1
|
Wang Y, Li C, Li J, Zhang S, Zhang Q, Duan J, Guo J. Abelmoschus manihot polysaccharide fortifies intestinal mucus barrier to alleviate intestinal inflammation by modulating Akkermansia muciniphila abundance. Acta Pharm Sin B 2024; 14:3901-3915. [PMID: 39309495 PMCID: PMC11413673 DOI: 10.1016/j.apsb.2024.06.002] [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: 03/26/2024] [Revised: 05/09/2024] [Accepted: 05/30/2024] [Indexed: 09/25/2024] Open
Abstract
The intestinal mucus barrier is an important line of defense against gut pathogens. Damage to this barrier brings bacteria into close contact with the epithelium, leading to intestinal inflammation. Therefore, its restoration is a promising strategy for alleviating intestinal inflammation. This study showed that Abelmoschus manihot polysaccharide (AMP) fortifies the intestinal mucus barrier by increasing mucus production, which plays a crucial role in the AMP-mediated amelioration of colitis. IL-10-deficient mouse models demonstrated that the effect of AMP on mucus production is dependent on IL-10. Moreover, bacterial depletion and replenishment confirmed that the effects of AMP on IL-10 secretion and mucus production were mediated by Akkermansia muciniphila. These findings suggest that plant polysaccharides fortify the intestinal mucus barrier by maintaining homeostasis in the gut microbiota. This demonstrates that targeting mucus barrier is a promising strategy for treating intestinal inflammation.
Collapse
Affiliation(s)
- Yumeng Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chengxi Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jianping Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shu Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qinyu Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jinao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jianming Guo
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
2
|
Seo KJ, Alam MR, Abdul-Ghafar J, Kim SW, Kim HK, Choi HH, Sin SH, Lee HK, Chae HS. Tranilast Treatment Prevents Chronic Radiation-Induced Colitis in Rats by Inhibiting Mast Cell Infiltration. Pharmacology 2024:1-10. [PMID: 39163845 DOI: 10.1159/000541003] [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: 12/06/2023] [Accepted: 08/12/2024] [Indexed: 08/22/2024]
Abstract
INTRODUCTION Mast cells are the principal cells involved in acute and chronic colitis due to radiation, known as radiation-induced colitis (RIC). In this study, we investigated whether pretreatment with tranilast, a mast cell inhibitor, could alleviate chronic RIC. METHODS A total of 23 Sprague-Dawley rats were randomly divided into three groups: control group (n = 5), radiation group (RG, n = 9), and tranilast-pretreated radiation group (TG, n = 9). The rats in the RG and the TG were irradiated in the pelvic area (1.5 cm from the anus) with a single dose of 20 Gy under general anesthesia. Tranilast (100 mg/kg) was administered intraperitoneally to the rats of the TG for 10 days, starting from the day of pelvic radiation. Ten weeks after radiation, the rats were euthanized. Rectal tissue samples were histologically evaluated for the total inflammation score (TIS) and mast cell count. The expression of MUC2, MUC5AC, and matrix metalloproteinase-9 (MMP-9) was also assessed immunohistochemically. RESULTS Both the TIS and specific components of TIS such as epithelial atypia, vascular sclerosis, and colitis cystica profunda (CCP) were significantly higher in the RG than in the TG (p = 0.02, 0.038, 0.025, and 0.01, respectively). Thein number of infiltrating mast cells was significantly higher in the RG than in the TG (median [range]: 20 [3-54] versus 6 [3-25], respectively; p = 0.034). Quantitatively, the number of MMP-9-positive cells was significantly higher in the RG (23.67 ± 19.00) than in the TG (10.25 ± 8.45) (mean ± standard deviation; p < 0.05). TIS and MMP-9 exhibited a strong association (correlation coefficient r = 0.56, p < 0.05). Immunohistochemically, the mucin-lake of CCP showed no staining for MUC5AC but was stained positive for MUC2. CONCLUSION Tranilast pretreatment of chronic RIC showed an anti-inflammatory effect associated with the reduction of mast cell infiltration and MMP-9 expression.
Collapse
Affiliation(s)
- Kyung Jin Seo
- Department of Hospital Pathology, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea,
| | - Mohammad Rizwan Alam
- Department of Hospital Pathology, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jamshid Abdul-Ghafar
- Department of Hospital Pathology, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sang Woo Kim
- Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyung Keun Kim
- Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyun Ho Choi
- Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Ho Sin
- Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hae Kyung Lee
- Department of Laboratory Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hiun Suk Chae
- Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
3
|
Bai J, Wang Y, Li F, Wu Y, Chen J, Li M, Wang X, Lv B. Research advancements and perspectives of inflammatory bowel disease: A comprehensive review. Sci Prog 2024; 107:368504241253709. [PMID: 38778725 PMCID: PMC11113063 DOI: 10.1177/00368504241253709] [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] [Indexed: 05/25/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease with increasing incidence, such as Crohn's disease and ulcerative colitis. The accurate etiology and pathogenesis of IBD remain unclear, and it is generally believed that it is related to genetic susceptibility, gut microbiota, environmental factors, immunological abnormalities, and potentially other factors. Currently, the mainstream therapeutic drugs are amino salicylic acid agents, corticosteroids, immunomodulators, and biological agents, but the remission rates do not surpass 30-60% of patients in a real-life setting. As a consequence, there are many studies focusing on emerging drugs and bioactive ingredients that have higher efficacy and long-term safety for achieving complete deep healing. This article begins with a review of the latest, systematic, and credible summaries of the pathogenesis of IBD. In addition, we provide a summary of the current treatments and drugs for IBD. Finally, we focus on the therapeutic effects of emerging drugs such as microRNAs and lncRNAs, nanoparticles-mediated drugs and natural products on IBD and their mechanisms of action.
Collapse
Affiliation(s)
- Junyi Bai
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Ying Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Fuhao Li
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yueyao Wu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jun Chen
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Meng Li
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xi Wang
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bin Lv
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
4
|
Kosmerl E, Miller C, Jiménez-Flores R. Preventative Effects of Milk Fat Globule Membrane Ingredients on DSS-Induced Mucosal Injury in Intestinal Epithelial Cells. Nutrients 2024; 16:954. [PMID: 38612988 PMCID: PMC11013169 DOI: 10.3390/nu16070954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/22/2024] [Accepted: 03/24/2024] [Indexed: 04/14/2024] Open
Abstract
The goblet cells of the gastrointestinal tract (GIT) produce glycoproteins called mucins that form a protective barrier from digestive contents and external stimuli. Recent evidence suggests that the milk fat globule membrane (MFGM) and its milk phospholipid component (MPL) can benefit the GIT through improving barrier function. Our objective was to compare the effects of two digested MFGM ingredients with or without dextran sodium sulfate (DSS)-induced barrier stress on mucin proteins. Co-cultured Caco-2/HT29-MTX intestinal cells were treated with in vitro digests of 2%, 5%, and 10% (w/v) MFGM or MPL alone for 6 h or followed by challenge with 2.5% DSS (6 h). Transepithelial electrical resistance and fluorescein isothiocyanate (FITC)-dextran (FD4) permeability measurements were used to measure changes in barrier integrity. Mucin characterization was performed using a combination of slot blotting techniques for secreted (MUC5AC, MUC2) and transmembrane (MUC3A, MUC1) mucins, scanning electron microscopy (SEM), and periodic acid Schiff (PAS)/Alcian blue staining. Digested MFGM and MPL prevented a DSS-induced reduction in secreted mucins, which corresponded to the prevention of DSS-induced increases in FD4 permeability. SEM and PAS/Alcian blue staining showed similar visual trends for secreted mucin production. A predictive bioinformatic approach was also used to identify potential KEGG pathways involved in MFGM-mediated mucosal maintenance under colitis conditions. This preliminary in silico evidence, combined with our in vitro findings, suggests the role of MFGM in inducing repair and maintenance of the mucosal barrier.
Collapse
Affiliation(s)
| | | | - Rafael Jiménez-Flores
- Department of Food Science and Technology, The Ohio State University, Columbus, OH 43210, USA; (E.K.); (C.M.)
| |
Collapse
|
5
|
Huang JJ, Feng YM, Zheng SM, Yu CL, Zhou RG, Liu MJ, Bo RN, Yu J, Li JG. Eugenol Possesses Colitis Protective Effects: Impacts on the TLR4/MyD88/NF-[Formula: see text]B Pathway, Intestinal Epithelial Barrier, and Macrophage Polarization. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:493-512. [PMID: 38480500 DOI: 10.1142/s0192415x24500216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2024]
Abstract
Eugenol (EU) has been shown to ameliorate experimental colitis due to its anti-oxidant and anti-inflammatory bioactivities. In this study, DSS-induced acute colitis was established and applied to clarify the regulation efficacy of EU on intestinal barrier impairment and macrophage polarization imbalance along with the inflammatory response. Besides, the adjusting effect of EU on macrophages was further investigated in vitro. The results confirmed that EU intervention alleviated DSS-induced colitis through methods such as restraining weight loss and colonic shortening and decreasing DAI scores. Microscopic observation manifested that EU maintained the intestinal barrier integrity in line with the mucus barrier and tight junction protection. Furthermore, EU intervention significantly suppressed the activation of TLR4/MyD88/NF-[Formula: see text]B signaling pathways and pro-inflammatory cytokines gene expressions, while enhancing the expressions of anti-inflammatory cytokines. Simultaneously, WB and FCM analyses of the CD86 and CD206 showed that EU could regulate the DSS-induced macrophage polarization imbalance. Overall, our data further elucidated the mechanism of EU's defensive effect on experimental colitis, which is relevant to the protective efficacy of intestinal barriers, inhibition of oxidative stress and excessive inflammatory response, and reprogramming of macrophage polarization. Hence, this study may facilitate a better understanding of the protective action of the EU against UC.
Collapse
Affiliation(s)
- Jun-Jie Huang
- College of Veterinary Medicine, Yangzhou University, Wenhui East Road 48, Yangzhou 225009, P. R. China
| | - Yue-Min Feng
- College of Veterinary Medicine, Yangzhou University, Wenhui East Road 48, Yangzhou 225009, P. R. China
| | - Shu-Mei Zheng
- College of Veterinary Medicine, Yangzhou University, Wenhui East Road 48, Yangzhou 225009, P. R. China
| | - Cheng-Long Yu
- College of Veterinary Medicine, Yangzhou University, Wenhui East Road 48, Yangzhou 225009, P. R. China
| | - Rui-Gang Zhou
- College of Veterinary Medicine, Yangzhou University, Wenhui East Road 48, Yangzhou 225009, P. R. China
| | - Ming-Jiang Liu
- College of Veterinary Medicine, Yangzhou University, Wenhui East Road 48, Yangzhou 225009, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, P. R. China
| | - Ruo-Nan Bo
- College of Veterinary Medicine, Yangzhou University, Wenhui East Road 48, Yangzhou 225009, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, P. R. China
| | - Jie Yu
- The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suzhi Road 120, Suqian 223800, P. R. China
| | - Jin-Gui Li
- College of Veterinary Medicine, Yangzhou University, Wenhui East Road 48, Yangzhou 225009, P. R. China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, P. R. China
| |
Collapse
|
6
|
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
|
7
|
Gutiérrez-Galicia JK, Drago-Serrano ME, Oros-Pantoja R, Godínez-Victoria M, Guzmán-Mejía F. Effect of chronic stress on gel-forming mucins in the small intestine of BALB/c mice. J Med Life 2024; 17:326-333. [PMID: 39044931 PMCID: PMC11262609 DOI: 10.25122/jml-2023-0473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/16/2024] [Indexed: 07/25/2024] Open
Abstract
Intestinal homeostasis involves the collaboration of gut barrier components, such as goblet cells and IgA-microbiota complexes, that are under the control of stress that promotes inflammatory responses addressed primarily in the colon. The aim of this study was to evaluate the effect of stress on mucins, goblet cells, and proinflammatory parameters in the proximal and distal regions of the small intestine. A group (n = 6) of female 8-week-old BALB/c mice underwent board immobilization stress (2 h per day for 4 days) and were sacrificed with isoflurane. Samples from proximal and distal small segments were collected to analyze the following: 1) goblet cells stained with periodic acid-Schiff (PAS) and with alcian blue (AB) to visualize histologically neutral and acidic mucins, respectively; 2) IgA-microbiota complexes identified by flow cytometry in intestinal lavages; and 3) MUC2, MUC5AC, and IL-18 mRNA levels in whole mucosal scrapings by reverse transcription-qPCR. Regarding the unstressed group, in the proximal region of small intestine both PAS+ and AB+ goblet cells were unchanged; however, MUC5AC and IL-18 mRNA levels were increased, and the percentage of IgA-microbiota complexes was reduced. In the distal segment, the number of PAS+ goblet cells was increased, whereas the number of AB+ goblet cells was reduced and did not affect the remaining parameters. The data suggest that stress induces inflammation in the proximal small intestine; these findings may provide an experimental reference for human diseases that may affect the proximal small intestine, such as Crohn's disease, in which stress contributes to the progression of intestinal inflammation or relapse.
Collapse
Affiliation(s)
- Jennifer Karume Gutiérrez-Galicia
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, México
| | - Maria Elisa Drago-Serrano
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México, México
| | - Rigoberto Oros-Pantoja
- Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca de Lerdo, México
| | - Marycarmen Godínez-Victoria
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, México
| | - Fabiola Guzmán-Mejía
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Ciudad de México, México
| |
Collapse
|
8
|
Fancy N, Nitin, Kniffen D, Melvin M, Kazemian N, Sadeghi J, Letef CA, D'Aloisio L, Copp AG, Inaba R, Hans G, Jafaripour S, Haskey N, Raman M, Daneshgar P, Chadee K, Ghosh S, Gibson DL, Pakpour S, Zandberg W, Bergstrom KSB. Fecal-adherent mucus is a non-invasive source of primary human MUC2 for structural and functional characterization in health and disease. J Biol Chem 2024; 300:105675. [PMID: 38272223 PMCID: PMC10891339 DOI: 10.1016/j.jbc.2024.105675] [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: 06/02/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/27/2024] Open
Abstract
The O-glycoprotein Mucin-2 (MUC2) forms the protective colon mucus layer. While animal models have demonstrated the importance of Muc2, few studies have explored human MUC2 in similar depth. Recent studies have revealed that secreted MUC2 is bound to human feces. We hypothesized human fecal MUC2 (HF-MUC2) was accessible for purification and downstream structural and functional characterization. We tested this via histologic and quantitative imaging on human fecal sections; extraction from feces for proteomic and O-glycomic characterization; and functional studies via growth and metabolic assays in vitro. Quantitative imaging of solid fecal sections showed a continuous mucus layer of varying thickness along human fecal sections with barrier functions intact. Lectin profiling showed HF-MUC2 bound several lectins but was weak to absent for Ulex europaeus 1 (α1,2 fucose-binding) and Sambucus nigra agglutinin (α2,6 sialic acid-binding), and did not have obvious b1/b2 barrier layers. HF-MUC2 separated by electrophoresis showed high molecular weight glycoprotein bands (∼1-2 MDa). Proteomics and Western analysis confirmed the enrichment of MUC2 and potential MUC2-associated proteins in HF-MUC2 extracts. MUC2 O-glycomics revealed diverse fucosylation, moderate sialylation, and little sulfation versus porcine colonic MUC2 and murine fecal Muc2. O-glycans were functional and supported the growth of Bacteroides thetaiotaomicron (B. theta) and short-chain fatty acid (SCFA) production in vitro. MUC2 could be similarly analyzed from inflammatory bowel disease stools, which displayed an altered glycomic profile and differential growth and SCFA production by B. theta versus healthy samples. These studies describe a new non-invasive platform for human MUC2 characterization in health and disease.
Collapse
Affiliation(s)
- Noah Fancy
- Biology, University of British Columbia-Okanagan, Kelowna, Canada
| | - Nitin
- Chemistry, University of British Columbia-Okanagan, Kelowna, Canada
| | - Darrek Kniffen
- Biology, University of British Columbia-Okanagan, Kelowna, Canada
| | - Mackenzie Melvin
- Biology, University of British Columbia-Okanagan, Kelowna, Canada
| | - Negin Kazemian
- School of Engineering, University of British Columbia-Okanagan, Kelowna, Canada
| | - Javad Sadeghi
- School of Engineering, University of British Columbia-Okanagan, Kelowna, Canada
| | - Clara A Letef
- Biology, University of British Columbia-Okanagan, Kelowna, Canada
| | - Leah D'Aloisio
- Biology, University of British Columbia-Okanagan, Kelowna, Canada
| | - Amanda G Copp
- Biology, University of British Columbia-Okanagan, Kelowna, Canada
| | - Rain Inaba
- Biology, University of British Columbia-Okanagan, Kelowna, Canada
| | - Geetkamal Hans
- Biology, University of British Columbia-Okanagan, Kelowna, Canada
| | - Simin Jafaripour
- Biology, University of British Columbia-Okanagan, Kelowna, Canada
| | - Natasha Haskey
- Biology, University of British Columbia-Okanagan, Kelowna, Canada
| | - Maitreyi Raman
- Cumming School of Medicine, University of Calgary, Calgary, Canada
| | | | - Kris Chadee
- Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Sanjoy Ghosh
- Biology, University of British Columbia-Okanagan, Kelowna, Canada
| | - Deanna L Gibson
- Biology, University of British Columbia-Okanagan, Kelowna, Canada
| | - Sepideh Pakpour
- School of Engineering, University of British Columbia-Okanagan, Kelowna, Canada
| | - Wesley Zandberg
- Chemistry, University of British Columbia-Okanagan, Kelowna, Canada
| | | |
Collapse
|
9
|
Miyazaki K, Sasaki A, Mizuuchi H. Advances in the Evaluation of Gastrointestinal Absorption Considering the Mucus Layer. Pharmaceutics 2023; 15:2714. [PMID: 38140055 PMCID: PMC10747107 DOI: 10.3390/pharmaceutics15122714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
Because of the increasing sophistication of formulation technology and the increasing polymerization of compounds directed toward undruggable drug targets, the influence of the mucus layer on gastrointestinal drug absorption has received renewed attention. Therefore, understanding the complex structure of the mucus layer containing highly glycosylated glycoprotein mucins, lipids bound to the mucins, and water held by glycans interacting with each other is critical. Recent advances in cell culture and engineering techniques have led to the development of evaluation systems that closely mimic the ecological environment and have been applied to the evaluation of gastrointestinal drug absorption while considering the mucus layer. This review provides a better understanding of the mucus layer components and the gastrointestinal tract's biological defense barrier, selects an assessment system for drug absorption in the mucus layer based on evaluation objectives, and discusses the overview and features of each assessment system.
Collapse
Affiliation(s)
- Kaori Miyazaki
- DMPK Research Laboratories, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida, Aoba-ku, Yokohama 227-0033, Japan; (A.S.); (H.M.)
| | | | | |
Collapse
|
10
|
Mules TC, Inns S, Le Gros G. Helminths' therapeutic potential to treat intestinal barrier dysfunction. Allergy 2023; 78:2892-2905. [PMID: 37449458 DOI: 10.1111/all.15812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/20/2023] [Accepted: 07/02/2023] [Indexed: 07/18/2023]
Abstract
The intestinal barrier is a dynamic multi-layered structure which can adapt to environmental changes within the intestinal lumen. It has the complex task of allowing nutrient absorption while limiting entry of harmful microbes and microbial antigens present in the intestinal lumen. Excessive entry of microbial antigens via microbial translocation due to 'intestinal barrier dysfunction' is hypothesised to contribute to the increasing incidence of allergic, autoimmune and metabolic diseases, a concept referred to as the 'epithelial barrier theory'. Helminths reside in the intestinal tract are in intimate contact with the mucosal surfaces and induce a range of local immunological changes which affect the layers of the intestinal barrier. Helminths are proposed to prevent, or even treat, many of the diseases implicated in the epithelial barrier theory. This review will focus on the effect of helminths on intestinal barrier function and explore whether this could explain the proposed health benefits delivered by helminths.
Collapse
Affiliation(s)
- Thomas C Mules
- Malaghan Institute of Medical Research, Wellington, New Zealand
- University of Otago, Wellington, New Zealand
| | | | - Graham Le Gros
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
11
|
Abaidullah M, La S, Liu M, Liu B, Cui Y, Wang Z, Sun H, Ma S, Shi Y. Polysaccharide from Smilax glabra Roxb Mitigates Intestinal Mucosal Damage by Therapeutically Restoring the Interactions between Gut Microbiota and Innate Immune Functions. Nutrients 2023; 15:4102. [PMID: 37836386 PMCID: PMC10574425 DOI: 10.3390/nu15194102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/09/2023] [Accepted: 09/13/2023] [Indexed: 10/15/2023] Open
Abstract
Smilax glabra Roxb (S. glabra) is a conventional Chinese medicine that is mainly used for the reliability of inflammation. However, bioactive polysaccharides from S. glabra (SGPs) have not been thoroughly investigated. Here, we demonstrate for the first time that SGPs preserve the integrity of the gut epithelial layer and protect against intestinal mucosal injury induced by dextran sulfate sodium. Mechanistically, SGPs mitigated colonic mucosal injury by restoring the association between the gut flora and innate immune functions. In particular, SGPs increased the number of goblet cells, reduced the proportion of apoptotic cells, improved the differentiation of gut tight junction proteins, and enhanced mucin production in the gut epithelial layer. Moreover, SGPs endorsed the propagation of probiotic bacteria, including Lachnospiraceae bacterium, which strongly correlated with decreased pro-inflammatory cytokines via the blocking of the TLR-4 NF-κB and MyD88 pathways. Overall, our study establishes a novel use of SGPs for the treatment of inflammatory bowel disease (IBD)-associated mucosal injury and provides a basis for understanding the therapeutic effects of natural polysaccharides from the perspective of symbiotic associations between host innate immune mechanisms and the gut microbiome.
Collapse
Affiliation(s)
- Muhammad Abaidullah
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (M.A.); (S.L.); (M.L.); (B.L.); (Y.C.); (Z.W.); (H.S.); (S.M.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| | - Shaokai La
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (M.A.); (S.L.); (M.L.); (B.L.); (Y.C.); (Z.W.); (H.S.); (S.M.)
| | - Mengqi Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (M.A.); (S.L.); (M.L.); (B.L.); (Y.C.); (Z.W.); (H.S.); (S.M.)
| | - Boshuai Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (M.A.); (S.L.); (M.L.); (B.L.); (Y.C.); (Z.W.); (H.S.); (S.M.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| | - Yalei Cui
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (M.A.); (S.L.); (M.L.); (B.L.); (Y.C.); (Z.W.); (H.S.); (S.M.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| | - Zhichang Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (M.A.); (S.L.); (M.L.); (B.L.); (Y.C.); (Z.W.); (H.S.); (S.M.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| | - Hao Sun
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (M.A.); (S.L.); (M.L.); (B.L.); (Y.C.); (Z.W.); (H.S.); (S.M.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| | - Sen Ma
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (M.A.); (S.L.); (M.L.); (B.L.); (Y.C.); (Z.W.); (H.S.); (S.M.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| | - Yinghua Shi
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (M.A.); (S.L.); (M.L.); (B.L.); (Y.C.); (Z.W.); (H.S.); (S.M.)
- Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou 450002, China
- Henan Forage Engineering Technology Research Center, Zhengzhou 450002, China
| |
Collapse
|
12
|
Park IS, Kim JH, Yu J, Shin Y, Kim K, Kim TI, Kim SW, Cheon JH. Bifidobacterium breve CBT BR3 is effective at relieving intestinal inflammation by augmenting goblet cell regeneration. J Gastroenterol Hepatol 2023; 38:1346-1354. [PMID: 37157108 DOI: 10.1111/jgh.16209] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/20/2023] [Accepted: 04/25/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND AND AIM Bifidobacterium breve was the first bacteria isolated in the feces of healthy infants and is a dominant species in the guts of breast-fed infants. Some strains of B. breve have been shown to be effective at relieving intestinal inflammation, but the modes of action have yet to be elucidated. In this study, we investigated the mechanisms of action of B. breve CBT BR3 isolated from South Korean infant feces in relieving colitis in vitro and in vivo. METHODS Colitis was induced in mice with dextran sodium sulfate (DSS) and dinitrobenzene sulfonic acid (DNBS). Quantitative reverse-transcription polymerase chain reaction, in vitro FITC-dextran flux permeability assay, and aryl hydrocarbon receptor (AhR) luciferase assay are performed using Caco-2 cells and HT29-Lucia™ AhR cells. RESULTS B. breve CBT BR3 was orally administered. B. breve CBT BR3 improved colitis symptoms in both DSS- and DNBS-induced colitis models. B. breve CBT BR3 increased the number of goblet cells per crypt. B. breve increased the mRNA expressions of Notch, Spdef, Muc5, and Il22. The mRNA expressions of Occludin, which encodes a membrane tight-junction protein, and Foxo3, which encodes a protein related to butyrate metabolism, were also increased in the DSS- and DNBS-induced colitis models. B. breve CBT BR3 protected inflammation-induced epithelial cell permeability and improved goblet cell function by inducing aryl hydrocarbon receptor in vitro. CONCLUSIONS These results indicate that B. breve CBT BR3 is effective at relieving intestinal inflammation by augmenting goblet cell regeneration.
Collapse
Affiliation(s)
- I Seul Park
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hyung Kim
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Jongwook Yu
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - YooJin Shin
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Kibeom Kim
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Tae Il Kim
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Won Kim
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Hee Cheon
- Department of Internal Medicine and Institute of Gastroenterology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
13
|
Golubkova A, Hunter CJ. Development of the Neonatal Intestinal Barrier, Microbiome, and Susceptibility to NEC. Microorganisms 2023; 11:1247. [PMID: 37317221 PMCID: PMC10221463 DOI: 10.3390/microorganisms11051247] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 06/16/2023] Open
Abstract
The function of the intestinal barrier is partially dependent on host maturity and the colonization patterns of the microbiome to which it is exposed. Premature birth and stressors of neonatal intensive care unit (NICU)-related support (e.g., antibiotics, steroids, etc.) can alter the host internal environment resulting in changes in the intestinal barrier. Pathogenic microbial proliferation and breach of the immature intestinal barrier are proposed to be crucial steps in the development of neonatal diseases such as necrotizing enterocolitis. This article will review the current literature on the intestinal barrier in the neonatal gut, the consequences of microbiome development for this defense system, and how prematurity can influence neonatal susceptibility to gastrointestinal infection.
Collapse
Affiliation(s)
| | - Catherine J. Hunter
- Division of Pediatric Surgery, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
14
|
Hamamoto Y, Kawamura M, Uchida H, Takagahara K, Katori C, Asai H, Harada H, Shimizu S, Morii E, Yoshida K. Aberrant MUC Immunohistochemical Expressions in Inflammatory Bowel Diseases. Appl Immunohistochem Mol Morphol 2023; 31:107-112. [PMID: 36728756 DOI: 10.1097/pai.0000000000001096] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 11/17/2022] [Indexed: 02/03/2023]
Abstract
Ulcerative colitis (UC) and Crohn disease (CD) are cryptogenic inflammatory bowel diseases that are suggestive of aberrant mucin (MUC) expression; however, their relationship remains unclear. Here, we examined aberrant MUC expression in intestinal samples from UC and CD patients in comparison to samples from patients with ischemic colitis and control groups. To study the expression of MUC1 , MUC5AC , and MUC6 in different patient groups, we reviewed the slides stained with hematoxylin and eosin and performed immunohistochemistry. The results revealed that MUC1 was expressed more in the UC group and MUC6 in the CD group. No significant changes were observed in MUC expression in the ischemic colitis group. Overall, we demonstrated changes in MUC expression in UC and CD, which can help in the diagnosis and early clinical management of UC and CD.
Collapse
Affiliation(s)
- Yuichiro Hamamoto
- Department of Diagnostic Pathology
- Department of Pathology, Osaka University Graduate School of Medicine, Suita
| | | | - Hiroki Uchida
- Department of Clinical Laboratory, Kinki Central Hospital, Itami, Hyogo
| | - Kojiro Takagahara
- Department of Clinical Laboratory, Kinki Central Hospital, Itami, Hyogo
| | - Chiaki Katori
- Department of Clinical Laboratory, Kinki Central Hospital, Itami, Hyogo
| | - Hinako Asai
- Department of Clinical Laboratory, Kinki Central Hospital, Itami, Hyogo
| | | | - Shigeki Shimizu
- Department of Clinical Laboratory, National Hospital Organization Kinki-Chuo Chest Medical Center, Kita-ku, Sakai, Osaka, Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, Suita
| | - Kyotaro Yoshida
- Department of Diagnostic Pathology
- Department of Clinical Laboratory, Kinki Central Hospital, Itami, Hyogo
| |
Collapse
|
15
|
Løkka G, Dhanasiri AKS, Krogdahl Å, Kortner TM. Bile components affect the functions and transcriptome of the rainbow trout intestinal epithelial cell line RTgutGC. FISH & SHELLFISH IMMUNOLOGY 2022; 131:1144-1156. [PMID: 36444097 DOI: 10.1016/j.fsi.2022.10.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/17/2022] [Accepted: 10/23/2022] [Indexed: 06/16/2023]
Abstract
The concomitant increase in cultivation of fish and decrease in supply of marine ingredients, have greatly increased the demand for new nutrient sources. This also regards so-called functional ingredients which may benefit health and welfare of the fish. In vitro cell line-based intestinal epithelial barrier models may serve as tools for narrowing down the broad range of ingredient options, to identify the most promising candidates before in vivo feeding trials are run. In vivo, differentiation of the various epithelial cells in the fish intestine, from the multipotent stem cells, takes place in the presence of a variety of substances from dietary and endogenous origin. Among these, bile salts have recently received attention as regulators of epithelial function in health and disease but have not, until now, been included in the medium when culturing fish gut epithelial cells in vitro. As bile salts are present at high levels in the chyme of the fish intestine, in particular in salmon and rainbow trout, mostly as taurocholate (>90%), their role for effects of diet ingredients on the in vitro gut cell model should be understood. With this study, we wanted to investigate whether inclusion of bile from rainbow trout or pure taurocholate in the culture media would modulate functions of the RTgutGC epithelial cells. Here, we demonstrated that the rainbow trout intestinal epithelial cell line RTgutGC responded significantly to the presence of bile components. Treatment with rainbow trout bile taken from the gall bladder (RTbile) or pure taurocholate (TC) at taurocholate concentrations of ≤0.5 mg/mL retained normal cell morphology, cell viability as in cell oxidation-reduction metabolic activity and membrane integrity, and barrier features, while high concentrations of bile salts (≥1 mg/mL) were cytotoxic to the cells. After long-term (4 days) bile treatment, transcriptome responses showed how bile salts play important roles in intestinal epithelial cell metabolism. qPCR data demonstrated that barrier function genes, brush border enzyme genes and immune genes were significantly affected. Although similar trends were seen, treatment with bile salt as a component of rainbow trout bile or pure taurocholate, induced somewhat different effects. In conclusion, this study clearly indicates that bile salts should be included in the cell medium when running in vitro studies of gut cell functions, not at least immune functions, preferably at the level of ∼0.5 mg/mL supplemented as pure taurocholate to ensure reproducibility.
Collapse
Affiliation(s)
- Guro Løkka
- Nutrition and Health Unit, Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, P.O. Box 5003, NO-1432 Ås, Norway.
| | - Anusha K S Dhanasiri
- Nutrition and Health Unit, Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, P.O. Box 5003, NO-1432 Ås, Norway
| | - Åshild Krogdahl
- Nutrition and Health Unit, Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, P.O. Box 5003, NO-1432 Ås, Norway
| | - Trond M Kortner
- Nutrition and Health Unit, Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, P.O. Box 5003, NO-1432 Ås, Norway
| |
Collapse
|
16
|
Busch M, Ramachandran H, Wahle T, Rossi A, Schins RPF. Investigating the Role of the NLRP3 Inflammasome Pathway in Acute Intestinal Inflammation: Use of THP-1 Knockout Cell Lines in an Advanced Triple Culture Model. Front Immunol 2022; 13:898039. [PMID: 35911682 PMCID: PMC9326178 DOI: 10.3389/fimmu.2022.898039] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/16/2022] [Indexed: 01/09/2023] Open
Abstract
The NLRP3 inflammasome plays an important role in intestinal homeostasis as well as inflammation. However, in vivo studies investigating the role of the NLRP3 inflammasome in inflammatory bowel disease (IBD) report contrasting results, leaving it unclear if the NLRP3 inflammasome augments or attenuates intestinal inflammation. To investigate the role of the NLRP3/caspase-1 pathway in a model of acute intestinal inflammation, we modified a previously established in vitro triple culture model of the healthy and inflamed intestine (Caco-2/HT29-MTX-E12/THP-1). Using THP-1 knockout cell lines, we analyzed how the NLRP3 inflammasome and its downstream enzyme caspase-1 (CASP1) affect inflammatory parameters including barrier integrity and cytotoxicity, as well as gene expression and secretion of pro-inflammatory cytokines and mucus. Furthermore, we investigated differences in inflammation-mediated cytotoxicity towards enterocyte-like (Caco-2) or goblet-like (HT29-MTX-E12) epithelial cells. As a complementary approach, inflammation-related cytotoxicity and gene expression of cytokines was analyzed in intestinal tissue explants from wildtype (WT) and Nlrp3-/- mice. Induction of intestinal inflammation impaired the barrier, caused cytotoxicity, and altered gene expression of pro-inflammatory cytokines and mucins in vitro, while the knockout of NLRP3 and CASP1 in THP 1 cells led to attenuation of these inflammatory parameters. The knockout of CASP1 tended to show a slightly stronger attenuating effect compared to the NLRP3 knockout model. We also found that the inflammation-mediated death of goblet-like cells is NLRP3/caspase-1 dependent. Furthermore, inflammation-related cytotoxicity and upregulation of pro-inflammatory cytokines was present in ileal tissue explants from WT, but not Nlrp3-/- mice. The here presented observations indicate a pro-inflammatory and adverse role of the NLRP3 inflammasome in macrophages during acute intestinal inflammation.
Collapse
|
17
|
Bein A, Fadel CW, Swenor B, Cao W, Powers RK, Camacho DM, Naziripour A, Parsons A, LoGrande N, Sharma S, Kim S, Jalili-Firoozinezhad S, Grant J, Breault DT, Iqbal J, Ali A, Denson LA, Moore SR, Prantil-Baun R, Goyal G, Ingber DE. Nutritional deficiency in an intestine-on-a-chip recapitulates injury hallmarks associated with environmental enteric dysfunction. Nat Biomed Eng 2022; 6:1236-1247. [PMID: 35739419 DOI: 10.1038/s41551-022-00899-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 05/12/2022] [Indexed: 01/03/2023]
Abstract
Environmental enteric dysfunction (EED)-a chronic inflammatory condition of the intestine-is characterized by villus blunting, compromised intestinal barrier function and reduced nutrient absorption. Here we show that essential genotypic and phenotypic features of EED-associated intestinal injury can be reconstituted in a human intestine-on-a-chip lined by organoid-derived intestinal epithelial cells from patients with EED and cultured in nutrient-deficient medium lacking niacinamide and tryptophan. Exposure of the organ chip to such nutritional deficiencies resulted in congruent changes in six of the top ten upregulated genes that were comparable to changes seen in samples from patients with EED. Chips lined with healthy epithelium or with EED epithelium exposed to nutritional deficiencies resulted in severe villus blunting and barrier dysfunction, and in the impairment of fatty acid uptake and amino acid transport; and the chips with EED epithelium exhibited heightened secretion of inflammatory cytokines. The organ-chip model of EED-associated intestinal injury may facilitate the analysis of the molecular, genetic and nutritional bases of the disease and the testing of candidate therapeutics for it.
Collapse
Affiliation(s)
- Amir Bein
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.,Quris Technologies, Boston, MA, USA
| | - Cicely W Fadel
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA.,Division of Neonatology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Ben Swenor
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Wuji Cao
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Rani K Powers
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.,Pluto Biosciences, Inc., Golden, CO, USA
| | - Diogo M Camacho
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.,Rheos Medicines, Cambridge, MA, USA
| | - Arash Naziripour
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Andrew Parsons
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Nina LoGrande
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Sanjay Sharma
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Seongmin Kim
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Sasan Jalili-Firoozinezhad
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.,Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Jennifer Grant
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - David T Breault
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.,Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA.,Harvard Stem Cell Institute, Harvard University, Boston, MA, USA
| | - Junaid Iqbal
- Department of Paediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Asad Ali
- Department of Paediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Lee A Denson
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sean R Moore
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology, and Nutrition, University of Virginia, Charlottesville, VA, USA
| | - Rachelle Prantil-Baun
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Girija Goyal
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA. .,Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA. .,Vascular Biology Program and Department of Surgery, Harvard Medical School and Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
18
|
Keeler SP, Yantis J, Gerovac BJ, Youkilis SL, Podgorny S, Mao D, Zhang Y, Whitworth KM, Redel B, Samuel MS, Wells KD, Prather RS, Holtzman MJ. Chloride channel accessory 1 gene deficiency causes selective loss of mucus production in a new pig model. Am J Physiol Lung Cell Mol Physiol 2022; 322:L842-L852. [PMID: 35438004 PMCID: PMC9142155 DOI: 10.1152/ajplung.00443.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/03/2022] [Accepted: 04/13/2022] [Indexed: 12/30/2022] Open
Abstract
Morbidity and mortality of respiratory diseases are linked to airway obstruction by mucus but there are still no specific, safe, and effective drugs to correct this phenotype. The need for better treatment requires a new understanding of the basis for mucus production. In that regard, studies of human airway epithelial cells in primary culture show that a mucin granule constituent known as chloride channel accessory 1 (CLCA1) is required for inducible expression of the inflammatory mucin MUC5AC in response to potent type 2 cytokines. However, it remained uncertain whether CLCLA1 is necessary for mucus production in vivo. Conventional approaches to functional biology using targeted gene knockout were difficult due to the functional redundancy of additional Clca genes in mice not found in humans. We reasoned that CLCA1 function might be better addressed in pigs that maintain the same four-member CLCA gene locus and the corresponding mucosal and submucosal populations of mucous cells found in humans. Here we develop to our knowledge the first CLCA1-gene-deficient (CLCA1-/-) pig and show that these animals exhibit loss of MUC5AC+ mucous cells throughout the airway mucosa of the lung without affecting comparable cells in the tracheal mucosa or MUC5B+ mucous cells in submucosal glands. Similarly, CLCA1-/- pigs exhibit loss of MUC5AC+ mucous cells in the intestinal mucosa without affecting MUC2+ mucous cells. These data establish CLCA1 function for controlling MUC5AC expression as a marker of mucus production and provide a new animal model to study mucus production at respiratory and intestinal sites.
Collapse
Affiliation(s)
- Shamus P Keeler
- Drug Discovery Program, Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Jennifer Yantis
- Drug Discovery Program, Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Benjamin J Gerovac
- Drug Discovery Program, Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Samuel L Youkilis
- Drug Discovery Program, Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Stephanie Podgorny
- Drug Discovery Program, Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Dailing Mao
- Drug Discovery Program, Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Yong Zhang
- Drug Discovery Program, Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Kristin M Whitworth
- Division of Animal Sciences, National Swine Resource and Research Center, University of Missouri, Columbia, Missouri
| | - Bethany Redel
- Division of Animal Sciences, National Swine Resource and Research Center, University of Missouri, Columbia, Missouri
| | - Melissa S Samuel
- Division of Animal Sciences, National Swine Resource and Research Center, University of Missouri, Columbia, Missouri
| | - Kevin D Wells
- Division of Animal Sciences, National Swine Resource and Research Center, University of Missouri, Columbia, Missouri
| | - Randall S Prather
- Division of Animal Sciences, National Swine Resource and Research Center, University of Missouri, Columbia, Missouri
| | - Michael J Holtzman
- Drug Discovery Program, Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
19
|
Breugelmans T, Oosterlinck B, Arras W, Ceuleers H, De Man J, Hold GL, De Winter BY, Smet A. The role of mucins in gastrointestinal barrier function during health and disease. Lancet Gastroenterol Hepatol 2022; 7:455-471. [DOI: 10.1016/s2468-1253(21)00431-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 12/23/2022]
|
20
|
Serigado JM, Foulke-Abel J, Hines WC, Hanson JA, In J, Kovbasnjuk O. Ulcerative Colitis: Novel Epithelial Insights Provided by Single Cell RNA Sequencing. Front Med (Lausanne) 2022; 9:868508. [PMID: 35530046 PMCID: PMC9068527 DOI: 10.3389/fmed.2022.868508] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/14/2022] [Indexed: 12/22/2022] Open
Abstract
Ulcerative Colitis (UC) is a chronic inflammatory disease of the intestinal tract for which a definitive etiology is yet unknown. Both genetic and environmental factors have been implicated in the development of UC. Recently, single cell RNA sequencing (scRNA-seq) technology revealed cell subpopulations contributing to the pathogenesis of UC and brought new insight into the pathways that connect genome to pathology. This review describes key scRNA-seq findings in two major studies by Broad Institute and University of Oxford, investigating the transcriptomic landscape of epithelial cells in UC. We focus on five major findings: (1) the identification of BEST4 + cells, (2) colonic microfold (M) cells, (3) detailed comparison of the transcriptomes of goblet cells, and (4) colonocytes and (5) stem cells in health and disease. In analyzing the two studies, we identify the commonalities and differences in methodologies, results, and conclusions, offering possible explanations, and validated several cell cluster markers. In systematizing the results, we hope to offer a framework that the broad scientific GI community and GI clinicians can use to replicate or corroborate the extensive new findings that RNA-seq offers.
Collapse
Affiliation(s)
- Joao M. Serigado
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Jennifer Foulke-Abel
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - William C. Hines
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Joshua A Hanson
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Julie In
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Olga Kovbasnjuk
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
- *Correspondence: Olga Kovbasnjuk,
| |
Collapse
|
21
|
Manuel CA, Johnson LK, Pugazhenthi U, Fong DL, Fink M, Habenicht LM, Leszczynski JK, Diana IR, Schurr MJ, Frank DN. Effect of Antimicrobial Prophylaxis on Corynebacterium bovis Infection and the Skin Microbiome of Immunodeficient Mice. Comp Med 2022; 72:78-89. [PMID: 35379380 PMCID: PMC9084569 DOI: 10.30802/aalas-cm-21-000082] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/21/2021] [Accepted: 12/22/2021] [Indexed: 11/05/2022]
Abstract
Corynebacterium bovis is an opportunistic pathogen of the skin of immunodeficient mice and is sensitive to oral antibiotics that reach therapeutic blood concentrations. However, prophylactic antibiotics are considered to be ineffective at preventing C. bovis infection. In addition, the effect of C. bovis on the skin microbiome (SM) of common immunodeficient mouse strains has yet to be characterized. Consequently, we evaluated whether oral prophylactic antibiotics prevent C. bovis infection after inoculation. An infectious dose of C. bovis was applied to the skin of Hsd:Athymic Nude (nude) and NOD. Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mice. Mice were then housed individually and assigned randomly to receive either untreated drinking water (Cb+Abx-group) or prophylactic amoxicillin-clavulanic acid in the drinking water (0.375 mg/mL) for 14 d (Cb+Abx+group). A third treatment group of each mouse strain was uninoculated and untreated (Cb-Abx-group). Mice from all groups were serially sampled by using dermal swabs to monitor C. bovis infection via quantitative real-time PCR and the SM via 16S rRNA sequence analysis. Fourteen days of prophylactic antibiotics prevented the perpetuation of C. bovis skin infection in both strains. Only the combination of C. bovis inoculation and oral antibiotics (Cb+Abx+) significantly affected the SM of NSG mice at day 14; this effect resolved by the end of the study (day 70). In mice that did not receive antibiotics, C. bovis significantly altered the SM of nude mice but not NSG mice at days 14 and 70. These findings demonstrate the potential benefit of prophylactic antibiotics for prevention of C. bovis infection. However, indirect effect of antibiotics on commensal bacteria and potential effects on xenograft models must be considered.
Collapse
Affiliation(s)
- Christopher A Manuel
- Office of Laboratory Animal Resources, University of Colorado Cancer Center, Aurora, CO; Department of Pathology, University of Colorado Cancer Center, Aurora, CO; University of Colorado Cancer Center, Aurora, CO;,
| | - Linda K Johnson
- Department of Pathology, University of Colorado Cancer Center, Aurora, CO; Deceased
| | - Uma Pugazhenthi
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Derek L Fong
- Office of Laboratory Animal Resources, University of Colorado Cancer Center, Aurora, CO; Department of Pathology, University of Colorado Cancer Center, Aurora, CO
| | - Michaelk Fink
- Office of Laboratory Animal Resources, University of Colorado Cancer Center, Aurora, CO; Department of Pathology; University of Colorado Cancer Center, Aurora, CO
| | - Lauren M Habenicht
- Office of Laboratory Animal Resources, University of Colorado Cancer Center, Aurora, CO; Department of Pathology, University of Colorado Cancer Center, Aurora, CO
| | - Jori K Leszczynski
- Office of Laboratory Animal Resources, University of Colorado Cancer Center, Aurora, CO; Department of Pathology, University of Colorado Cancer Center, Aurora, CO
| | - I R Diana
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Michael J Schurr
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Daniel N Frank
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
22
|
Liu H, Gao P, Jia B, Lu N, Zhu B, Zhang F. IBD-Associated Atg16L1T300A Polymorphism Regulates Commensal Microbiota of the Intestine. Front Immunol 2022; 12:772189. [PMID: 35154071 PMCID: PMC8829142 DOI: 10.3389/fimmu.2021.772189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/13/2021] [Indexed: 11/13/2022] Open
Abstract
The development of inflammatory bowel disease (IBD) is driven by the interaction among host genetics, microbiota, and the immune system of the entire digestive tract. Atg16L1T300A polymorphism is a genetic factor that confers increased risk for the pathogenesis of Crohn's disease. However, the exact contributions of Atg16L1T300A to intestinal mucosal homeostasis are not well understood. Here we show that Atg16L1T300A polymorphism impacts commensal bacterial flora in the intestine under a steady state. Analysis of intestinal bacteria from Atg16L1T300A/T300A mice showed that they harbored an altered microbiota in both the terminal ileum and colon compared to cohoused WT mice. Interestingly, Atg16L1T300A/T300A mice harbored a significant increase in the abundance of Tyzzerella, Mucispirillum, Ruminococcaceae, and Cyanobacteria which were known associated with IBD. Moreover, Akkermansia, a bacterium that is mucin-associated, was reduced greatly in Atg16L1T300A/T300A mice. Further analysis indicated that goblet cells of Atg16L1T300A/T300A mice had diminished mucin secretion that resulted from defective autophagy. Finally, Atg16L1T300A/T300A mice developed more severe inflammation in the DSS colitis model than in WT mice. These results indicate that the altered microbiota in Atg16L1T300A/T300A mice might be an important factor that contributed to the risk of Atg16L1T300A carriers to Crohn's disease and supports a multi-hit disease model involving specific gene-microbe interactions.
Collapse
Affiliation(s)
- Hongtao Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.,College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Ping Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Baoqian Jia
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Na Lu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Baoli Zhu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.,Department of Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Fuping Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.,Department of Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
23
|
Role of microRNAs in the Pathophysiology of Ulcerative Colitis. IMMUNO 2021. [DOI: 10.3390/immuno1040039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Ulcerative colitis (UC) is an intractable disorder characterized by a chronic inflammation of the colon. Studies have identified UC as a multifactorial disorder affected by both genetic and environmental factors; however, the precise mechanism remains unclear. Recent advances in the field of microRNA (miRNA) research have identified an association between this small non-coding RNA in the pathophysiology of UC and altered miRNA expression profiles in patients with UC. Nevertheless, the roles of individual miRNAs are uncertain due to heterogeneity in both research samples and clinical backgrounds. In this review, we focus on miRNA expression in colonic mucosa where inflammation occurs in UC and discuss the potential roles of individual miRNAs in disease development, outlining the pathophysiology of UC.
Collapse
|
24
|
Bredeck G, Kämpfer AAM, Sofranko A, Wahle T, Büttner V, Albrecht C, Schins RPF. Ingested Engineered Nanomaterials Affect the Expression of Mucin Genes-An In Vitro-In Vivo Comparison. NANOMATERIALS 2021; 11:nano11102621. [PMID: 34685068 PMCID: PMC8537393 DOI: 10.3390/nano11102621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 12/22/2022]
Abstract
The increasing use of engineered nanomaterials (ENM) in food has fueled the development of intestinal in vitro models for toxicity testing. However, ENM effects on intestinal mucus have barely been addressed, although its crucial role for intestinal health is evident. We investigated the effects of ENM on mucin expression and aimed to evaluate the suitability of four in vitro models of increasing complexity compared to a mouse model exposed through feed pellets. We assessed the gene expression of the mucins MUC1, MUC2, MUC5AC, MUC13 and MUC20 and the chemokine interleukin-8 in pre-confluent and confluent HT29-MTX-E12 cells, in stable and inflamed triple cultures of Caco-2, HT29-MTX-E12 and THP-1 cells, and in the ileum of mice following exposure to TiO2, Ag, CeO2 or SiO2. All ENM had shared and specific effects. CeO2 downregulated MUC1 in confluent E12 cells and in mice. Ag induced downregulation of Muc2 in mice. Overall, the in vivo data were consistent with the findings in the stable triple cultures and the confluent HT29-MTX-E12 cells but not in pre-confluent cells, indicating the higher relevance of advanced models for hazard assessment. The effects on MUC1 and MUC2 suggest that specific ENM may lead to an elevated susceptibility towards intestinal infections and inflammations.
Collapse
|
25
|
Gut Microbiota and Dietary Factors as Modulators of the Mucus Layer in Inflammatory Bowel Disease. Int J Mol Sci 2021; 22:ijms221910224. [PMID: 34638564 PMCID: PMC8508624 DOI: 10.3390/ijms221910224] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 09/20/2021] [Indexed: 12/14/2022] Open
Abstract
The gastrointestinal tract is optimized to efficiently absorb nutrients and provide a competent barrier against a variety of lumen environmental compounds. Different regulatory mechanisms jointly collaborate to maintain intestinal homeostasis, but alterations in these mechanisms lead to a dysfunctional gastrointestinal barrier and are associated to several inflammatory conditions usually found in chronic pathologies such as inflammatory bowel disease (IBD). The gastrointestinal mucus, mostly composed of mucin glycoproteins, covers the epithelium and plays an essential role in digestive and barrier functions. However, its regulation is very dynamic and is still poorly understood. This review presents some aspects concerning the role of mucus in gut health and its alterations in IBD. In addition, the impact of gut microbiota and dietary compounds as environmental factors modulating the mucus layer is addressed. To date, studies have evidenced the impact of the three-way interplay between the microbiome, diet and the mucus layer on the gut barrier, host immune system and IBD. This review emphasizes the need to address current limitations on this topic, especially regarding the design of robust human trials and highlights the potential interest of improving our understanding of the regulation of the intestinal mucus barrier in IBD.
Collapse
|
26
|
Wang Z, Guo K, Gao P, Pu Q, Lin P, Qin S, Xie N, Hur J, Li C, Huang C, Wu M. Microbial and genetic-based framework identifies drug targets in inflammatory bowel disease. Theranostics 2021; 11:7491-7506. [PMID: 34158863 PMCID: PMC8210594 DOI: 10.7150/thno.59196] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/14/2021] [Indexed: 02/05/2023] Open
Abstract
Rationale: With increasing incidence and prevalence of inflammatory bowel disease (IBD), it has become one of the major public health threats, and there is an urgent need to develop new therapeutic agents. Although the pathogenesis of IBD is still unclear, previous research has provided evidence for complex interplays between genetic, immune, microbial, and environmental factors. Here, we constructed a gene-microbiota interaction-based framework to discover IBD biomarkers and therapeutics. Methods: We identified candidate biomarkers for IBD by analyzing the publicly available transcriptomic and microbiome data from IBD cohorts. Animal models of IBD and diarrhea were established. The inflammation-correlated microbial and genetic variants in gene knockout mice were identified by 16S rRNA sequences and PCR array. We performed bioinformatic analysis of microbiome functional prediction and drug repurposing. Our validation experiments with cells and animals confirmed anti-inflammatory properties of a drug candidate. Results: We identified the DNA-sensing enzyme cyclic GMP-AMP synthase (cGAS) as a potential biomarker for IBD in both patients and murine models. cGAS knockout mice were less susceptible to DSS-induced colitis. cGAS-associated gut microbiota and host genetic factors relating to IBD pathogenesis were also identified. Using a computational drug repurposing approach, we predicted 43 candidate drugs with high potency to reverse colitis-associated gene expression and validated that brefeldin-a mitigates inflammatory response in colitis mouse model and colon cancer cell lines. Conclusions: By integrating computational screening, microbiota interference, gene knockout techniques, and in vitro and in vivo validation, we built a framework for predicting biomarkers and host-microbe interaction targets and identifying repurposing drugs for IBD, which may be tested further for clinical application. This approach may also be a tool for repurposing drugs for treating other diseases.
Collapse
Affiliation(s)
- Zhihan Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Kai Guo
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Pan Gao
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
- Medical Research Institute, Wuhan University, Wuhan 430071, China
| | - Qinqin Pu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Ping Lin
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing 400038, China
| | - Shugang Qin
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Na Xie
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Junguk Hur
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Changlong Li
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Canhua Huang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Min Wu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| |
Collapse
|
27
|
Tolomeo AM, Castagliuolo I, Piccoli M, Grassi M, Magarotto F, De Lazzari G, Malvicini R, Caicci F, Franzin C, Scarpa M, Macchi V, De Caro R, Angriman I, Viola A, Porzionato A, Pozzobon M, Muraca M. Extracellular Vesicles Secreted by Mesenchymal Stromal Cells Exert Opposite Effects to Their Cells of Origin in Murine Sodium Dextran Sulfate-Induced Colitis. Front Immunol 2021; 12:627605. [PMID: 33927713 PMCID: PMC8076641 DOI: 10.3389/fimmu.2021.627605] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/17/2021] [Indexed: 12/15/2022] Open
Abstract
Several reports have described a beneficial effect of Mesenchymal Stromal Cells (MSCs) and of their secreted extracellular vesicles (EVs) in mice with experimental colitis. However, the effects of the two treatments have not been thoroughly compared in this model. Here, we compared the effects of MSCs and of MSC-EV administration in mice with colitis induced by dextran sulfate sodium (DSS). Since cytokine conditioning was reported to enhance the immune modulatory activity of MSCs, the cells were kept either under standard culture conditions (naïve, nMSCs) or primed with a cocktail of pro-inflammatory cytokines, including IL1β, IL6 and TNFα (induced, iMSCs). In our experimental conditions, nMSCs and iMSCs administration resulted in both clinical and histological worsening and was associated with pro-inflammatory polarization of intestinal macrophages. However, mice treated with iEVs showed clinico-pathological improvement, decreased intestinal fibrosis and angiogenesis and a striking increase in intestinal expression of Mucin 5ac, suggesting improved epithelial function. Moreover, treatment with iEVs resulted in the polarization of intestinal macrophages towards and anti-inflammatory phenotype and in an increased Treg/Teff ratio at the level of the intestinal lymph node. Collectively, these data confirm that MSCs can behave either as anti- or as pro-inflammatory agents depending on the host environment. In contrast, EVs showed a beneficial effect, suggesting a more predictable behavior, a safer therapeutic profile and a higher therapeutic efficacy with respect to their cells of origin.
Collapse
Affiliation(s)
- Anna Maria Tolomeo
- Department of Women’s and Children’s Health, University of Padova, Padua, Italy
- Laboratory of Extracellular Vesicles as Therapeutic Tools, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Padua, Italy
| | | | - Martina Piccoli
- Laboratory of Tissue Engineering, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - Michele Grassi
- Department of Women’s and Children’s Health, University of Padova, Padua, Italy
| | - Fabio Magarotto
- Department of Women’s and Children’s Health, University of Padova, Padua, Italy
- Laboratory of Stem Cells and Regenerative Medicine, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - Giada De Lazzari
- Department of Women’s and Children’s Health, University of Padova, Padua, Italy
- Laboratory of Extracellular Vesicles as Therapeutic Tools, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Padua, Italy
| | - Ricardo Malvicini
- Laboratory of Extracellular Vesicles as Therapeutic Tools, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Padua, Italy
- Instituto de medicina traslacional, trasplante y bioingenieria (IMeTTyB-CONICET), Buenos Aires, Argentina
| | | | - Chiara Franzin
- Laboratory of Tissue Engineering, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - Melania Scarpa
- Laboratory of Advanced Translational Research, Veneto Institute of Oncology IOV–IRCCS, Padua, Italy
| | - Veronica Macchi
- Department of Neurosciences, University of Padova, Padua, Italy
| | - Raffaele De Caro
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Padua, Italy
- Department of Neurosciences, University of Padova, Padua, Italy
| | - Imerio Angriman
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Antonella Viola
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Andrea Porzionato
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Padua, Italy
- Department of Neurosciences, University of Padova, Padua, Italy
| | - Michela Pozzobon
- Department of Women’s and Children’s Health, University of Padova, Padua, Italy
- Laboratory of Stem Cells and Regenerative Medicine, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
| | - Maurizio Muraca
- Department of Women’s and Children’s Health, University of Padova, Padua, Italy
- Laboratory of Extracellular Vesicles as Therapeutic Tools, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padua, Italy
- L.i.f.e.L.a.b. Program, Consorzio per la Ricerca Sanitaria (CORIS), Padua, Italy
| |
Collapse
|
28
|
Carroll-Portillo A, Lin HC. Exploring Mucin as Adjunct to Phage Therapy. Microorganisms 2021; 9:microorganisms9030509. [PMID: 33670927 PMCID: PMC7997181 DOI: 10.3390/microorganisms9030509] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/21/2022] Open
Abstract
Conventional phage therapy using bacteriophages (phages) for specific targeting of pathogenic bacteria is not always useful as a therapeutic for gastrointestinal (GI) dysfunction. Complex dysbiotic GI disorders such as small intestinal bowel overgrowth (SIBO), ulcerative colitis (UC), or Crohn’s disease (CD) are even more difficult to treat as these conditions have shifts in multiple populations of bacteria within the microbiome. Such community-level structural changes in the gut microbiota may require an alternative to conventional phage therapy such as fecal virome transfer or a phage cocktail capable of targeting multiple bacterial species. Additionally, manipulation of the GI microenvironment may enhance beneficial bacteria–phage interactions during treatment. Mucin, produced along the entire length of the GI tract to protect the underlying mucosa, is a prominent contributor to the GI microenvironment and may facilitate bacteria–phage interactions in multiple ways, potentially serving as an adjunct during phage therapy. In this review, we will describe what is known about the role of mucin within the GI tract and how its facilitation of bacteria–phage interactions should be considered in any effort directed at optimizing effectiveness of a phage therapy for gastrointestinal dysbiosis.
Collapse
Affiliation(s)
- Amanda Carroll-Portillo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA;
| | - Henry C. Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA;
- Medicine Service, New Mexico VA Health Care System, Albuquerque, NM 87108, USA
- Correspondence: ; Tel.: +1-505-265-1711 (ext. 4552)
| |
Collapse
|
29
|
Flores-Sanchez F, Chavez-Dueñas L, Sanchez-Villamil J, Navarro-Garcia F. Pic Protein From Enteroaggregative E. coli Induces Different Mechanisms for Its Dual Activity as a Mucus Secretagogue and a Mucinase. Front Immunol 2020; 11:564953. [PMID: 33281812 PMCID: PMC7705071 DOI: 10.3389/fimmu.2020.564953] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022] Open
Abstract
A hallmark of enteroaggregative Escherichia coli (EAEC) infection is the formation of an intestinal biofilm, which comprises a mucus layer with immersed bacteria. Pic is an autotransporter secreted by EAEC, and other E. coli pathotypes, and has been involved in two apparently contradictory phenotypes, as a mucus secretagogue and as a mucinase. Here, we investigated this Pic dual activity, mucus secretagogue capability and mucinolytic activity, in human goblet cells that secrete MUC2 and MUC5AC. Pic induced mucus hypersecretion directly in the goblet cells, without other intestinal cell types involved. At the same time, Pic exhibited strong proteolytic activity on the secreted mucins. These activities were independent since a mutation in the serine protease motif (PicS258I) abolished mucin degradation while maintaining the mucus secretagogue activity intact. Furthermore, deoxycholic acid (DCA)-induced mucins were proteolytically degraded when goblet cells were co-incubated with DCA/Pic, while co-incubation with DCA/PicS258I induced a synergistic effect on mucus hypersecretion. Pic was more efficient degrading MUC5AC than MUC2, but no degradation was detected with Pic inactivated at the active site by mutation or pharmacological inhibition. Remarkably, Pic cleaved MUC2 and MUC5AC in the C-terminal domain, leaving N-terminal subproducts, impacting the feature of gel-forming mucins and allowing mucus layer penetration by EAEC. Astonishingly, Pic stimulated rapid mucin secretion in goblet-like cells by activating the intracellular calcium pathway resulting from the PLC signal transduction pathway, leading to the production of DAG and releasing IP3, a second messenger of calcium signaling. Therefore, the dual activity of Pic, as a mucus secretagogue and a mucinase, is relevant in the context of carbon source generation and mucus layer penetration, allowing EAEC to live within the layer of mucus but also access epithelial cells.
Collapse
Affiliation(s)
- Fernando Flores-Sanchez
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), México DF, México
| | - Lucia Chavez-Dueñas
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), México DF, México
| | - Javier Sanchez-Villamil
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), México DF, México
| | - Fernando Navarro-Garcia
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), México DF, México
| |
Collapse
|