultural Sciences, Yangzhou 225125, China;
| | - Adileidys Ruiz Barcenas
- Plant Protein and Bionatural Products Research Center, Ministry of Agriculture, Havana 999075, Cuba; (A.R.B.); (B.R.S.)
| | - Boris Ramos Serrano
- Plant Protein and Bionatural Products Research Center, Ministry of Agriculture, Havana 999075, Cuba; (A.R.B.); (B.R.S.)
| | - Manman Shen
- Jiangsu Key Laboratory of Sericultural Sericulture and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (C.Z.); (M.W.); (T.L.); (D.L.); (Y.F.); (Y.W.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Weiguo Zhao
- Jiangsu Key Laboratory of Sericultural Sericulture and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (C.Z.); (M.W.); (T.L.); (D.L.); (Y.F.); (Y.W.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| |
Collapse
|
23
|
Shaban SF, Abdel-Fattah EA, Ali MM, Dessouky AA. The therapeutic efficacy of adipose mesenchymal stem cell-derived microvesicles versus infliximab in a dextran sodium sulfate induced ulcerative colitis rat model. Ultrastruct Pathol 2024; 48:526-549. [PMID: 39545690 DOI: 10.1080/01913123.2024.2426566] [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: 08/18/2024] [Revised: 10/25/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
Ulcerative colitis (UC) is a chronic relapsing intestinal inflammation that is becoming of increasing incidence worldwide and has insufficient treatment. Therefore, finding effective therapies remains a priority. A dextran sodium sulfate colitis model was established to elucidate colonic layers alterations and compare adipose mesenchymal stem cell-derived microvesicles (MSC-MVs) versus infliximab (IFX) efficacy through biochemical, light, and electron microscope studies. Fifty-four rats were allocated to 4 groups: Control (Con), UC, UC+IFX, and UC+MSC-MVs groups. End body weights (BW) and serum malondialdehyde (MDA) levels were recorded. Colitis severity was estimated by disease activity index (DAI). Colonic specimens were processed to evaluate the histological structure, collagen content, surface mucous and goblet cells, CD44, TNF-α, and GFAP immune expression. Morphometric and statistical analyses were performed. The UC group revealed congested, stenosed colons, a significant decline in end BW, and a significant increase in serum MDA and DAI. Furthermore, disturbed histoarchitecture, inflammatory infiltration, depletion of surface mucous and goblet cells, increased collagen, and TNF-α expression and decreased GFAP expression were observed. Alterations were partially attenuated by IFX therapy, whereas MSC-MVs significantly improved all parameters. In conclusion, MSC-MVs were a superior therapeutic option, via attenuating oxidative stress and inflammatory infiltration, in addition to restoring intestinal epithelial integrity and mucosal barrier.
Collapse
Affiliation(s)
- Sahar F Shaban
- Department of Medical Histology and Cell Biology, Medicine, Faculty of medicine, Zagazig University, Zagazig city, Egypt
| | - Eman A Abdel-Fattah
- Department of Medical Histology and Cell Biology, Medicine, Faculty of medicine, Zagazig University, Zagazig city, Egypt
| | - Manar M Ali
- Department of Medical Histology and Cell Biology, Medicine, Faculty of medicine, Zagazig University, Zagazig city, Egypt
| | - Arigue A Dessouky
- Department of Medical Histology and Cell Biology, Medicine, Faculty of medicine, Zagazig University, Zagazig city, Egypt
| |
Collapse
|
24
|
Wang X, Wang P, Li Y, Guo H, Wang R, Liu S, Qiu J, Wang X, Hao Y, Zhao Y, Liao H, Zou Z, Thinwa J, Liu R. Procyanidin C1 Modulates the Microbiome to Increase FOXO1 Signaling and Valeric Acid Levels to Protect the Mucosal Barrier in Inflammatory Bowel Disease. ENGINEERING 2024; 42:108-120. [DOI: 10.1016/j.eng.2023.10.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
25
|
Wang YN, Zhai XY, Wang Z, Gao CL, Mi SC, Tang WL, Fu XM, Li HB, Yue LF, Li PF, Xi SY. Jianpi-Huatan-Huoxue-Anshen formula ameliorates gastrointestinal inflammation and microecological imbalance in chemotherapy-treated mice transplanted with H22 hepatocellular carcinoma. World J Gastrointest Oncol 2024; 16:4209-4231. [DOI: 10.4251/wjgo.v16.i10.4209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 08/06/2024] [Accepted: 09/03/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND Jianpi-Huatan-Huoxue-Anshen formula [Tzu-Chi cancer-antagonizing & life-protecting II decoction (TCCL)] is a Chinese medical formula that has been clinically shown to reduce the gastrointestinal side effects of chemotherapy in cancer patients and improve their quality of life. However, its effect and mechanism on the intestinal microecology after chemotherapy are not yet clear.
AIM To discover the potential mechanisms of TCCL on gastrointestinal inflammation and microecological imbalance in chemotherapy-treated mice transplanted with hepatocellular carcinoma (HCC).
METHODS Ninety-six mice were inoculated subcutaneously with HCC cells. One week later, the mice received a large dose of 5-fluorouracil by intraperitoneal injection to establish a HCC chemotherapy model. Thirty-six mice were randomly selected before administration, and feces, ileal tissue, and ileal contents were collected from each mouse. The remaining mice were randomized into normal saline, continuous chemotherapy, Yangzheng Xiaoji capsules-treated, and three TCCL-treated groups. After treatment, feces, tumors, liver, spleen, thymus, stomach, jejunum, ileum, and colon tissues, and ileal contents were collected. Morphological changes, serum levels of IL-1β, IL-6, IL-8, IL-10, IL-22, TNF-α, and TGF-β, intestinal SIgA, and protein and mRNA expression of ZO-1, NF-κB, Occludin, MUC-2, Claudin-1, and IκB-α in colon tissues were documented. The effect of TCCL on the abundance and diversity of intestinal flora was analyzed using 16S rDNA sequencing.
RESULTS TCCL treatment improved thymus and spleen weight, thymus and spleen indexes, and body weight, decreased tumor volumes and tumor tissue cell density, and alleviated injury to gastric, ileal, and colonic mucosal tissues. Among proteins and genes associated with inflammation, IL-10, TGF-β, SIgA, ZO-1, MUC-2, and Occludin were upregulated, whereas NF-κB, IL-1β, IL-6, TNF-α, IL-22, IL-8, and IκB-α were downregulated. Additionally, TCCL increased the proportions of fecal Actinobacteria, AF12, Adlercreutzia, Clostridium, Coriobacteriaceae, and Paraprevotella in the intermediate stage of treatment, decreased the proportions of Mucipirillum, Odoribacter, RF32, YS2, and Rikenellaceae but increased the proportions of p_Deferribacteres and Lactobacillus at the end of treatment. Studies on ileal mucosal microbiota showed similar findings. Moreover, TCCL improved community richness, evenness, and the diversity of fecal and ileal mucosal flora.
CONCLUSION TCCL relieves pathological changes in tumor tissue and chemotherapy-induced gastrointestinal injury, potentially by reducing the release of pro-inflammatory factors to repair the gastrointestinal mucosa, enhancing intestinal barrier function, and maintaining gastrointestinal microecological balance. Hence, TCCL is a very effective adjuvant to chemotherapy.
Collapse
Affiliation(s)
- Ya-Nan Wang
- Department of TCM, Xiang’an Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Xiang-Yang Zhai
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Zheng Wang
- Department of TCM, Xiang’an Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Chun-Ling Gao
- Department of Radiotherapy, Chenggong Hospital of Xiamen University, PLA 73rd Army Hospital, Xiamen 361003, Fujian Province, China
| | - Sui-Cai Mi
- Department of Oncology, Xiamen Hospital of Traditional Chinese Medicine, Xiamen 361015, Fujian Province, China
| | - Wen-Li Tang
- Department of TCM, Xiang’an Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Xue-Min Fu
- Department of TCM, Xiang’an Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Huai-Bang Li
- Department of TCM, Xiang’an Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Li-Feng Yue
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Peng-Fei Li
- Department of TCM, Xiang’an Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Sheng-Yan Xi
- Department of TCM, Xiang’an Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| |
Collapse
|
26
|
Cunha AF, Delou JM, Barbosa PS, Conceição JSM, Souza KCS, Chagas V, Soletti RC, de Souza HSP, Borges HL. Trp53 Deletion Promotes Exacerbated Colitis, Facilitates Lgr5+ Cancer Stem Cell Expansion, and Fuels Tumorigenesis in AOM/DSS-Induced Colorectal Cancer. Int J Mol Sci 2024; 25:10953. [PMID: 39456736 PMCID: PMC11507199 DOI: 10.3390/ijms252010953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Colorectal cancer CRC remains one of the leading causes of cancer-related deaths worldwide, with chronic intestinal inflammation identified as a major risk factor. Notably, the tumor suppressor TP53 undergoes mutation at higher rates and earlier stages during human inflammation-driven colon tumorigenesis than in sporadic cases. We investigated whether deleting Trp53 affects inflammation-induced tumor growth and the expression of Lgr5+ cancer stem cells in mice. We examined azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced colon tumorigenesis in wild-type Trp53 (+/+), heterozygous (+/-), and knockout (-/-) mice. Trp53-/- mice showed increased sensitivity to DSS colitis and earlier accelerated tumorigenesis with 100% incidence. All groups could develop invasive tumors, but knockouts displayed the most aggressive features. Unlike wild-type CRC, knockouts selectively showed increased populations of Lgr5+ colon cancer stem-like cells. Trp53 loss also boosted laminin, possibly facilitating the disruption of the tumor border. This study highlights how Trp53 deletion promotes the perfect storm of inflammation and stemness, driving colon cancer progression. Trp53 deletion dramatically shortened AOM/DSS latency and improved tumor induction efficiency, offering an excellent inflammation-driven CRC model.
Collapse
Affiliation(s)
- Anderson F. Cunha
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-90, RJ, Brazil; (A.F.C.); (J.M.D.); (P.S.B.); (J.S.M.C.)
- Instituto D’Or de Ensino e Pesquisa, Rio de Janeiro 22281-100, RJ, Brazil
| | - João M. Delou
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-90, RJ, Brazil; (A.F.C.); (J.M.D.); (P.S.B.); (J.S.M.C.)
- Instituto D’Or de Ensino e Pesquisa, Rio de Janeiro 22281-100, RJ, Brazil
| | - Pedro S. Barbosa
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-90, RJ, Brazil; (A.F.C.); (J.M.D.); (P.S.B.); (J.S.M.C.)
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, RJ, Brazil; (K.C.S.S.); (H.S.P.d.S.)
| | - Julia S. M. Conceição
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-90, RJ, Brazil; (A.F.C.); (J.M.D.); (P.S.B.); (J.S.M.C.)
| | - Karen C. S. Souza
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, RJ, Brazil; (K.C.S.S.); (H.S.P.d.S.)
| | - Vera Chagas
- Departamento de Patologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, RJ, Brazil;
| | - Rossana C. Soletti
- Departamento Interdisciplinar, Universidade Federal do Rio Grande do Sul, Tramandaí 95590-000, RS, Brazil;
| | - Heitor S. P. de Souza
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-913, RJ, Brazil; (K.C.S.S.); (H.S.P.d.S.)
| | - Helena L. Borges
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-90, RJ, Brazil; (A.F.C.); (J.M.D.); (P.S.B.); (J.S.M.C.)
| |
Collapse
|
27
|
Wang X, Peng J, Cai P, Xia Y, Yi C, Shang A, Akanyibah FA, Mao F. The emerging role of the gut microbiota and its application in inflammatory bowel disease. Biomed Pharmacother 2024; 179:117302. [PMID: 39163678 DOI: 10.1016/j.biopha.2024.117302] [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: 06/19/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/22/2024] Open
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a complex disorder with an unknown cause. However, the dysbiosis of the gut microbiome has been found to play a role in IBD etiology, including exacerbated immune responses and defective intestinal barrier integrity. The gut microbiome can also be a potential biomarker for several diseases, including IBD. Currently, conventional treatments targeting pro-inflammatory cytokines and pathways in IBD-associated dysbiosis do not yield effective results. Other therapies that directly target the dysbiotic microbiome for effective outcomes are emerging. We review the role of the gut microbiome in health and IBD and its potential as a diagnostic, prognostic, and therapeutic target for IBD. This review also explores emerging therapeutic advancements that target gut microbiome-associated alterations in IBD, such as nanoparticle or encapsulation delivery, fecal microbiota transplantation, nutritional therapies, microbiome/probiotic engineering, phage therapy, mesenchymal stem cells (MSCs), gut proteins, and herbal formulas.
Collapse
Affiliation(s)
- Xiu Wang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu 222006, China
| | - Jianhua Peng
- The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, Jiangsu 212300, China
| | - Peipei Cai
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yuxuan Xia
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Chengxue Yi
- School of Medical Technology, Zhenjiang College, Zhenjiang 212028, China
| | - Anquan Shang
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu 222006, China
| | - Francis Atim Akanyibah
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu 222006, China.
| |
Collapse
|
28
|
Zhao X, Yuan W, Yang L, Yan F, Cui D. Ginsenoside Rh2 suppresses ferroptosis in ulcerative colitis by targeting specific protein 1 by upregulating microRNA-125a-5p. Eur J Med Res 2024; 29:450. [PMID: 39223620 PMCID: PMC11370063 DOI: 10.1186/s40001-024-02025-w] [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: 12/06/2023] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Worldwide, ulcerative colitis (UC) is becoming increasingly fast growing. Ginsenoside Rh2 has been reported to alleviate UC. However, the latent biological mechanism of Rh2 in the treatment of UC remains uncertain. In this study, the goal was to determine the therapeutic effect of Rh2 on dextran sulfate sodium (DSS)-induced UC. METHODS A DSS-induced UC mouse model was established and divided into 7 groups for Rh2 gavage and/or miR-125a-5p lentivirus injection (n = 10 per group). Colonic specimens were collected for phenotypic and pathological analysis. miR-125a-5p and specific protein 1 (SP1) expression, inflammation-related factors IL-6 and IL-10, and apoptosis were detected in mice. Human normal colon epithelial cell line NCM460 was treated with H2O2 and ferric chloride hexahydrate to construct an in vitro cell model of colitis and induce ferroptosis. Independent sample t-test was used to compare cell proliferation, cell entry, apoptosis, and oxidative stress between the two groups. One way analysis of variance combined with the least significant difference t test was used for comparison between groups. Multiple time points were compared by repeated measurement analysis of variance. RESULTS DSS-induced UC mice had significantly decreased body weight, increased disease activity index, decreased colon length, and decreased miR-125a-5p expression (all P < 0.05). In the DSS-induced mouse model, the expression of miR-125a-5p rebounded and ferroptosis was inhibited after Rh2 treatment (all P < 0.05). Inhibition of miR-125a-5p or upregulation of SP1 expression counteracted the protective effects of Rh2 on UC mice and ferroptosis cell models (all P < 0.05). CONCLUSIONS Rh2 mitigated DSS-induced colitis in mice and restrained ferroptosis by targeting miR-125a-5p. Downregulating miR-125a-5p or elevating SP1 could counteract the protective impacts of Rh2 on ferroptotic cells. The findings convey that Rh2 has a latent application value in the treatment of UC.
Collapse
Affiliation(s)
- Xun Zhao
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No. 83, East Zhongshan Road, Guiyang, 550002, Guizhou, China
| | - WenQiang Yuan
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No. 83, East Zhongshan Road, Guiyang, 550002, Guizhou, China
| | - LiuChan Yang
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No. 83, East Zhongshan Road, Guiyang, 550002, Guizhou, China
| | - Fang Yan
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No. 83, East Zhongshan Road, Guiyang, 550002, Guizhou, China
| | - DeJun Cui
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, No. 83, East Zhongshan Road, Guiyang, 550002, Guizhou, China.
| |
Collapse
|
29
|
Demirturk M, Cinar MS, Avci FY. The immune interactions of gut glycans and microbiota in health and disease. Mol Microbiol 2024; 122:313-330. [PMID: 38703041 DOI: 10.1111/mmi.15267] [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: 10/13/2023] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 05/06/2024]
Abstract
The human digestive system harbors a vast diversity of commensal bacteria and maintains a symbiotic relationship with them. However, imbalances in the gut microbiota accompany various diseases, such as inflammatory bowel diseases (IBDs) and colorectal cancers (CRCs), which significantly impact the well-being of populations globally. Glycosylation of the mucus layer is a crucial factor that plays a critical role in maintaining the homeostatic environment in the gut. This review delves into how the gut microbiota, immune cells, and gut mucus layer work together to establish a balanced gut environment. Specifically, the role of glycosylation in regulating immune cell responses and mucus metabolism in this process is examined.
Collapse
Affiliation(s)
- Mahmut Demirturk
- Department of Biochemistry, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mukaddes Sena Cinar
- Department of Biochemistry, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Fikri Y Avci
- Department of Biochemistry, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
30
|
Mitchell C, Staley S, Williams MC, Saxena A, Bogdon R, Roark K, Hailey M, Miranda K, Becker W, Dopkins N, Pena MM, Hogan KM, Baird M, Wilson K, Nagarkatti P, Nagarkatti M, Busbee PB. Regulation of Bacteroides acidifaciens by the aryl hydrocarbon receptor in IL-22-producing immune cells has sex-dependent consequential impact on colitis. Front Immunol 2024; 15:1444045. [PMID: 39229279 PMCID: PMC11368719 DOI: 10.3389/fimmu.2024.1444045] [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: 06/05/2024] [Accepted: 07/31/2024] [Indexed: 09/05/2024] Open
Abstract
Introduction Colitis is an inflammatory bowel disease (IBD) characterized by immune cell dysregulation and alterations in the gut microbiome. In our previous report, we showed a natural product in cruciferous vegetables and ligand of the aryl hydrocarbon receptor (AhR), indole-3-carbinol (I3C), was able to reduce colitis-induced disease severity and microbial dysbiosis in an interleukin-22 (IL-22) dependent manner. Methods In the current study, we performed single-cell RNA sequencing (scRNAseq) from colonocytes during colitis induction and supplementation with I3C and show how this treatment alters expression of genes involved in IL-22 signaling. To further define the role of IL-22 signaling in I3C-mediated protection during colitis and disease-associated microbial dysbiosis, we generated mice with AhR deficiency in RAR-related orphan receptor c (Rorc)-expressing cells (AhR ΔRorc ) which depletes this receptor in immune cells involved in production of IL-22. Colitis was induced in wildtype (WT), AhR ΔRorc , and littermate (LM) mice with or without I3C treatment. Results Results showed AhR ΔRorc mice lost the efficacy effects of I3C treatment which correlated with a loss of ability to increase IL-22 by innate lymphoid type 3 (ILC3s), not T helper 22 (Th22) cells. 16S rRNA microbiome profiling studies showed AhR ΔRorc mice were unable to regulate disease-associated increases in Bacteroides, which differed between males and females. Lastly, inoculation with a specific disease-associated Bacteroides species, Bacteroides acidifaciens (B. acidifaciens), was shown to exacerbate colitis in females, but not males. Discussion Collectively, this report highlights the cell and sex-specific role of AhR in regulating microbes that can impact colitis disease.
Collapse
Affiliation(s)
- Chandani Mitchell
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Shanieka Staley
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Michal Claire Williams
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Archana Saxena
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Raymond Bogdon
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Kasie Roark
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Michele Hailey
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Kathryn Miranda
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - William Becker
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Nicholas Dopkins
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Maria Marjorette Pena
- Department of Biological Sciences, College of Arts and Sciences, University of South Carolina, Columbia, SC, United States
| | - Kristen M. Hogan
- Department of Biological Sciences, College of Arts and Sciences, University of South Carolina, Columbia, SC, United States
| | - Maredith Baird
- Department of Biological Sciences, College of Arts and Sciences, University of South Carolina, Columbia, SC, United States
| | - Kiesha Wilson
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Philip Brandon Busbee
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
31
|
Sikdar S, Mitra D, Das O, Bhaumik M, Dutta S. The functional antagonist of sphingosine-1-phosphate, FTY720, impairs gut barrier function. Front Pharmacol 2024; 15:1407228. [PMID: 39224783 PMCID: PMC11366638 DOI: 10.3389/fphar.2024.1407228] [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: 03/26/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
FTY720 or fingolimod is a known functional antagonist of sphingosine-1-phosphate (S1P), and it is effective in treating multiple sclerosis and preventing inflammatory bowel disease (IBD). Evidence shows that its use in mice can increase the susceptibility to mucosal infections. Despite the significant contribution of S1P to barrier function, the effect of the administration of FTY720 on the mucosal barrier has never been investigated. In this study, we looked into how FTY720 therapy affected the function of the gut barrier susceptibility. Administration of FTY720 to C57BL/6 mice enhances the claudin-2 expression and reduces the expression of claudin-4 and occludin, as studied by qPCR, Western blot, and immunofluorescence. FTY720 inhibits the Akt-mTOR pathway to decrease occludin and claudin-4 expression and increase claudin-2 expression. FTY720 treatment induced increased colonic inflammation, with notably greater immune cell infiltration, colon histopathology, and increased production of TNF-α, IFN-γ, CXCL-1, and CXCL-2 than that in control mice. Taking into account the close association of "the leaky gut" and gut dysbiosis among the major diseases, we therefore can infer that the vigilance of gut pathology should be maintained, where FTY720 is used as a treatment option.
Collapse
Affiliation(s)
- Sohini Sikdar
- Division of Immunology , ICMR-National Institute for Research in Bacterial Infections (NIRBI), Kolkata, India
| | - Debmalya Mitra
- Center of Radiological Research, Columbia University Irving Medical Center, New York, NY, United States
| | - Oishika Das
- Division of Immunology , ICMR-National Institute for Research in Bacterial Infections (NIRBI), Kolkata, India
| | - Moumita Bhaumik
- Division of Immunology , ICMR-National Institute for Research in Bacterial Infections (NIRBI), Kolkata, India
| | - Shanta Dutta
- Division of Immunology , ICMR-National Institute for Research in Bacterial Infections (NIRBI), Kolkata, India
| |
Collapse
|
32
|
Dilixiati Y, Aipire A, Song M, Nijat D, Wubuli A, Cao Q, Li J. The Potential Role of Plant Polysaccharides in Treatment of Ulcerative Colitis. Pharmaceutics 2024; 16:1073. [PMID: 39204418 PMCID: PMC11360206 DOI: 10.3390/pharmaceutics16081073] [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: 05/13/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Ulcerative colitis (UC) results in inflammation and ulceration of the colon and the rectum's inner lining. The application of herbal therapy in UC is increasing worldwide. As natural macromolecular compounds, polysaccharides have a significant role in the treatment of UC due to advantages of better biodegradation, good biocompatibility, immunomodulatory activity, and low reactogenicity. Therefore, polysaccharide drug formulation is becoming a potential candidate for UC treatment. In this review, we summarize the etiology and pathogenesis of UC and the therapeutic effects of polysaccharides on UC, such as regulating the expression of cytokines and tight junction proteins and modulating the balance of immune cells and intestinal microbiota. Polysaccharides can also serve as drug delivery carriers to enhance drug targeting and reduce side effects. This review provides a theoretical basis for applying natural plant polysaccharides in the prevention and treatment of UC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China; (Y.D.); (A.A.); (M.S.); (D.N.); (A.W.); (Q.C.)
| |
Collapse
|
33
|
Bernardi F, Ungaro F, D’Amico F, Zilli A, Parigi TL, Massimino L, Allocca M, Danese S, Furfaro F. The Role of Viruses in the Pathogenesis of Immune-Mediated Gastro-Intestinal Diseases. Int J Mol Sci 2024; 25:8301. [PMID: 39125870 PMCID: PMC11313478 DOI: 10.3390/ijms25158301] [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: 06/18/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Immune-mediated gastrointestinal (GI) diseases, including achalasia, celiac disease, and inflammatory bowel diseases, pose significant challenges in diagnosis and management due to their complex etiology and diverse clinical manifestations. While genetic predispositions and environmental factors have been extensively studied in the context of these conditions, the role of viral infections and virome dysbiosis remains a subject of growing interest. This review aims to elucidate the involvement of viral infections in the pathogenesis of immune-mediated GI diseases, focusing on achalasia and celiac disease, as well as the virome dysbiosis in IBD. Recent evidence suggests that viral pathogens, ranging from common respiratory viruses to enteroviruses and herpesviruses, may trigger or exacerbate achalasia and celiac disease by disrupting immune homeostasis in the GI tract. Furthermore, alterations in the microbiota and, specifically, in the virome composition and viral-host interactions have been implicated in perpetuating chronic intestinal inflammation in IBD. By synthesizing current knowledge on viral contributions to immune-mediated GI diseases, this review aims to provide insights into the complex interplay between viral infections, host genetics, and virome dysbiosis, shedding light on novel therapeutic strategies aimed at mitigating the burden of these debilitating conditions on patients' health and quality of life.
Collapse
Affiliation(s)
- Francesca Bernardi
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.B.); (F.U.); (F.D.); (A.Z.); (T.L.P.); (L.M.); (M.A.); (S.D.)
- Gastroenterology and Endoscopy, Vita-Salute San Raffaele University, Via Olgettina, 58, 20132 Milan, Italy
| | - Federica Ungaro
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.B.); (F.U.); (F.D.); (A.Z.); (T.L.P.); (L.M.); (M.A.); (S.D.)
| | - Ferdinando D’Amico
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.B.); (F.U.); (F.D.); (A.Z.); (T.L.P.); (L.M.); (M.A.); (S.D.)
| | - Alessandra Zilli
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.B.); (F.U.); (F.D.); (A.Z.); (T.L.P.); (L.M.); (M.A.); (S.D.)
| | - Tommaso Lorenzo Parigi
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.B.); (F.U.); (F.D.); (A.Z.); (T.L.P.); (L.M.); (M.A.); (S.D.)
| | - Luca Massimino
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.B.); (F.U.); (F.D.); (A.Z.); (T.L.P.); (L.M.); (M.A.); (S.D.)
| | - Mariangela Allocca
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.B.); (F.U.); (F.D.); (A.Z.); (T.L.P.); (L.M.); (M.A.); (S.D.)
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.B.); (F.U.); (F.D.); (A.Z.); (T.L.P.); (L.M.); (M.A.); (S.D.)
- Gastroenterology and Endoscopy, Vita-Salute San Raffaele University, Via Olgettina, 58, 20132 Milan, Italy
| | - Federica Furfaro
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (F.B.); (F.U.); (F.D.); (A.Z.); (T.L.P.); (L.M.); (M.A.); (S.D.)
| |
Collapse
|
34
|
Dzhalilova D, Silina M, Tsvetkov I, Kosyreva A, Zolotova N, Gantsova E, Kirillov V, Fokichev N, Makarova O. Changes in the Expression of Genes Regulating the Response to Hypoxia, Inflammation, Cell Cycle, Apoptosis, and Epithelial Barrier Functioning during Colitis-Associated Colorectal Cancer Depend on Individual Hypoxia Tolerance. Int J Mol Sci 2024; 25:7801. [PMID: 39063041 PMCID: PMC11276979 DOI: 10.3390/ijms25147801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
One of the factors contributing to colorectal cancer (CRC) development is inflammation, which is mostly hypoxia-associated. This study aimed to characterize the morphological and molecular biological features of colon tumors in mice that were tolerant and susceptible to hypoxia based on colitis-associated CRC (CAC). Hypoxia tolerance was assessed through a gasping time evaluation in a decompression chamber. One month later, the animals were experimentally modeled for colitis-associated CRC by intraperitoneal azoxymethane administration and three dextran sulfate sodium consumption cycles. The incidence of tumor development in the distal colon in the susceptible to hypoxia mice was two times higher and all tumors (100%) were represented by adenocarcinomas, while in the tolerant mice, only 14% were adenocarcinomas and 86% were glandular intraepithelial neoplasia. The tumor area assessed on serially stepped sections was statistically significantly higher in the susceptible animals. The number of macrophages, CD3-CD19+, CD3+CD4+, and NK cells in tumors did not differ between animals; however, the number of CD3+CD8+ and vimentin+ cells was higher in the susceptible mice. Changes in the expression of genes regulating the response to hypoxia, inflammation, cell cycle, apoptosis, and epithelial barrier functioning in tumors and the peritumoral area depended on the initial mouse's hypoxia tolerance, which should be taken into account for new CAC diagnostics and treatment approaches development.
Collapse
Affiliation(s)
- Dzhuliia Dzhalilova
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia; (M.S.); (I.T.); (A.K.); (N.Z.); (E.G.); (N.F.); (O.M.)
| | - Maria Silina
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia; (M.S.); (I.T.); (A.K.); (N.Z.); (E.G.); (N.F.); (O.M.)
| | - Ivan Tsvetkov
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia; (M.S.); (I.T.); (A.K.); (N.Z.); (E.G.); (N.F.); (O.M.)
| | - Anna Kosyreva
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia; (M.S.); (I.T.); (A.K.); (N.Z.); (E.G.); (N.F.); (O.M.)
- Research Institute of Molecular and Cellular Medicine, People’s Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Natalia Zolotova
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia; (M.S.); (I.T.); (A.K.); (N.Z.); (E.G.); (N.F.); (O.M.)
| | - Elena Gantsova
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia; (M.S.); (I.T.); (A.K.); (N.Z.); (E.G.); (N.F.); (O.M.)
- Research Institute of Molecular and Cellular Medicine, People’s Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Vladimir Kirillov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Health of Russian Federation, 117513 Moscow, Russia;
| | - Nikolay Fokichev
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia; (M.S.); (I.T.); (A.K.); (N.Z.); (E.G.); (N.F.); (O.M.)
| | - Olga Makarova
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia; (M.S.); (I.T.); (A.K.); (N.Z.); (E.G.); (N.F.); (O.M.)
| |
Collapse
|
35
|
Yang J, Xiao Y, Zhao N, Pei G, Sun Y, Sun X, Yu K, Miao C, Liu R, Lv J, Chu H, Zhou L, Wang B, Yao Z, Wang Q. PIM1-HDAC2 axis modulates intestinal homeostasis through epigenetic modification. Acta Pharm Sin B 2024; 14:3049-3067. [PMID: 39027246 PMCID: PMC11252454 DOI: 10.1016/j.apsb.2024.04.017] [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: 10/11/2023] [Revised: 03/07/2024] [Accepted: 03/14/2024] [Indexed: 07/20/2024] Open
Abstract
The mucosal barrier is crucial for intestinal homeostasis, and goblet cells are essential for maintaining the mucosal barrier integrity. The proviral integration site for Moloney murine leukemia virus-1 (PIM1) kinase regulates multiple cellular functions, but its role in intestinal homeostasis during colitis is unknown. Here, we demonstrate that PIM1 is prominently elevated in the colonic epithelia of both ulcerative colitis patients and murine models, in the presence of intestinal microbiota. Epithelial PIM1 leads to decreased goblet cells, thus impairing resistance to colitis and colitis-associated colorectal cancer (CAC) in mice. Mechanistically, PIM1 modulates goblet cell differentiation through the Wnt and Notch signaling pathways. Interestingly, PIM1 interacts with histone deacetylase 2 (HDAC2) and downregulates its level via phosphorylation, thereby altering the epigenetic profiles of Wnt signaling pathway genes. Collectively, these findings investigate the unknown function of the PIM1-HDAC2 axis in goblet cell differentiation and ulcerative colitis/CAC pathogenesis, which points to the potential for PIM1-targeted therapies of ulcerative colitis and CAC.
Collapse
Affiliation(s)
- Jianming Yang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Yawen Xiao
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Ningning Zhao
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Geng Pei
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center of Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center of Cancer, Tianjin 30060, China
| | - Yan Sun
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center of Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center of Cancer, Tianjin 30060, China
| | - Xinyu Sun
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Kaiyuan Yu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Chunhui Miao
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Ran Liu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Junqiang Lv
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Hongyu Chu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Lu Zhou
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin 300070, China
| | - Zhi Yao
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Quan Wang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Immunology, School of Basic Medical Sciences, Tianjin Institute of Urology, the Second Hospital of Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
36
|
Francis KL, Zheng HB, Suskind DL, Murphree TA, Phan BA, Quah E, Hendrickson AS, Zhou X, Nuding M, Hudson AS, Guttman M, Morton GJ, Schwartz MW, Alonge KM, Scarlett JM. Characterizing the human intestinal chondroitin sulfate glycosaminoglycan sulfation signature in inflammatory bowel disease. Sci Rep 2024; 14:11839. [PMID: 38782973 PMCID: PMC11116513 DOI: 10.1038/s41598-024-60959-x] [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: 12/21/2023] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
The intestinal extracellular matrix (ECM) helps maintain appropriate tissue barrier function and regulate host-microbial interactions. Chondroitin sulfate- and dermatan sulfate-glycosaminoglycans (CS/DS-GAGs) are integral components of the intestinal ECM, and alterations in CS/DS-GAGs have been shown to significantly influence biological functions. Although pathologic ECM remodeling is implicated in inflammatory bowel disease (IBD), it is unknown whether changes in the intestinal CS/DS-GAG composition are also linked to IBD in humans. Our aim was to characterize changes in the intestinal ECM CS/DS-GAG composition in intestinal biopsy samples from patients with IBD using mass spectrometry. We characterized intestinal CS/DS-GAGs in 69 pediatric and young adult patients (n = 13 control, n = 32 active IBD, n = 24 IBD in remission) and 6 adult patients. Here, we report that patients with active IBD exhibit a significant decrease in the relative abundance of CS/DS isomers associated with matrix stability (CS-A and DS) compared to controls, while isomers implicated in matrix instability and inflammation (CS-C and CS-E) were significantly increased. This imbalance of intestinal CS/DS isomers was restored among patients in clinical remission. Moreover, the abundance of pro-stabilizing CS/DS isomers negatively correlated with clinical disease activity scores, whereas both pro-inflammatory CS-C and CS-E content positively correlated with disease activity scores. Thus, pediatric patients with active IBD exhibited increased pro-inflammatory and decreased pro-stabilizing CS/DS isomer composition, and future studies are needed to determine whether changes in the CS/DS-GAG composition play a pathogenic role in IBD.
Collapse
Affiliation(s)
- Kendra L Francis
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, WA, USA
- Department of Medicine, University of Washington Medicine Diabetes Institute, 750 Republican St, Box 358062, Seattle, WA, 98195, USA
| | - Hengqi B Zheng
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, WA, USA
| | - David L Suskind
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, WA, USA
| | - Taylor A Murphree
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Bao Anh Phan
- Department of Medicine, University of Washington Medicine Diabetes Institute, 750 Republican St, Box 358062, Seattle, WA, 98195, USA
| | - Emily Quah
- Department of Medicine, University of Washington Medicine Diabetes Institute, 750 Republican St, Box 358062, Seattle, WA, 98195, USA
| | - Aarun S Hendrickson
- Department of Medicine, University of Washington Medicine Diabetes Institute, 750 Republican St, Box 358062, Seattle, WA, 98195, USA
| | - Xisheng Zhou
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Mason Nuding
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, WA, USA
| | - Alexandra S Hudson
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, WA, USA
| | - Miklos Guttman
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Gregory J Morton
- Department of Medicine, University of Washington Medicine Diabetes Institute, 750 Republican St, Box 358062, Seattle, WA, 98195, USA
| | - Michael W Schwartz
- Department of Medicine, University of Washington Medicine Diabetes Institute, 750 Republican St, Box 358062, Seattle, WA, 98195, USA
| | - Kimberly M Alonge
- Department of Medicine, University of Washington Medicine Diabetes Institute, 750 Republican St, Box 358062, Seattle, WA, 98195, USA
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Jarrad M Scarlett
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, WA, USA.
- Department of Medicine, University of Washington Medicine Diabetes Institute, 750 Republican St, Box 358062, Seattle, WA, 98195, USA.
| |
Collapse
|
37
|
Jurjus A, El Masri J, Ghazi M, El Ayoubi LM, Soueid L, Gerges Geagea A, Jurjus R. Mechanism of Action of Melatonin as a Potential Adjuvant Therapy in Inflammatory Bowel Disease and Colorectal Cancer. Nutrients 2024; 16:1236. [PMID: 38674926 PMCID: PMC11054672 DOI: 10.3390/nu16081236] [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: 03/22/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
Inflammatory bowel disease (IBD), a continuum of chronic inflammatory diseases, is tightly associated with immune system dysregulation and dysbiosis, leading to inflammation in the gastrointestinal tract (GIT) and multiple extraintestinal manifestations. The pathogenesis of IBD is not completely elucidated. However, it is associated with an increased risk of colorectal cancer (CRC), which is one of the most common gastrointestinal malignancies. In both IBD and CRC, a complex interplay occurs between the immune system and gut microbiota (GM), leading to the alteration in GM composition. Melatonin, a neuroendocrine hormone, was found to be involved with this interplay, especially since it is present in high amounts in the gut, leading to some protective effects. Actually, melatonin enhances the integrity of the intestinal mucosal barrier, regulates the immune response, alleviates inflammation, and attenuates oxidative stress. Thereby, the authors summarize the multifactorial interaction of melatonin with IBD and with CRC, focusing on new findings related to the mechanisms of action of this hormone, in addition to its documented positive outcomes on the treatment of these two pathologies and possible future perspectives to use melatonin as an adjuvant therapy.
Collapse
Affiliation(s)
- Abdo Jurjus
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon; (J.E.M.); (M.G.); (L.S.); (A.G.G.); (R.J.)
| | - Jad El Masri
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon; (J.E.M.); (M.G.); (L.S.); (A.G.G.); (R.J.)
- Faculty of Medical Sciences, Lebanese University, Beirut 6573, Lebanon;
| | - Maya Ghazi
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon; (J.E.M.); (M.G.); (L.S.); (A.G.G.); (R.J.)
- Faculty of Medical Sciences, Lebanese University, Beirut 6573, Lebanon;
| | | | - Lara Soueid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon; (J.E.M.); (M.G.); (L.S.); (A.G.G.); (R.J.)
| | - Alice Gerges Geagea
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon; (J.E.M.); (M.G.); (L.S.); (A.G.G.); (R.J.)
| | - Rosalyn Jurjus
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon; (J.E.M.); (M.G.); (L.S.); (A.G.G.); (R.J.)
| |
Collapse
|
38
|
Wang Z, Shen J. The role of goblet cells in Crohn' s disease. Cell Biosci 2024; 14:43. [PMID: 38561835 PMCID: PMC10985922 DOI: 10.1186/s13578-024-01220-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 03/14/2024] [Indexed: 04/04/2024] Open
Abstract
The prevalence of Crohn's disease (CD), a subtype of inflammatory bowel disease (IBD), is increasing worldwide. The pathogenesis of CD is hypothesized to be related to environmental, genetic, immunological, and bacterial factors. Current studies have indicated that intestinal epithelial cells, including columnar, Paneth, M, tuft, and goblet cells dysfunctions, are strongly associated with these pathogenic factors. In particular, goblet cells dysfunctions have been shown to be related to CD pathogenesis by direct or indirect ways, according to the emerging studies. The mucus barrier was established with the help of mucins secreted by goblet cells. Not only do the mucins mediate the mucus barrier permeability and bacterium selection, but also, they are closely linked with the endothelial reticulum stress during the synthesis process. Goblet cells also play a vital role in immune response. It was indicated that goblet cells take part in the antigen presentation and cytokines secretion process. Disrupted goblet cells related immune process were widely discovered in CD patients. Meanwhile, dysbiosis of commensal and pathogenic microbiota can induce myriad immune responses through mucus and goblet cell-associated antigen passage. Microbiome dysbiosis lead to inflammatory reaction against pathogenic bacteria and abnormal tolerogenic response. All these three pathways, including the loss of mucus barrier function, abnormal immune reaction, and microbiome dysbiosis, may have independent or cooperative effect on the CD pathogenesis. However, many of the specific mechanisms underlying these pathways remain unclear. Based on the current understandings of goblet cell's role in CD pathogenesis, substances including butyrate, PPARγagonist, Farnesoid X receptor agonist, nuclear factor-Kappa B, nitrate, cytokines mediators, dietary and nutrient therapies were all found to have potential therapeutic effects on CD by regulating the goblet cells mediated pathways. Several monoclonal antibodies already in use for the treatment of CD in the clinical settings were also found to have some goblet cells related therapeutic targets. In this review, we introduce the disease-related functions of goblet cells, their relationship with CD, their possible mechanisms, and current CD treatments targeting goblet cells.
Collapse
Affiliation(s)
- Zichen Wang
- Division of Gastroenterology and Hepatology, Baoshan Branch, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Inflammatory Bowel Disease Research Center, Renji Hospital, School of Medicine, Ministry of Health, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, No.160 PuJian Road, Shanghai, 200127, China
| | - Jun Shen
- Division of Gastroenterology and Hepatology, Baoshan Branch, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Inflammatory Bowel Disease Research Center, Renji Hospital, School of Medicine, Ministry of Health, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, No.160 PuJian Road, Shanghai, 200127, China.
| |
Collapse
|
39
|
Riemann B, Antoine T, Béduneau A, Pellequer Y, Lamprecht A, Moulari B. Active nanoparticle targeting of MUC5AC ameliorates therapeutic outcome in experimental colitis. NANOSCALE 2024; 16:5715-5728. [PMID: 38407269 DOI: 10.1039/d3nr05681c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Inflammatory bowel diseases (IBDs), which include Crohn's disease (CD) and ulcerative colitis (UC), are chronic inflammatory diseases of the gastrointestinal tract and are characterized by chronic recurrent ulceration of the bowels. Colon-targeted drug delivery systems (DDS) have received significant attention for their potential to treat IBD by improving the inflamed tissue selectivity. Herein, antiMUC5AC-decorated drug loaded nanoparticles (NP) are suggested for active epithelial targeting and selective adhesion to the inflamed tissue in experimental colitis. NPs conjugated with antiMUC5AC (anti-MUC5) were tested for their degree of bioadhesion with HT29-MTX cells by comparison with non-targeted BSA-NP conjugates. In vivo, the selectivity of bioadhesion and the influence of ligand density in bioadhesion efficiency as well as the therapeutic benefit for glucocorticoid loaded anti-MUC5-NP were studied in a murine colitis model. Quantitative adhesion analyses showed that anti-MUC5-conjugated NP exhibited a much higher binding and selectivity to inflamed tissue compared to PNA-, IgG1- and BSA-NP conjugates used as controls. This bioadhesion efficiency was found to be dependent on the ligand density, present at the NP surface. The binding specificity between anti-MUC5 ligand and inflamed tissues was confirmed by fluorescence imaging. Both anti-MUC5-NP and all other glucocorticoid containing formulations led to a significant mitigation of the experimental colitis, as became evident from the substantial reduction of myeloperoxidase activity and pro-inflammatory cytokine concentrations (TNF-α, IL-1β). Targeted NP by using anti-MUC5 appears to be a very promising tool in future treatment of various types of local disorders affecting the gastro-intestinal tract but not limited to colitis.
Collapse
Affiliation(s)
- Bernadette Riemann
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Germany
| | - Thomas Antoine
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France
| | - Arnaud Béduneau
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France
| | - Yann Pellequer
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France
| | - Alf Lamprecht
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Germany
| | - Brice Moulari
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, F-25000 Besançon, France
| |
Collapse
|
40
|
Liu X, Zhang B, Zhang Y, Li W, Yin J, Shi A, Wang J, Wang S. 2'-Fucosyllactose Promotes Colonization of Akkermansia muciniphila and Prevents Colitis In Vitro and in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:4765-4776. [PMID: 38393978 DOI: 10.1021/acs.jafc.3c08305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
Akkermansia muciniphila is a potential candidate for ulcerative colitis prevention. Considering that it utilizes 2'-fucosyllactose (2'FL) for growth, 2'FL can be used to enrich the abundance of A. muciniphila in feces. However, whether the crosswalk between 2'FL and A. muciniphila can promote the intestinal colonization of A. muciniphila remains unclear. In this study, we explored the effect and the underlying mechanism of 2'FL on the colonization of A. muciniphila in vitro and in vivo as well as its alleviating effect on colitis. Our results revealed that 2'FL can serve as a carbon source of A. muciniphila to support the growth and increase cell-surface hydrophobicity and the expression of the genes coding fibronectin-binding autotransporter adhesin to promote the adhesion to Caco2/HT29 methotrexate (MTX) cells but not of galactooligosaccharides (GOS) and glucose. Moreover, 2'FL could increase the host mucin formation to promote the adhesion of A. muciniphila to Caco2/HT29 MTX cells but not of GOS and glucose. Furthermore, 2'FL could significantly increase the colonization of A. muciniphila in the gut to alleviate colitis in mice. Overall, the interplay between A. muciniphila and 2'FL is expected to provide an advantageous ecological niche for A. muciniphila so as to confer further health benefits against colitis.
Collapse
Affiliation(s)
- Xiaoxia Liu
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Bowei Zhang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Yunhui Zhang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Wanhua Li
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Jia Yin
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Aiying Shi
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Jin Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
41
|
Zhan M, Yang X, Zhao C, Han Y, Xie P, Mo Z, Xiao J, Cao Y, Xiao H, Song M. Dietary nobiletin regulated cefuroxime- and levofloxacin-associated "gut microbiota-metabolism" imbalance and intestinal barrier dysfunction in mice. Food Funct 2024; 15:1265-1278. [PMID: 38196314 DOI: 10.1039/d3fo04378a] [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: 01/11/2024]
Abstract
Nobiletin (NOB) exhibits significant biological activities and may be a potential dietary treatment for antibiotic-associated gut dysbiosis. In this study, mice were gavaged with 0.2 mL day-1 of 12.5 g L-1 cefuroxime (LFX) and 10 g L-1 levofloxacin (LVX) for a duration of 10 days, accompanied by 0.05% NOB to investigate the regulatory effect and potential mechanisms of NOB on antibiotic-induced intestinal microbiota disorder and intestinal barrier dysfunction. Our results indicated that dietary NOB improved the pathology of intestinal epithelial cells and the intestinal permeability by upregulating the expression of intestinal tight junction proteins (TJs) and the number of goblet cells. Furthermore, dietary NOB reduced the levels of serum lipopolysaccharide (LPS) and pro-inflammatory factors (TNF-α and IL-1β), thereby facilitating the restoration of the intestinal mucosal barrier. Additionally, dietary NOB increased the abundance of beneficial bacteria f_Lachnospiraceae and regulated the metabolic disorders of short-chain fatty acids (SCFAs) and bile acids (BAs). Notably, NOB supplementation resulted in elevated levels of butyric acid and lithocholic acid (LCA), which contributed to the repair of the intestinal mucosal barrier function and the maintenance of intestinal homeostasis. Collectively, our results propose a healthy dietary strategy for the prevention or mitigation of antibiotic-associated gut dysbiosis by dietary NOB.
Collapse
Affiliation(s)
- Minmin Zhan
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, China.
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| | - Xiaoshuang Yang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, China.
| | - Chenxi Zhao
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| | - Yanhui Han
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shanxi 710062, P.R. China
| | - Peichun Xie
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, China.
| | - Zheqi Mo
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, China.
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, China.
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, China.
| | - Hang Xiao
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | - Mingyue Song
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Sciences, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
42
|
Leoncini G, Cari L, Ronchetti S, Donato F, Caruso L, Calafà C, Villanacci V. Mucin Expression Profiles in Ulcerative Colitis: New Insights on the Histological Mucosal Healing. Int J Mol Sci 2024; 25:1858. [PMID: 38339134 PMCID: PMC10855303 DOI: 10.3390/ijms25031858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
A structural weakness of the mucus barrier (MB) is thought to be a cause of ulcerative colitis (UC). This study aims to investigate the mucin (MUC) composition of MB in normal mucosa and UC. Ileocolonic biopsies were taken at disease onset and after treatment in 40 patients, including 20 with relapsing and 20 with remitting UC. Ileocolonic biopsies from 10 non-IBD patients were included as controls. Gut-specific MUC1, MUC2, MUC4, MUC5B, MUC12, MUC13, MUC15, and MUC17 were evaluated immunohistochemically. The promoters of mucin genes were also examined. Normal mucosa showed MUC2, MUC5B, and MUC13 in terminal ileum and colon, MUC17 in ileum, and MUC1, MUC4, MUC12, and MUC15 in colon. Membranous, cytoplasmic and vacuolar expressions were highlighted. Overall, the mucin expression was abnormal in UC. Derangements in MUC1, MUC4, and MUC5B were detected both at onset and after treatment. MUC2 and MUC13 were unaffected. Sequence analysis revealed glucocorticoid-responsive elements in the MUC1 promoter, retinoic-acid-responsive elements in the MUC4 promoter, and butyrate-responsive elements in the MUC5B promoter. In conclusion, MUCs exhibited distinct expression patterns in the gut. Their expression was disrupted in UC, regardless of the treatment protocols. Abnormal MUC1, MUC4, and MUC5B expression marked the barrier dysfunction in UC.
Collapse
Affiliation(s)
- Giuseppe Leoncini
- First Pathology Division, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Luigi Cari
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Simona Ronchetti
- Pharmacology Division, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Francesco Donato
- Unit of Hygiene, Epidemiology and Public Health, University of Brescia, 25123 Brescia, Italy
| | - Laura Caruso
- Pathology Unit, Department of Pathology and Laboratory Medicine, Desenzano del Garda Hospital, ASST del Garda, 25015 Brescia, Italy
| | - Cristina Calafà
- Pathology Unit, Department of Pathology and Laboratory Medicine, Desenzano del Garda Hospital, ASST del Garda, 25015 Brescia, Italy
| | | |
Collapse
|
43
|
Xiu W, Ding W, Mou S, Li Y, Sultan Y, Ma J, Li X. Adverse effects of fenpropathrin on the intestine of common carp (Cyprinus carpio L.) and the mechanism involved. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 199:105799. [PMID: 38458669 DOI: 10.1016/j.pestbp.2024.105799] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/10/2024] [Accepted: 01/19/2024] [Indexed: 03/10/2024]
Abstract
Fenpropathrin (FEN), a pyrethroid pesticide, is frequently detected in natural water bodies, unavoidable pose adverse effects to aquatic organisms. However, the harmful effects and potential mechanisms of FEN on aquatic species are poorly understood. In this study, common carp were treatment with FEN at 0.45 and 1.35 μg/L for 14 d, and the toxic effects and underlying mechanisms of FEN on the intestine of carp were revealed. RNA-seq results showed that FEN exposure cause a wide range of transcriptional alterations in the intestine and the differentially expressed genes were mainly enrichment in the pathways related to immune and metabolism. Specifically, FEN exposure induced pathological damage and altered submicroscopic structure of the intestine, elevated the levels of Bacteroides fragilis enterotoxin, altered the contents of claudin-1, occludin, and zonula occluden-1 (ZO-1), and causing injury to the intestinal barrier. In addition, inflammation-related index TNF-α in the serum and IL-6 in the intestinal tissues were generally increased after FEN exposure. Moreover, FEN exposure promoted an increase in reactive oxygen species (ROS), altered the levels of superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH), upregulated the contents of malondialdehyde (MDA) in the intestines. The apoptosis-related parameter cytochrome c, caspase-9, and caspase-3 were significantly altered, indicating that inflammation reaction, oxidative stress, and apoptosis may be involved in the toxic mechanism of FEN on carp. Moreover, FEN treatment also altered the intestinal flora community significantly, which may affect the intestinal normal physiological function and thus affect the growth of fish. Overall, the present study help to clarify the intestinal reaction mechanisms after FEN treatment, and provide a basis for the risk assessment of FEN.
Collapse
Affiliation(s)
- Wenyao Xiu
- College of Life Science, Henan Normal University, Xinxiang, Henan 453007, China
| | - Weikai Ding
- Henan International Joint Laboratory of Aquatic Toxicology and Health Protection, Henan Normal University, Xinxiang, Henan 453007, China
| | - Shaoyu Mou
- College of Life Science, Henan Normal University, Xinxiang, Henan 453007, China
| | - Yuanyuan Li
- Henan International Joint Laboratory of Aquatic Toxicology and Health Protection, Henan Normal University, Xinxiang, Henan 453007, China.
| | - Yousef Sultan
- Department of Food Toxicology and Contaminants, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Junguo Ma
- College of Life Science, Henan Normal University, Xinxiang, Henan 453007, China.
| | - Xiaoyu Li
- Henan International Joint Laboratory of Aquatic Toxicology and Health Protection, Henan Normal University, Xinxiang, Henan 453007, China
| |
Collapse
|
44
|
Dorohavtseva HA, Dorofieiev AE, Dyadyk OO, Myroshnychenko MS. Morphological and functional features of the colonic mucus barrier in patients with symptomatic uncomplicated diverticular disease and acute uncomplicated diverticulitis. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2024; 77:1331-1337. [PMID: 39241130 DOI: 10.36740/wlek202407105] [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: 09/08/2024]
Abstract
OBJECTIVE Aim: The purpose was to identify the morphological and functional features of the colonic mucus barrier in patients with symptomatic uncomplicated diverticular disease and acute uncomplicated diverticulitis. PATIENTS AND METHODS Materials and Methods: In the research, three groups were formed. Group 1 included fragments of the mucous membrane of the large intestine, which were collected from 12 people during autopsies. The results of autopsies and histological examination of the material did not reveal any gastrointestinal pathology. Group 2 included biopsies of the mucous membrane of the large intestine from the area of the diverticulum of 34 patients with symptomatic uncomplicated diverticular disease. Group 3 included biopsies of the mucous membrane of the large intestine of 26 patients with acute uncomplicated diverticulitis. Histological (hematoxylin and eosin staining), histochemical (PAS reaction) and immunohistochemical (mouse monoclonal antibodies to Mucin 2 (MUC2) and Mucin 4 (MUC4)) staining methods were used. A morphometric study was also carried out. RESULTS Results: In patients with diverticular disease, the authors identified disturbances in the morphofunctional state of the mucus barrier of the colon, the structure and function of goblet cells contained in its mucous membrane, characterized by a decrease in the thickness of the mucus layer covering the surface of the mucous membrane; a decrease in the size and number of goblet cells with a decrease in their mucus-producing ability; a change in the mucin profile, characterized by a violation of the content of MUC2 and MUC4. These changes were greatest in patients with acute uncomplicated diverticulitis compared with patients with symptomatic uncomplicated diverticular disease. CONCLUSION Conclusions: The identified disturbances in the morphofunctional state of the mucus barrier of the colon, structural and functional changes in goblet cells may be one of the mechanisms for the development of acute uncomplicated diverticulitis and symptomatic uncomplicated diverticular disease.
Collapse
Affiliation(s)
- Hanna A Dorohavtseva
- FEOFANIYA CLINICAL HOSPITAL OF THE STATE ADMINISTRATION OF AFFAIRS, KYIV, UKRAINE
| | | | - Olena O Dyadyk
- SHUPYK NATIONAL HEALTHCARE UNIVERSITY OF UKRAINE, KYIV, UKRAINE
| | | |
Collapse
|
45
|
Yang Y, Zhang Y, Song J, Li Y, Zhou L, Xu H, Wu K, Gao J, Zhao M, Zheng Y. Bergamot polysaccharides relieve DSS-induced ulcerative colitis via regulating the gut microbiota and metabolites. Int J Biol Macromol 2023; 253:127335. [PMID: 37820919 DOI: 10.1016/j.ijbiomac.2023.127335] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/16/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
This study aimed to explore the efficacy of polysaccharides from bergamot (BP) in alleviating DSS-induced colitis. Results showed that BP was primarily composed of two components, BP-1 and BP-2, with similar monosaccharide compositions to BP (mainly glucose and xylose) and molecular weights (Mw) of 4.50 × 105 and 2.35 × 105 Da. This study found BP relieved disease symptoms such as weight loss and colon shortening in mice with colitis. Gut microbiota and metabolomics analysis revealed that the BP could also promote the proliferation of beneficial bacteria such as Bifidobacteria, Butyrivibrio, and Blautia, resulting in increased levels of SCFAs and L-phenylalanine, which were associated with phenylalanine, tyrosine, and tryptophan metabolism pathways. Further analysis validated the inflammatory activity of L-phenylalanine. Hence, BP may relieve colitis symptoms by regulating the gut microbiota and metabolism, which reduced inflammation and enhanced the expression of tight junctional proteins (TJ proteins) and mucin in the intestine.
Collapse
Affiliation(s)
- Yiren Yang
- Chaozhou Branch of Chemistry and Chemical Engineering Guangdong Laboratory, Chaozhou 521031, China; School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Yuwei Zhang
- Chaozhou Branch of Chemistry and Chemical Engineering Guangdong Laboratory, Chaozhou 521031, China; School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Jiangping Song
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Yaqian Li
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Liuyang Zhou
- Chaozhou Branch of Chemistry and Chemical Engineering Guangdong Laboratory, Chaozhou 521031, China; School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Hongtao Xu
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Kaizhang Wu
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Jie Gao
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Mouming Zhao
- Chaozhou Branch of Chemistry and Chemical Engineering Guangdong Laboratory, Chaozhou 521031, China; School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China.
| | - Yang Zheng
- Chaozhou Branch of Chemistry and Chemical Engineering Guangdong Laboratory, Chaozhou 521031, China; School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China.
| |
Collapse
|
46
|
Jiménez-Meléndez A, Shakya R, Markussen T, Robertson LJ, Myrmel M, Makvandi-Nejad S. Gene expression profile of HCT-8 cells following single or co-infections with Cryptosporidium parvum and bovine coronavirus. Sci Rep 2023; 13:22106. [PMID: 38092824 PMCID: PMC10719361 DOI: 10.1038/s41598-023-49488-1] [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: 03/09/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023] Open
Abstract
Among the causative agents of neonatal diarrhoea in calves, two of the most prevalent are bovine coronavirus (BCoV) and the intracellular parasite Cryptosporidium parvum. Although several studies indicate that co-infections are associated with greater symptom severity, the host-pathogen interplay remains unresolved. Here, our main objective was to investigate the modulation of the transcriptome of HCT-8 cells during single and co-infections with BCoV and C. parvum. For this, HCT-8 cells were inoculated with (1) BCoV alone, (2) C. parvum alone, (3) BCoV and C. parvum simultaneously. After 24 and 72 h, cells were harvested and analyzed using high-throughput RNA sequencing. Following differential expression analysis, over 6000 differentially expressed genes (DEGs) were identified in virus-infected and co-exposed cells at 72 hpi, whereas only 52 DEGs were found in C. parvum-infected cells at the same time point. Pathway (KEGG) and gene ontology (GO) analysis showed that DEGs in the virus-infected and co-exposed cells were mostly associated with immune pathways (such as NF-κB, TNF-α or, IL-17), apoptosis and regulation of transcription, with a more limited effect exerted by C. parvum. Although the modulation observed in the co-infection was apparently dominated by the virus, over 800 DEGs were uniquely expressed in co-exposed cells at 72 hpi. Our findings provide insights on possible biomarkers associated with co-infection, which could be further explored using in vivo models.
Collapse
Affiliation(s)
- Alejandro Jiménez-Meléndez
- Department of Paraclinical Sciences (PARAFAG), Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Ås, Norway.
| | - Ruchika Shakya
- Department of Paraclinical Sciences (PARAFAG), Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Turhan Markussen
- Department of Paraclinical Sciences (PARAFAG), Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Lucy J Robertson
- Department of Paraclinical Sciences (PARAFAG), Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Mette Myrmel
- Department of Paraclinical Sciences (PARAFAG), Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Ås, Norway
| | - Shokouh Makvandi-Nejad
- Research Group Animal Health, Vaccinology, Norwegian Veterinary Institute, Ås, Norway
- Nykode Therapeutics ASA, Oslo Science Park, Oslo, Norway
| |
Collapse
|
47
|
Filippi L, Nardini P, Zizi V, Molino M, Fazi C, Calvani M, Carrozzo F, Cavallaro G, Giuseppetti G, Calosi L, Crociani O, Pini A. β3 Adrenoceptor Agonism Prevents Hyperoxia-Induced Colonic Alterations. Biomolecules 2023; 13:1755. [PMID: 38136626 PMCID: PMC10741994 DOI: 10.3390/biom13121755] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/30/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Oxygen level is a key regulator of organogenesis and its modification in postnatal life alters the maturation process of organs, including the intestine, which do not completely develop in utero. The β3-adrenoreceptor (β3-AR) is expressed in the colon and has an oxygen-dependent regulatory mechanism. This study shows the effects of the β3-AR agonist BRL37344 in a neonatal model of hyperoxia-driven colonic injury. For the first 14 days after birth, Sprague-Dawley rat pups were exposed to ambient oxygen levels (21%) or hyperoxia (85%) and treated daily with BRL37344 at 1, 3, 6 mg/kg or untreated. At the end of day 14, proximal colon samples were collected for analysis. Hyperoxia deeply influences the proximal colon development by reducing β3-AR-expressing cells (27%), colonic length (26%) and mucin production (47%), and altering the neuronal chemical coding in the myenteric plexus without changes in the neuron number. The administration of BRL37344 at 3 mg/kg, but not at 1 mg/kg, significantly prevented these alterations. Conversely, it was ineffective in preventing hyperoxia-induced body weight loss. BRL37344 at 6 mg/kg was toxic. These findings pave the way for β3-AR pharmacological targeting as a therapeutic option for diseases caused by hyperoxia-impaired development, typical prematurity disorders.
Collapse
Affiliation(s)
- Luca Filippi
- Department of Clinical and Experimental Medicine, University of Pisa, 56124 Pisa, Italy;
| | - Patrizia Nardini
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (V.Z.); (M.M.); (L.C.); (O.C.)
- Imaging Platform, Department Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy
| | - Virginia Zizi
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (V.Z.); (M.M.); (L.C.); (O.C.)
| | - Marta Molino
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (V.Z.); (M.M.); (L.C.); (O.C.)
| | - Camilla Fazi
- Department of Pediatric, Meyer Children’s University Hospital, 50139 Florence, Italy;
| | - Maura Calvani
- Division of Pediatric Oncology/Hematology, Meyer University Children’s Hospital, 50139 Florence, Italy; (M.C.); (F.C.)
| | - Francesco Carrozzo
- Division of Pediatric Oncology/Hematology, Meyer University Children’s Hospital, 50139 Florence, Italy; (M.C.); (F.C.)
| | - Giacomo Cavallaro
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Giorgia Giuseppetti
- Department of Clinical and Experimental Medicine, University of Pisa, 56124 Pisa, Italy;
| | - Laura Calosi
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (V.Z.); (M.M.); (L.C.); (O.C.)
| | - Olivia Crociani
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (V.Z.); (M.M.); (L.C.); (O.C.)
| | - Alessandro Pini
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (P.N.); (V.Z.); (M.M.); (L.C.); (O.C.)
- Imaging Platform, Department Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy
| |
Collapse
|
48
|
Liu W, Fan X, Jian B, Wen D, Wang H, Liu Z, Li B. The signaling pathway of hypoxia inducible factor in regulating gut homeostasis. Front Microbiol 2023; 14:1289102. [PMID: 37965556 PMCID: PMC10641782 DOI: 10.3389/fmicb.2023.1289102] [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: 09/05/2023] [Accepted: 10/02/2023] [Indexed: 11/16/2023] Open
Abstract
Hypoxia represent a condition in which an adequate amount of oxygen supply is missing in the body, and it could be caused by a variety of diseases, including gastrointestinal disorders. This review is focused on the role of hypoxia in the maintenance of the gut homeostasis and related treatment of gastrointestinal disorders. The effects of hypoxia on the gut microbiome and its role on the intestinal barrier functionality are also covered, together with the potential role of hypoxia in the development of gastrointestinal disorders, including inflammatory bowel disease and irritable bowel syndrome. Finally, we discussed the potential of hypoxia-targeted interventions as a novel therapeutic approach for gastrointestinal disorders. In this review, we highlighted the importance of hypoxia in the maintenance of the gut homeostasis and the potential implications for the treatment of gastrointestinal disorders.
Collapse
Affiliation(s)
- Wei Liu
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, China
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Xueni Fan
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, China
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Boshuo Jian
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Dongxu Wen
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, China
| | - Hongzhuang Wang
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, China
| | - Zhenjiang Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Bin Li
- Institute of Animal Husbandry and Veterinary, Tibet Academy of Agricultural and Animal Husbandry Sciences, Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lhasa, China
| |
Collapse
|
49
|
Ephraim R, Fraser S, Devereaux J, Stavely R, Feehan J, Eri R, Nurgali K, Apostolopoulos V. Differential Gene Expression of Checkpoint Markers and Cancer Markers in Mouse Models of Spontaneous Chronic Colitis. Cancers (Basel) 2023; 15:4793. [PMID: 37835487 PMCID: PMC10571700 DOI: 10.3390/cancers15194793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/17/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
The presence of checkpoint markers in cancer cells aids in immune escape. The identification of checkpoint markers and early cancer markers is of utmost importance to gain clarity regarding the relationship between colitis and progressive inflammation leading to cancer. Herein, the gene expression levels of checkpoint makers, cancer-related pathways, and cancer genes in colon tissues of mouse models of chronic colitis (Winnie and Winnie-Prolapse mice) using next-generation sequencing are determined. Winnie mice are a result of a Muc2 missense mutation. The identification of such genes and their subsequent expression and role at the protein level would enable novel markers for the early diagnosis of cancer in IBD patients. The differentially expressed genes in the colonic transcriptome were analysed based on the Kyoto Encyclopedia of Genes and Genomes pathway. The expression of several oncogenes is associated with the severity of IBD, with Winnie-Prolapse mice expressing a large number of key genes associated with development of cancer. This research presents a number of new targets to evaluate for the development of biomarkers and therapeutics.
Collapse
Affiliation(s)
- Ramya Ephraim
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (R.E.); (S.F.); (J.D.); (J.F.); (K.N.)
| | - Sarah Fraser
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (R.E.); (S.F.); (J.D.); (J.F.); (K.N.)
| | - Jeannie Devereaux
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (R.E.); (S.F.); (J.D.); (J.F.); (K.N.)
| | - Rhian Stavely
- Pediatric Surgery Research Laboratories, Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA;
| | - Jack Feehan
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (R.E.); (S.F.); (J.D.); (J.F.); (K.N.)
- Immunology Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
| | - Rajaraman Eri
- STEM/School of Science, RMIT University, Melbourne, VIC 3001, Australia;
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (R.E.); (S.F.); (J.D.); (J.F.); (K.N.)
- Department of Medicine Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC 3010, Australia
- Regenerative Medicine and Stem Cells Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (R.E.); (S.F.); (J.D.); (J.F.); (K.N.)
- Immunology Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
| |
Collapse
|
50
|
Francis KL, Alonge KM, Pacheco MC, Hu SJ, Krutzsch CA, Morton GJ, Schwartz MW, Scarlett JM. Diabetes exacerbates inflammatory bowel disease in mice with diet-induced obesity. World J Gastroenterol 2023; 29:4991-5004. [PMID: 37731997 PMCID: PMC10507503 DOI: 10.3748/wjg.v29.i33.4991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/22/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND The increased prevalence of inflammatory bowel disease (IBD) among patients with obesity and type 2 diabetes suggests a causal link between these diseases, potentially involving the effect of hyperglycemia to disrupt intestinal barrier integrity. AIM To investigate whether the deleterious impact of diabetes on the intestinal barrier is associated with increased IBD severity in a murine model of colitis in mice with and without diet-induced obesity. METHODS Mice were fed chow or a high-fat diet and subsequently received streptozotocin to induce diabetic-range hyperglycemia. Six weeks later, dextran sodium sulfate was given to induce colitis. In select experiments, a subset of diabetic mice was treated with the antidiabetic drug dapagliflozin prior to colitis onset. Endpoints included both clinical and histological measures of colitis activity as well as histochemical markers of colonic epithelial barrier integrity. RESULTS In mice given a high-fat diet, but not chow-fed animals, diabetes was associated with significantly increased clinical colitis activity and histopathologic markers of disease severity. Diabetes was also associated with a decrease in key components that regulate colonic epithelial barrier integrity (colonic mucin layer content and epithelial tight junction proteins) in diet-induced obese mice. Each of these effects of diabetes in diet-induced obese mice was ameliorated by restoring normoglycemia. CONCLUSION In obese mice, diabetes worsened clinical and pathologic outcomes of colitis via mechanisms that are reversible with treatment of hyperglycemia. Hyperglycemia-induced intestinal barrier dysfunction offers a plausible mechanism linking diabetes to increased colitis severity. These findings suggest that effective diabetes management may decrease the clinical severity of IBD.
Collapse
Affiliation(s)
- Kendra L Francis
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children’s Hospital, Seattle, WA 98105, United States
- Diabetes Institute, University of Washington, Seattle, WA 98109, United States
| | - Kimberly M Alonge
- Diabetes Institute, University of Washington, Seattle, WA 98109, United States
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, United States
| | - Maria Cristina Pacheco
- Department of Laboratory Medicine and Pathology, Seattle Children's Hospital, Seattle, WA 98105, United States
| | - Shannon J Hu
- Diabetes Institute, University of Washington, Seattle, WA 98109, United States
| | - Cody A Krutzsch
- Diabetes Institute, University of Washington, Seattle, WA 98109, United States
| | - Gregory J Morton
- Diabetes Institute, University of Washington, Seattle, WA 98109, United States
| | - Michael W Schwartz
- Diabetes Institute, University of Washington, Seattle, WA 98109, United States
| | - Jarrad M Scarlett
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children’s Hospital, Seattle, WA 98105, United States
- Diabetes Institute, University of Washington, Seattle, WA 98109, United States
| |
Collapse
|