1
|
Kaden T, Alonso-Román R, Stallhofer J, Gresnigt MS, Hube B, Mosig AS. Leveraging Organ-on-Chip Models to Investigate Host-Microbiota Dynamics and Targeted Therapies for Inflammatory Bowel Disease. Adv Healthc Mater 2024:e2402756. [PMID: 39491534 DOI: 10.1002/adhm.202402756] [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: 07/25/2024] [Revised: 09/29/2024] [Indexed: 11/05/2024]
Abstract
Inflammatory bowel disease (IBD) is an idiopathic gastrointestinal disease with drastically increasing incidence rates. Due to its multifactorial etiology, a precise investigation of the pathogenesis is extremely difficult. Although reductionist cell culture models and more complex disease models in animals have clarified the understanding of individual disease mechanisms and contributing factors of IBD in the past, it remains challenging to bridge research and clinical practice. Conventional 2D cell culture models cannot replicate complex host-microbiota interactions and stable long-term microbial culture. Further, extrapolating data from animal models to patients remains challenging due to genetic and environmental diversity leading to differences in immune responses. Human intestine organ-on-chip (OoC) models have emerged as an alternative in vitro model approach to investigate IBD. OoC models not only recapitulate the human intestinal microenvironment more accurately than 2D cultures yet may also be advantageous for the identification of important disease-driving factors and pharmacological interventions targets due to the possibility of emulating different complexities. The predispositions and biological hallmarks of IBD focusing on host-microbiota interactions at the intestinal mucosal barrier are elucidated here. Additionally, the potential of OoCs to explore microbiota-related therapies and personalized medicine for IBD treatment is discussed.
Collapse
Affiliation(s)
- Tim Kaden
- Dynamic42 GmbH, 07745, Jena, Germany
- Institute of Biochemistry II, Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany
| | - Raquel Alonso-Román
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, 07745, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, 07745, Jena, Germany
| | - Johannes Stallhofer
- Department of Internal Medicine IV, Jena University Hospital, 07747, Jena, Germany
| | - Mark S Gresnigt
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, 07745, Jena, Germany
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, 07745, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, 07745, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, 07745, Jena, Germany
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University, 07743, Jena, Germany
| | - Alexander S Mosig
- Institute of Biochemistry II, Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, 07745, Jena, Germany
| |
Collapse
|
2
|
Wang Z, Ma Z, Tian Z, Jia H, Zhang L, Mao Y, Yang Z, Liu X, Li M. Microbial dysbiosis in the gut–mammary axis as a mechanism for mastitis in dairy cows. INT J DAIRY TECHNOL 2024. [DOI: 10.1111/1471-0307.13150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Mastitis is a significant and costly disease in dairy cows, reducing milk production and affecting herd health. Recent research highlights the role of gastrointestinal microbial dysbiosis in the development of mastitis. This review focuses on how microbial imbalances in the rumen and intestines can compromise the integrity of the gastrointestinal barriers, allowing harmful bacteria and endotoxins, such as lipopolysaccharide, to enter the bloodstream and reach the mammary gland, triggering inflammation. This process links gastrointestinal health to mammary gland inflammation through the gut–mammary axis. Furthermore, disruptions in glucose metabolism and immune responses are implicated in the progression of mastitis. This review underscores the potential for non‐antibiotic interventions aimed at restoring microbial balance to reduce mastitis incidence, providing new insights into improving dairy cow health and farm productivity. Our findings emphasise the critical need to explore preventive measures targeting the rumen and intestinal microbiota for effective mastitis control.
Collapse
Affiliation(s)
- Zhiwei Wang
- College of Animal Science and Technology Yangzhou University Yangzhou Jiangsu 225009 China
| | - Zheng Ma
- College of Animal Science and Technology Yangzhou University Yangzhou Jiangsu 225009 China
| | - Zhichen Tian
- College of Animal Science and Technology Yangzhou University Yangzhou Jiangsu 225009 China
| | - Haoran Jia
- College of Animal Science and Technology Yangzhou University Yangzhou Jiangsu 225009 China
| | - Lei Zhang
- College of Animal Science and Technology Yangzhou University Yangzhou Jiangsu 225009 China
| | - Yongjiang Mao
- College of Animal Science and Technology Yangzhou University Yangzhou Jiangsu 225009 China
- Joint International Research Laboratory of Agriculture and Agri‐Product Safety the Ministry of Education, Yangzhou University Yangzhou Jiangsu 225009 China
| | - Zhangping Yang
- College of Animal Science and Technology Yangzhou University Yangzhou Jiangsu 225009 China
- Joint International Research Laboratory of Agriculture and Agri‐Product Safety the Ministry of Education, Yangzhou University Yangzhou Jiangsu 225009 China
| | - Xu Liu
- College of Veterinary Medicine Northwest A&F University Yangling Shanxi 712100 China
| | - Mingxun Li
- College of Animal Science and Technology Yangzhou University Yangzhou Jiangsu 225009 China
- Joint International Research Laboratory of Agriculture and Agri‐Product Safety the Ministry of Education, Yangzhou University Yangzhou Jiangsu 225009 China
| |
Collapse
|
3
|
Jin J, Li Q, Zhou Q, Li X, Lan F, Wen C, Wu G, Li G, Yan Y, Yang N, Sun C. Calcium deposition in chicken eggshells: role of host genetics and gut microbiota. Poult Sci 2024; 103:104073. [PMID: 39068697 PMCID: PMC11339253 DOI: 10.1016/j.psj.2024.104073] [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: 05/07/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/30/2024] Open
Abstract
Eggshell is predominantly composed of calcium carbonate, making up about 95% of its composition. Eggshell quality is closely related to the amount of calcium deposition in the shell, which requires chickens to maintain a robust state of calcium metabolism. In this study, we introduced a novel parameter, Total Eggshell Weight (TESW), which measures the total weight of eggshells produced by chickens over a period of 10 consecutive d, providing valuable information on the intensity of calcium metabolism in chickens. Genome-wide association study (GWAS) was conducted to explore the genetic determinants of eggshell calcification in a population of 570 Rhode Island Red laying hens at 90 wk of age. This study revealed a significant association between a specific SNP (rs14249431) and TESW. Additionally, using random forest modeling and 2-tailed testing, we identified 3 genera, Lactobacillus in the jejunum, Lactobacillus, and Fournierella in the cecum, that exhibited a significant association with TESW. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis of claudin-1 and occludin genes in individuals with low TESW and high abundance of jejunal Lactobacillus confirmed that the inhibitory effect of jejunal Lactobacillus on calcium uptake was achieved through the up-regulation of tight junctions in intestinal epithelial cells. Notably, both host and microbial factors influence TESW, displaying a mutually influential relationship between them. The microbiome-wide Genome-Wide Association Study (mb-GWAS) identified significant associations between these 3 genera and specific genomic variants, such as rs316115020 and rs316420452 on chromosome 5, rs313198529 on chromosome 11, linked to Lactobacillus in the cecum. Moreover, rs312552529 on chromosome 1 exhibited potential association with Fournierella in the cecum. This study highlights the influence of host genetics and gut microbiota on calcium deposition in eggshells during the late laying phase, providing a foundational reference for studying calcium metabolism in hens.
Collapse
Affiliation(s)
- Jiaming Jin
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Quanlin Li
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Qianqian Zhou
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xiaochang Li
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Fangren Lan
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Chaoliang Wen
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Guiqin Wu
- Beijing Engineering Research Centre of Layer, Beijing 101206, China
| | - Guangqi Li
- Beijing Engineering Research Centre of Layer, Beijing 101206, China
| | - Yiyuan Yan
- Beijing Engineering Research Centre of Layer, Beijing 101206, China
| | - Ning Yang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Congjiao Sun
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
4
|
Pan H, Hu T, He Y, Zhong G, Wu S, Jiang X, Rao G, You Y, Ruan Z, Tang Z, Hu L. Curcumin attenuates aflatoxin B1-induced ileum injury in ducks by inhibiting NLRP3 inflammasome and regulating TLR4/NF-κB signaling pathway. Mycotoxin Res 2024; 40:255-268. [PMID: 38400893 DOI: 10.1007/s12550-024-00524-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 01/02/2024] [Accepted: 01/29/2024] [Indexed: 02/26/2024]
Abstract
Aflatoxin B1 (AFB1) is a widespread toxic contamination in feed for animals. The primary active component of turmeric, curcumin (Cur), is an antioxidant and an anti-inflammatory. However, it is yet unknown how AFB1 affects the intestinal epithelial barrier and whether Cur acts as a protective mechanism when exposed to AFB1. Here, we explored the mechanism of AFB1-induced intestinal injury from intestinal epithelial barrier, inflammation, pyroptosis, and intestinal flora, and evaluated the protective role of Cur. We found that AFB1 caused weight loss and intestinal morphological damage that is mainly characterized by shortened intestinal villi, deepened crypts, and damaged intestinal epithelium. Exposure to AFB1 decreased the expression of Claudin-1, MUC2, ZO-1, and Occludin and increased the expression of pyroptosis-related factors (NLRP3, GSDMD, Caspase-1, IL-1β, and IL-18) and inflammation-related factors (TLR4, NF-κB, IκB, IFN-γ, and TNF-α). Furthermore, ileal gut microbiota was altered, and simultaneously, the Lactobacillus abundance was decreased. The gut microbiota interacts with a wide range of physiologic functions and disease development in the host through its metabolites, and disturbances in gut microbial metabolism can cause functional impairment of the ileum. Meanwhile, Cur can ameliorate histological ileum injuries and intestinal flora disturbance caused by AFB1. We found that Cur reversed the effects of AFB1 through modulating both NLRP3 inflammasome and the TLR4/NF-κB signaling pathway. In conclusion, AFB1 can induce inflammatory damage and pyroptosis in duck ileum, while Cur has obviously protective effects on all the above damages.
Collapse
Affiliation(s)
- Hang Pan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- College of Life Science, Yantai University, Yantai City, 264005, Shandong Province, China
| | - Ting Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Ying He
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001, China
- Guangxi Key Laboratory of Veterinary Biotechnology, Nanning, Guangxi, China
- Key Laboratory of China(Guangxi)-ASEAN Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
| | - Gaolong Zhong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Shaofeng Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Xuanxuan Jiang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Gan Rao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yanli You
- College of Life Science, Yantai University, Yantai City, 264005, Shandong Province, China
| | - Zhiyan Ruan
- School of Pharmacy, Guangdong Food & Drug Vocational College, No. 321, Longdong North Road, Tianhe District, Guangzhou, 510520, Guangdong Province, People's Republic of China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Lianmei Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
5
|
Wang X, Xie X, Li Y, Xie X, Huang S, Pan S, Zou Y, Pan Z, Wang Q, Chen J, Zhou L, Luo X. Quercetin ameliorates ulcerative colitis by activating aryl hydrocarbon receptor to improve intestinal barrier integrity. Phytother Res 2024; 38:253-264. [PMID: 37873559 DOI: 10.1002/ptr.8027] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 09/05/2023] [Accepted: 09/19/2023] [Indexed: 10/25/2023]
Abstract
Ulcerative colitis (UC) pathogenesis is largely associated with intestinal epithelial barrier dysfunction. A therapeutic approach to UC involves the repair of damaged intestinal barrier. Our study aimed to investigate whether aryl hydrocarbon receptor (AhR) mediated the intestinal barrier repair effects of quercetin to ameliorate UC. 3% dextran sulfate sodium was used to induce colitic mice, and quercetin (25, 50, and 100 mg/kg) was administered orally for 10 days to assess the therapeutic effects. In vitro, Caco-2 cells were used to explore the effect of quercetin on tight junction protein expression and AhR activation. The results showed that quercetin alleviated colitic mice by restoring tight junctions (TJs) integrity via an AhR-dependent manner (p < 0.05). In vitro, quercetin dose-dependently elevated the expressions of TJs protein ZO-1 and Claudin1, and activated AhR by enhancing the expression of CYP1A1 and facilitating AhR nuclear translocation in Caco-2 cells (p < 0.05). While AhR antagonist CH223191 reversed the therapeutic effects of quercetin (p < 0.05) and blocked quercetin-induced AhR activation and enhancement of TJs protein (p < 0.05). In conclusion, quercetin repaired intestinal barrier dysfunction by activating AhR-mediated enhancement of TJs to alleviate UC. Our research offered new perspectives on how quercetin enhanced intestinal barrier function.
Collapse
Affiliation(s)
- Xiaojing Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuting Xie
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanyang Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xueqian Xie
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shaowei Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Simin Pan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanling Zou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zengfeng Pan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qing Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinyan Chen
- Basic Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lian Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xia Luo
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
6
|
Ahmad R, Kumar B, Thapa I, Talmon GA, Salomon J, Ramer-Tait AE, Bastola DK, Dhawan P, Singh AB. Loss of claudin-3 expression increases colitis risk by promoting Gut Dysbiosis. Gut Microbes 2023; 15:2282789. [PMID: 38010872 PMCID: PMC10730149 DOI: 10.1080/19490976.2023.2282789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 11/08/2023] [Indexed: 11/29/2023] Open
Abstract
Dysregulation of both the gut barrier and microbiota (dysbiosis) promotes susceptibility to and severity of Inflammatory Bowel Diseases (IBD). Leaky gut and dysbiosis often coexist; however, potential interdependence and molecular regulation are not well understood. Robust expression of claudin-3 (CLDN3) characterizes the gut epithelium, and studies have demonstrated a positive association between CLDN3 expression and gut barrier maturity and integrity, including in response to probiotics. However, the exact status and causal role of CLDN3 in IBD and regulation of gut dysbiosis remain unknown. Analysis of mouse and human IBD cohorts helped examine CLDN3 expression in IBD. The causal role was determined by modeling CLDN3 loss of expression during experimental colitis. 16S sequencing and in silico analysis helped examine gut microbiota diversity between Cldn3KO and WT mice and potential host metabolic responses. Fecal microbiota transplant (FMT) studies were performed to assess the role of gut dysbiosis in the increased susceptibility of Cldn3KO mice to colitis. A significant decrease in CLDN3 expression characterized IBD and CLDN3 loss of expression promoted colitis. 16S sequencing analysis suggested gut microbiota changes in Cldn3KO mice that were capable of modulating fatty acid metabolism and oxidative stress response. FMT from naïve Cldn3KO mice promoted colitis susceptibility in recipient germ-free mice (GFM) compared with GFM-receiving microbiota from WT mice. Our data demonstrate a critical role of CLDN3 in maintaining normal gut microbiota and inflammatory responses, which can be harnessed to develop novel therapeutic opportunities for patients with IBD.
Collapse
Affiliation(s)
- Rizwan Ahmad
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Balawant Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ishwor Thapa
- School of Interdisciplinary Informatics, College of Information Science & Technology, University of Nebraska at Omaha, Omaha, NE, USA
| | - Geoffrey A. Talmon
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jeffrey Salomon
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Amanda E. Ramer-Tait
- Department of Food Science and Technology and the Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Dhundy K. Bastola
- School of Interdisciplinary Informatics, College of Information Science & Technology, University of Nebraska at Omaha, Omaha, NE, USA
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Amar B. Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| |
Collapse
|
7
|
Vázquez-Gómez G, Petráš J, Dvořák Z, Vondráček J. Aryl hydrocarbon receptor (AhR) and pregnane X receptor (PXR) play both distinct and common roles in the regulation of colon homeostasis and intestinal carcinogenesis. Biochem Pharmacol 2023; 216:115797. [PMID: 37696457 DOI: 10.1016/j.bcp.2023.115797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/13/2023]
Abstract
Both aryl hydrocarbon receptor (AhR) and pregnane X receptor (PXR) belong among key regulators of xenobiotic metabolism in the intestinal tissue. AhR in particular is activated by a wide range of environmental and dietary carcinogens. The data accumulated over the last two decades suggest that both of these transcriptional regulators play a much wider role in the maintenance of gut homeostasis, and that both transcription factors may affect processes linked with intestinal tumorigenesis. Intestinal epithelium is continuously exposed to a wide range of AhR, PXR and dual AhR/PXR ligands formed by intestinal microbiota or originating from diet. Current evidence suggests that specific ligands of both AhR and PXR can protect intestinal epithelium against inflammation and assist in the maintenance of epithelial barrier integrity. AhR, and to a lesser extent also PXR, have been shown to play a protective role against inflammation-induced colon cancer, or, in mouse models employing overactivation of Wnt/β-catenin signaling. In contrast, other evidence suggests that both receptors may contribute to modulation of transformed colon cell behavior, with a potential to promote cancer progression and/or chemoresistance. The review focuses on both overlapping and separate roles of the two receptors in these processes, and on possible implications of their activity within the context of intestinal tissue.
Collapse
Affiliation(s)
- Gerardo Vázquez-Gómez
- Department of Cytokinetics, Institute of Biophysics of the CAS, Královopolská 135, 61265 Brno, Czech Republic
| | - Jiří Petráš
- Department of Cytokinetics, Institute of Biophysics of the CAS, Královopolská 135, 61265 Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
| | - Zdeněk Dvořák
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, 783 71 Olomouc, Czech Republic
| | - Jan Vondráček
- Department of Cytokinetics, Institute of Biophysics of the CAS, Královopolská 135, 61265 Brno, Czech Republic.
| |
Collapse
|
8
|
Quansah E, Gardey E, Ramoji A, Meyer-Zedler T, Goehrig B, Heutelbeck A, Hoeppener S, Schmitt M, Waldner M, Stallmach A, Popp J. Intestinal epithelial barrier integrity investigated by label-free techniques in ulcerative colitis patients. Sci Rep 2023; 13:2681. [PMID: 36792686 PMCID: PMC9931702 DOI: 10.1038/s41598-023-29649-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 02/08/2023] [Indexed: 02/17/2023] Open
Abstract
The intestinal epithelial barrier, among other compartments such as the mucosal immune system, contributes to the maintenance of intestinal homeostasis. Therefore, any disturbance within the epithelial layer could lead to intestinal permeability and promote mucosal inflammation. Considering that disintegration of the intestinal epithelial barrier is a key element in the etiology of ulcerative colitis, further assessment of barrier integrity could contribute to a better understanding of the role of epithelial barrier defects in ulcerative colitis (UC), one major form of chronic inflammatory bowel disease. Herein, we employ fast, non-destructive, and label-free non-linear methods, namely coherent anti-Stokes Raman scattering (CARS), second harmonic generation (SHG), two-photon excited fluorescence (TPEF), and two-photon fluorescence lifetime imaging (2P-FLIM), to assess the morpho-chemical contributions leading to the dysfunction of the epithelial barrier. For the first time, the formation of epithelial barrier gaps was directly visualized, without sophisticated data analysis procedures, by the 3D analysis of the colonic mucosa from severely inflamed UC patients. The results were compared with histopathological and immunofluorescence images and validated using transmission electron microscopy (TEM) to indicate structural alterations of the apical junction complex as the underlying cause for the formation of the epithelial barrier gaps. Our findings suggest the potential advantage of non-linear multimodal imaging is to give precise, detailed, and direct visualization of the epithelial barrier in the gastrointestinal tract, which can be combined with a fiber probe for future endomicroscopy measurements during real-time in vivo imaging.
Collapse
Affiliation(s)
- Elsie Quansah
- Institute of Physical Chemistry and Abbe Center of Photonics (IPC), Member of the Leibniz Centre for Photonics in Infection Research (LPI), Friedrich Schiller University Jena, Helmholtzweg 4, 07743, Jena, Germany
- Leibniz Institute of Photonic Technology (IPHT), Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745, Jena, Germany
| | - Elena Gardey
- Department of Internal Medicine IV (Gastroenterology, Hepatology, Infectious Diseases and Interdisciplinary Endoscopy), Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747, Jena, Germany
- Friedrich Schiller University Jena, Jena Center for Soft Matter (JCSM), Philosophenweg 7, 07743, Jena, Germany
| | - Anuradha Ramoji
- Institute of Physical Chemistry and Abbe Center of Photonics (IPC), Member of the Leibniz Centre for Photonics in Infection Research (LPI), Friedrich Schiller University Jena, Helmholtzweg 4, 07743, Jena, Germany.
- Leibniz Institute of Photonic Technology (IPHT), Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745, Jena, Germany.
- Jena University Hospital, Center for Sepsis Control and Care (CSCC), Friedrich Schiller University Jena, Erlanger Allee 101, 07747, Jena, Germany.
| | - Tobias Meyer-Zedler
- Institute of Physical Chemistry and Abbe Center of Photonics (IPC), Member of the Leibniz Centre for Photonics in Infection Research (LPI), Friedrich Schiller University Jena, Helmholtzweg 4, 07743, Jena, Germany
- Leibniz Institute of Photonic Technology (IPHT), Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745, Jena, Germany
| | - Bianca Goehrig
- Institute for Occupational, Social, and Environmental Medicine, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Astrid Heutelbeck
- Institute for Occupational, Social, and Environmental Medicine, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Stephanie Hoeppener
- Friedrich Schiller University Jena, Jena Center for Soft Matter (JCSM), Philosophenweg 7, 07743, Jena, Germany
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743, Jena, Germany
| | - Michael Schmitt
- Institute of Physical Chemistry and Abbe Center of Photonics (IPC), Member of the Leibniz Centre for Photonics in Infection Research (LPI), Friedrich Schiller University Jena, Helmholtzweg 4, 07743, Jena, Germany
- Leibniz Institute of Photonic Technology (IPHT), Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745, Jena, Germany
| | - Maximillian Waldner
- Department of Medicine, University of Erlangen-Nuremberg, 91054, Erlangen, Germany
| | - Andreas Stallmach
- Department of Internal Medicine IV (Gastroenterology, Hepatology, Infectious Diseases and Interdisciplinary Endoscopy), Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747, Jena, Germany
- Friedrich Schiller University Jena, Jena Center for Soft Matter (JCSM), Philosophenweg 7, 07743, Jena, Germany
| | - Jürgen Popp
- Institute of Physical Chemistry and Abbe Center of Photonics (IPC), Member of the Leibniz Centre for Photonics in Infection Research (LPI), Friedrich Schiller University Jena, Helmholtzweg 4, 07743, Jena, Germany
- Leibniz Institute of Photonic Technology (IPHT), Member of Leibniz Health Technologies, Member of the Leibniz Centre for Photonics in Infection Research (LPI), Albert-Einstein-Straße 9, 07745, Jena, Germany
| |
Collapse
|
9
|
Huang Y, He C, Hu Z, Chu X, Zhou S, Hu X, Deng J, Xiao D, Tao T, Yang H, Chen AF, Yin Y, Yang X. The beneficial effects of alpha-tocopherol on intestinal function and the expression of tight junction proteins in differentiated segments of the intestine in piglets. Food Sci Nutr 2023; 11:677-687. [PMID: 36789053 PMCID: PMC9922147 DOI: 10.1002/fsn3.3103] [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: 01/02/2022] [Revised: 09/12/2022] [Accepted: 09/28/2022] [Indexed: 11/10/2022] Open
Abstract
Alpha (α)-tocopherol is a major component of dietary vitamin E. Despite being one of the most widely used food supplements in both animals and humans, its role in intestinal functions remains unknown. We were able to examine and accurately demonstrate its permeability effect in vitro and its differentiated effect on tight junction expression in different segments of the intestine in vivo using cultured intestinal porcine epithelial cell line (IPEC-J2) and piglets. A cultured IPEC-J2 demonstrated that α-tocopherol upregulated the expression of tight junction proteins and improved their integrity, with a maximum effect at concentrations ranging from 20 to 40 μmol/L. In vivo data from weaned pigs fed different doses of α-tocopherol for 2 weeks revealed that α-tocopherol effectively increases the expression of tight junction proteins in all sections of the intestinal mucosa, with the highest effect on the duodenum at an optimum dose of 20-50 mg/kg. In contrast, α-tocopherol did not affect intestinal inflammation. These findings suggest that α-tocopherol maintains intestinal integrity and increases the expression of tight junction proteins both in vitro and in vivo.
Collapse
Affiliation(s)
- Yanjun Huang
- Key Laboratory for Study and Discovery of Small Targeted Molecules of Hunan ProvinceDepartment of Pharmacy, School of MedicineHunan Normal UniversityChangshaChina
| | - Caimei He
- Key Laboratory for Study and Discovery of Small Targeted Molecules of Hunan ProvinceDepartment of Pharmacy, School of MedicineHunan Normal UniversityChangshaChina
| | - Zheng Hu
- Key Laboratory for Study and Discovery of Small Targeted Molecules of Hunan ProvinceDepartment of Pharmacy, School of MedicineHunan Normal UniversityChangshaChina
| | - Xuetong Chu
- Key Laboratory for Study and Discovery of Small Targeted Molecules of Hunan ProvinceDepartment of Pharmacy, School of MedicineHunan Normal UniversityChangshaChina
| | - Sichun Zhou
- Key Laboratory for Study and Discovery of Small Targeted Molecules of Hunan ProvinceDepartment of Pharmacy, School of MedicineHunan Normal UniversityChangshaChina
| | - Xin Hu
- Key Laboratory for Study and Discovery of Small Targeted Molecules of Hunan ProvinceDepartment of Pharmacy, School of MedicineHunan Normal UniversityChangshaChina
| | - Jun Deng
- Key Laboratory for Study and Discovery of Small Targeted Molecules of Hunan ProvinceDepartment of Pharmacy, School of MedicineHunan Normal UniversityChangshaChina
| | - Di Xiao
- Key Laboratory for Study and Discovery of Small Targeted Molecules of Hunan ProvinceDepartment of Pharmacy, School of MedicineHunan Normal UniversityChangshaChina
| | - Ting Tao
- Key Laboratory for Study and Discovery of Small Targeted Molecules of Hunan ProvinceDepartment of Pharmacy, School of MedicineHunan Normal UniversityChangshaChina
| | - Huansheng Yang
- Research Center for Healthy Breeding of Livestock and PoultryHunan Engineering and Research Center of Animal and Poultry Science and Key Laboratory for Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical Agriculture, The Chinese Academy of SciencesChangshaChina
| | - Alex F. Chen
- Key Laboratory for Study and Discovery of Small Targeted Molecules of Hunan ProvinceDepartment of Pharmacy, School of MedicineHunan Normal UniversityChangshaChina
| | - Yulong Yin
- Research Center for Healthy Breeding of Livestock and PoultryHunan Engineering and Research Center of Animal and Poultry Science and Key Laboratory for Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical Agriculture, The Chinese Academy of SciencesChangshaChina
| | - Xiaoping Yang
- Key Laboratory for Study and Discovery of Small Targeted Molecules of Hunan ProvinceDepartment of Pharmacy, School of MedicineHunan Normal UniversityChangshaChina
- Research Center for Healthy Breeding of Livestock and PoultryHunan Engineering and Research Center of Animal and Poultry Science and Key Laboratory for Agro‐ecological Processes in Subtropical RegionInstitute of Subtropical Agriculture, The Chinese Academy of SciencesChangshaChina
| |
Collapse
|
10
|
The Effects and Cell Barrier Mechanism of Main Dietary Nutrients on Intestinal Barrier. Curr Opin Food Sci 2022. [DOI: 10.1016/j.cofs.2022.100942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
11
|
Zuo F, Wei H, Peng J, Li S, Zhou Y. Effects on the Cell Barrier Function of L-Met and DL-HMTBA Is Related to Metabolic Characteristics and m 6A Modification. Front Nutr 2022; 9:836069. [PMID: 35464013 PMCID: PMC9020446 DOI: 10.3389/fnut.2022.836069] [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: 12/15/2021] [Accepted: 03/07/2022] [Indexed: 11/15/2022] Open
Abstract
Methionine is a substrate for protein synthesis and participates in many other biological events via its metabolism. We have previously demonstrated significant differences in the metabolism of L-methionine (L-Met) and its precursor DL-2-hydroxy-4-methylthiobutyric acid (DL-HMTBA) in IPEC-J2 cells. When DL–HMTBA is added to the diet, intracellular methionine (Met) sources also contain the natural form of L-Met. Then, what is the effect on Met metabolism when these two Met sources exist simultaneously? Moreover, the effects of metabolic differences on cell function remain unclear. In this study, it was found that when the proportion of L-Met to DL–HMTBA was ≤ 40%:60%, Met transmethylation was promoted and when the proportion of L-Met to DL-HMTBA was ≤ 85%:15%, Met trans-sulfuration and regeneration were improved. In addition, DL-HMTBA improved the cell barrier function when the ratio of L-Met to DL-HMTBA was ≤ 40%:60%. This finding may be due to the decrease in the proportion of S-adenosylmethionine to S-adenosylhomocysteine and mRNA N6-methyladenosine (m6A) levels, which increase the mRNA stability and protein expression of tight junction zona occludens-1. To sum up, the effects of L-Met and DL–HMTBA on Met metabolism, especially transmethylation, suggest that DL–HMTBA has the potential to influence the intestinal barrier function of animals through epigenetic processes.
Collapse
Affiliation(s)
- Fangrui Zuo
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.,Wuhan Sun HY Biology Co., Ltd, Wuhan, China
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Shengqing Li
- State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan, China
| | - Yuanfei Zhou
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
12
|
Vitamin D Receptor Influences Intestinal Barriers in Health and Disease. Cells 2022; 11:cells11071129. [PMID: 35406694 PMCID: PMC8997406 DOI: 10.3390/cells11071129] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 12/12/2022] Open
Abstract
Vitamin D receptor (VDR) executes most of the biological functions of vitamin D. Beyond this, VDR is a transcriptional factor regulating the expression levels of many target genes, such as genes for tight junction proteins claudin-2, -5, -12, and -15. In this review, we discuss the progress of research on VDR that influences intestinal barriers in health and disease. We searched PubMed and Google Scholar using key words vitamin D, VDR, tight junctions, cancer, inflammation, and infection. We summarize the literature and progress reports on VDR regulation of tight junction distribution, cellular functions, and mechanisms (directly or indirectly). We review the impacts of VDR on barriers in various diseases, e.g., colon cancer, infection, inflammatory bowel disease, and chronic inflammatory lung diseases. We also discuss the limits of current studies and future directions. Deeper understanding of the mechanisms by which the VDR signaling regulates intestinal barrier functions allow us to develop efficient and effective therapeutic strategies based on levels of tight junction proteins and vitamin D/VDR statuses for human diseases.
Collapse
|
13
|
Bolan S, Seshadri B, Keely S, Kunhikrishnan A, Bruce J, Grainge I, Talley NJ, Naidu R. Bioavailability of arsenic, cadmium, lead and mercury as measured by intestinal permeability. Sci Rep 2021; 11:14675. [PMID: 34282255 PMCID: PMC8289861 DOI: 10.1038/s41598-021-94174-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 06/25/2021] [Indexed: 12/17/2022] Open
Abstract
In this study, the intestinal permeability of metal(loid)s (MLs) such as arsenic (As), cadmium (Cd), lead (Pb) and mercury (Hg) was examined, as influenced by gut microbes and chelating agents using an in vitro gastrointestinal/Caco-2 cell intestinal epithelium model. The results showed that in the presence of gut microbes or chelating agents, there was a significant decrease in the permeability of MLs (As-7.5%, Cd-6.3%, Pb-7.9% and Hg-8.2%) as measured by apparent permeability coefficient value (Papp), with differences in ML retention and complexation amongst the chelants and the gut microbes. The decrease in ML permeability varied amongst the MLs. Chelating agents reduce intestinal absorption of MLs by forming complexes thereby making them less permeable. In the case of gut bacteria, the decrease in the intestinal permeability of MLs may be associated to a direct protection of the intestinal barrier against the MLs or indirect intestinal ML sequestration by the gut bacteria through adsorption on bacterial surface. Thus, both gut microbes and chelating agents can be used to decrease the intestinal permeability of MLs, thereby mitigating their toxicity.
Collapse
Affiliation(s)
- Shiv Bolan
- Global Centre for Environmental Remediation, University of Newcastle, Callaghan, NSW, Australia
- Cooperative Research Centre for Contamination Assessment and Remediation of the Environment, University of Newcastle, Callaghan, NSW, Australia
| | - Balaji Seshadri
- Global Centre for Environmental Remediation, University of Newcastle, Callaghan, NSW, Australia
- Cooperative Research Centre for Contamination Assessment and Remediation of the Environment, University of Newcastle, Callaghan, NSW, Australia
| | - Simon Keely
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Anitha Kunhikrishnan
- Global Centre for Environmental Remediation, University of Newcastle, Callaghan, NSW, Australia
| | - Jessica Bruce
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Ian Grainge
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - Nicholas J Talley
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Ravi Naidu
- Global Centre for Environmental Remediation, University of Newcastle, Callaghan, NSW, Australia.
- Cooperative Research Centre for Contamination Assessment and Remediation of the Environment, University of Newcastle, Callaghan, NSW, Australia.
| |
Collapse
|
14
|
Petito V, Greco V, Laterza L, Graziani C, Fanali C, Lucchetti D, Barbaro MR, Bugli F, Pieroni L, Lopetuso LR, Sgambato A, Sanguinetti M, Scaldaferri F, Urbani A, Gasbarrini A. Impact of the Trophic Effects of the Secretome From a Multistrain Probiotic Preparation on the Intestinal Epithelia. Inflamm Bowel Dis 2021; 27:902-913. [PMID: 33300553 DOI: 10.1093/ibd/izaa298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Probiotics are defined as live, nonpathogenic bacteria that confer health benefits beyond their nutritional value. In particular, VSL#3 exhibits demonstrated efficacy in the management of diseases characterized by an increased intestinal permeability. Our study aimed to understand how VSL#3 promotes gut health by secreting bioactive factors and identify which human pathways are modulated by secretome derived from the VSL#3 formula. METHODS Two different lots of VSL#3 were used, and Caco-2 cell line was treated with conditioned media (CM) prepared using 1 g of the probiotic formula. We evaluated the effects of the probiotics on cellular proliferation and apoptosis by cytometry and the expression of tight junction proteins by western blotting. A proteomics analysis of both culture media and the whole proteome of Caco-2 cells treated with VSL#3-CM was performed by nano-ultra performance liquid chromatography - tandem mass (nUPLC MS/MS) spectrometry. RESULTS The probiotic formula increased cell proliferation, decreased cellular apoptosis cells, and increased re-epithelialization in the scratch assay. Several peptides specifically synthetized by all the species within the probiotic preparation were recognized in the proteomics analysis. Human proteins synthesized by CaCo-2 cells were also identified. CONCLUSIONS To our knowledge, this manuscript describes the first evaluation of the probiotic secretome, and the results showed that the improvement in intestinal barrier functions induced by probiotics seems to be accompanied by the modulation of some human cellular pathways.
Collapse
Affiliation(s)
- Valentina Petito
- Università Cattolica del Sacro Cuore, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Rome, Italy
| | - Viviana Greco
- Università Cattolica del Sacro Cuore, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Rome, Italy.,Università Cattolica del Sacro Cuore, Dipartimento Universitario di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Rome, Italy
| | - Lucrezia Laterza
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Dipartimento di Scienze di Laboratorio e Infettivologiche, Rome, Italy
| | - Cristina Graziani
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Dipartimento di Scienze di Laboratorio e Infettivologiche, Rome, Italy
| | - Caterina Fanali
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Dipartimento di Scienze di Laboratorio e Infettivologiche, Rome, Italy
| | - Donatella Lucchetti
- Università Cattolica del Sacro Cuore, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Rome, Italy
| | - Maria Raffaella Barbaro
- Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Dipartimento di Scienze Mediche e Chirurgiche, Rome, Italy
| | - Francesca Bugli
- Policlinico Sant'Orsola- Malpighi, Università di Bologna, Dipartimento di Scienze Mediche e Chirurgiche, Bologna, Italia
| | - Luisa Pieroni
- Fondazione Santa Lucia IRCCS, Unitá di Proteomica e Metabolomica, Rome, Italy
| | - Loris Riccardo Lopetuso
- Università Cattolica del Sacro Cuore, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Rome, Italy.,Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Dipartimento di Scienze di Laboratorio e Infettivologiche, Rome, Italy
| | - Alessandro Sgambato
- Università Cattolica del Sacro Cuore, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Rome, Italy
| | - Maurizio Sanguinetti
- Università Cattolica del Sacro Cuore, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Rome, Italy.,Università Cattolica del Sacro Cuore, Dipartimento Universitario di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Rome, Italy
| | - Franco Scaldaferri
- Università Cattolica del Sacro Cuore, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Rome, Italy.,Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Dipartimento di Scienze di Laboratorio e Infettivologiche, Rome, Italy
| | - Andrea Urbani
- Università Cattolica del Sacro Cuore, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Rome, Italy.,Università Cattolica del Sacro Cuore, Dipartimento Universitario di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Rome, Italy
| | - Antonio Gasbarrini
- Università Cattolica del Sacro Cuore, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Rome, Italy.,Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Dipartimento di Scienze di Laboratorio e Infettivologiche, Rome, Italy
| |
Collapse
|
15
|
Liu F, Liu J, Liu Y, Zhang Y, Ding X. Shen-Fu Decoction could ameliorate intestinal permeability by regulating the intestinal expression of tight junction proteins and p-VASP in septic rats. JOURNAL OF ETHNOPHARMACOLOGY 2021; 268:113562. [PMID: 33217519 DOI: 10.1016/j.jep.2020.113562] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 10/31/2020] [Accepted: 11/02/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shen-Fu Decoction (SFD), a classic Traditional Chinese paired herb formulation, has been widely used for the treatment of sepsis in China. This study was carried out to assess the effects of SFD in sepsis-induced intestinal permeability and intestinal epithelial tight junction damage in rats with sepsis. MATERIALS AND METHODS A rat model of sepsis was created by cecal ligation and puncture (CLP). Rats in Sham and CLP + vehicle groups received equal distilled water, while rats in SFD group were treated by gavage of SFD (3 mg/kg, twice a day) for 72h. Mortality, sepsis-induced peritoneal inflammation, intestinal and liver histopathology damage, intestinal permeability (serum FITC-dextran and D-lactate), serum LPS, serum inflammation (PCT, TNF-α, and IL-6), and liver function (AST and ALT) were evaluated. The levels of zonula occluden (ZO-1), Occludin, Claudin-1 were analyzed by Real-time quantitative PCR and Western blotting (WB) respectively. Vasodilator-stimulated phosphoprotein (VASP) and p-VASP in intestinal epithelium were analyzed by WB. RESULTS Our study showed that SFD markedly reduced the mortality rate of CLP rats, prevented intestine and liver damage, relieved sepsis-induced intestinal permeability and inflammation elevation, ameliorated sepsis-induced impaired intestinal permeability by regulating the expression of ZO-1, Occludin, Claudin-1 and p-VASP. CONCLUSIONS The herbal formula SFD may be useful for reducing sepsis-induced organic damage and mortality by ameliorating the condition of sepsis-induced intestinal permeability by regulating tight junction proteins and p-VASP.
Collapse
Affiliation(s)
- Fusheng Liu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, China
| | - Jin Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Yinuo Liu
- First Clinical College of Hubei University of Medicine, Shiyan City, Hubei province, 442000, China
| | - Yin Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xia Ding
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
16
|
Liang H, Cheng R, Wang J, Xie H, Li R, Shimizu K, Zhang C. Mogrol, an aglycone of mogrosides, attenuates ulcerative colitis by promoting AMPK activation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 81:153427. [PMID: 33296813 DOI: 10.1016/j.phymed.2020.153427] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 11/03/2020] [Accepted: 11/27/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Ulcerative colitis (UC) is a non-specific chronic inflammatory disease. The incidence of UC in China has been increasing in recent years. Mogrol is an aglycone of mogrosides. Studies have shown that mogrosides have anti-oxygenation, anti-inflammatory, and laxative effects as well as other biological activities. PURPOSE To investigate the beneficial effects of mogrol on UC and identify its underlying mechanisms. STUDY DESIGN We used the dextran sodium sulphate (DSS)-induced UC model in mice, TNF-α-damaged NCM460 colonic epithelial cells, macrophage cells THP-M stimulated with lipopolysaccharide (LPS) / adenosine triphosphate (ATP) and compound C (an AMPK inhibitor) to confirm the key role of AMPK (AMP-activated protein kinase) activation. METHODS Histological evaluation, immunohistochemical staining, Western blot analysis, immunofluorescence assay and quantitative real time-PCR were used in the study. RESULTS Oral administration of mogrol (5 mg/kg/daily) in vivo significantly attenuated pathological colonic damage, inhibited inflammatory infiltration and improved the abnormal expression of NLRP3 inflammasome in colonic mucosa via the AMPK and NF-κB signaling pathways. In vitro, mogrol protected against intestinal epithelial barrier dysfunction by activating AMPK in TNF-α-treated NCM460 cells and inhibited the production of inflammatory mediator in LPS-stimulated THP-M cells. Furthermore, mogrol's effects were reversed by compound C intervention in DSS-induced UC model. CONCLUSION Mogrol exerts protective effects in experimental UC and inhibits production of inflammatory mediators through activation of AMPK-mediated signaling pathways.
Collapse
Affiliation(s)
- Han Liang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China; Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Rui Cheng
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Jiaoyang Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China
| | - Haifeng Xie
- Research and Development Department, Chengdu Biopurify Phytochemicals Ltd., Chengdu, China
| | - Renshi Li
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China.
| | - Kuniyoshi Shimizu
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China; Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Chaofeng Zhang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China; Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
17
|
Ibrahim D, Sewid AH, Arisha AH, abd El-fattah AH, Abdelaziz AM, Al-Jabr OA, Kishawy ATY. Influence of Glycyrrhiza glabra Extract on Growth, Gene Expression of Gut Integrity, and Campylobacter jejuni Colonization in Broiler Chickens. Front Vet Sci 2020; 7:612063. [PMID: 33415133 PMCID: PMC7782238 DOI: 10.3389/fvets.2020.612063] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/20/2020] [Indexed: 12/20/2022] Open
Abstract
Phytogenic feed additives have been gaining considerable interest due to their ability to improve gut health and thereby performance of broiler chickens. The impact of Glycyrrhiza glabra (licorice) extract (GE) on expression of genes coding for tight junction proteins and gut protection and Campylobacter jejuni colonization in broilers has not been discussed until now. Thus, the current study assessed the effective dose of GE for maximum growth in broiler chickens, clear-cut molecular mechanisms related to integrity and health of intestine, and controlling C. jejuni colonization. Over a 35-day feeding period, a total of 500 Ross broiler chicks were allocated to five groups; the first group was fed a control diet without GE and the second group to the fifth group were fed a control diet with GE (0.25, 0.5, 1, and 2 g/kg of diet); each group comprised 100 chicks with 10 replicates (10 birds/replicate). Birds fed GE had an improved body weight gain and feed conversion ratio. Furthermore, the highest body weight gain was observed in the group that received 1 g/kg of GE (P < 0.05). The expression of genes coding for tight junction proteins [occludin and junctional adhesion molecules (JAM)] was upregulated in all groups supplemented with GE. Moreover, birds fed 1 g/kg of GE exhibited the maximum gene expression of occludin and JAM [0.2 and 0.3 fold change, respectively (P < 0.05)]. In relation to enterocyte protective genes [glucagon-like peptide (GLP-2) and fatty acid-binding protein (FABP-6)], use of GE significantly upregulated expression of GLP-2 gene with 0.8 fold change in 2 g/kg of the GE supplemented group (P < 0.05) while the expression of FABP-6 gene was not affected by GE supplementation (P > 0.05). After challenge with C. jejuni, the expression of mucin (MUC-2) gene was upregulated and the inflammatory markers such as Toll-like receptors (TLR-4) and interleukin (IL-1β) were downregulated with increasing level of supplemented GE (P < 0.05). The mean log10 count of C. jejuni in cecal samples after 7 days post-infection by culture and real-time qPCR was decreased in groups fed GE in a dose-dependent manner (P < 0.05). In addition, the highest reduction of C. jejuni count in cecal samples by culture and real-time qPCR was observed in the group fed 2 g/kg of GE (2.58 and 2.28 log10 CFU/g, respectively). Results from this study suggested that G. glabra extract (1 g/kg) improved growth performance of broiler chickens, as well as influenced the maintenance of intestinal integrity and reduced C. jejuni shedding from infected birds.
Collapse
Affiliation(s)
- Doaa Ibrahim
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Alaa H. Sewid
- Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
- Department of Biomedical and Diagnostic Sciences, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Ahmed H. Arisha
- Department of Physiology, Faculty of Veterinary Medicine, Zaagazig University, Zagazig, Egypt
- Department of Animal Physiology and Biochemistry, Faculty of Veterinary Medicine, Badr University in Cairo (BUC), Badr City, Egypt
| | - Amir H. abd El-fattah
- Department of Animal Wealth Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Adel M. Abdelaziz
- Faculty of Veterinary Medicine, Veterinary Educational Hospital, Zagazig University, Zagazig, Egypt
| | - Omar A. Al-Jabr
- Department of Microbiology, College of Veterinary Medicine, King Faisal University, Hofuf, Saudi Arabia
| | - Asmaa T. Y. Kishawy
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
18
|
Increased intestinal permeability and gut dysbiosis in the R6/2 mouse model of Huntington's disease. Sci Rep 2020; 10:18270. [PMID: 33106549 PMCID: PMC7589489 DOI: 10.1038/s41598-020-75229-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 10/12/2020] [Indexed: 12/18/2022] Open
Abstract
Huntington's disease (HD) is a progressive, multifaceted neurodegenerative disease associated with weight loss and gut problems. Under healthy conditions, tight junction (TJ) proteins maintain the intestinal barrier integrity preventing bacterial translocation from the intestinal lumen to the systemic circulation. Reduction of TJs expression in Parkinson's disease patients has been linked with increased intestinal permeability-leaky gut syndrome. The intestine contains microbiota, most dominant phyla being Bacteroidetes and Firmicutes; in pathogenic or disease conditions the balance between these bacteria might be disrupted. The present study investigated whether there is evidence for an increased intestinal permeability and dysbiosis in the R6/2 mouse model of HD. Our data demonstrate that decreased body weight and body length in R6/2 mice is accompanied by a significant decrease in colon length and increased gut permeability compared to wild type littermates, without any significant changes in the protein levels of the tight junction proteins (occludin, zonula occludens). Moreover, we found an altered gut microbiota in R6/2 mice with increased relative abundance of Bacteroidetes and decreased of Firmicutes. Our results indicate an increased intestinal permeability and dysbiosis in R6/2 mice and further studies investigating the clinical relevance of these findings are warranted.
Collapse
|
19
|
Roodsant T, Navis M, Aknouch I, Renes IB, van Elburg RM, Pajkrt D, Wolthers KC, Schultsz C, van der Ark KCH, Sridhar A, Muncan V. A Human 2D Primary Organoid-Derived Epithelial Monolayer Model to Study Host-Pathogen Interaction in the Small Intestine. Front Cell Infect Microbiol 2020; 10:272. [PMID: 32656095 PMCID: PMC7326037 DOI: 10.3389/fcimb.2020.00272] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
Gut organoids are stem cell derived 3D models of the intestinal epithelium that are useful for studying interactions between enteric pathogens and their host. While the organoid model has been used for both bacterial and viral infections, this is a closed system with the luminal side being inaccessible without microinjection or disruption of the organoid polarization. In order to overcome this and simplify their applicability for transepithelial studies, permeable membrane based monolayer approaches are needed. In this paper, we demonstrate a method for generating a monolayer model of the human fetal intestinal polarized epithelium that is fully characterized and validated. Proximal and distal small intestinal organoids were used to generate 2D monolayer cultures, which were characterized with respect to epithelial cell types, polarization, barrier function, and gene expression. In addition, viral replication and bacterial translocation after apical infection with enteric pathogens Enterovirus A71 and Listeria monocytogenes were evaluated, with subsequent monitoring of the pro-inflammatory host response. This human 2D fetal intestinal monolayer model will be a valuable tool to study host-pathogen interactions and potentially reduce the use of animals in research.
Collapse
Affiliation(s)
- Thomas Roodsant
- Department of Global Health-Amsterdam Institute for Global Health and Development, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands.,Department of Medical Microbiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Marit Navis
- Tytgat Institute for Intestinal and Liver Research, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Ikrame Aknouch
- Department of Medical Microbiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands.,Viroclinics Xplore, Schaijk, Netherlands
| | - Ingrid B Renes
- Danone Nutricia Research, Utrecht, Netherlands.,Department of Pediatrics, Amsterdam University Medical Center (UMC), Emma Children's Hospital, University of Amsterdam, Amsterdam, Netherlands
| | - Ruurd M van Elburg
- Department of Pediatrics, Amsterdam University Medical Center (UMC), Emma Children's Hospital, University of Amsterdam, Amsterdam, Netherlands
| | - Dasja Pajkrt
- Department of Pediatric Infectious Diseases, Amsterdam University Medical Center (UMC), Emma Children's Hospital, University of Amsterdam, Amsterdam, Netherlands
| | - Katja C Wolthers
- Department of Medical Microbiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Constance Schultsz
- Department of Global Health-Amsterdam Institute for Global Health and Development, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands.,Department of Medical Microbiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Kees C H van der Ark
- Department of Global Health-Amsterdam Institute for Global Health and Development, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands.,Department of Medical Microbiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Adithya Sridhar
- Department of Medical Microbiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Vanesa Muncan
- Tytgat Institute for Intestinal and Liver Research, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
20
|
Nattramilarasu PK, Bücker R, Lobo de Sá FD, Fromm A, Nagel O, Lee IFM, Butkevych E, Mousavi S, Genger C, Kløve S, Heimesaat MM, Bereswill S, Schweiger MR, Nielsen HL, Troeger H, Schulzke JD. Campylobacter concisus Impairs Sodium Absorption in Colonic Epithelium via ENaC Dysfunction and Claudin-8 Disruption. Int J Mol Sci 2020; 21:ijms21020373. [PMID: 31936044 PMCID: PMC7013563 DOI: 10.3390/ijms21020373] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/31/2019] [Accepted: 12/31/2019] [Indexed: 12/12/2022] Open
Abstract
The epithelial sodium channel (ENaC) can increase the colonic absorptive capacity for salt and water. Campylobacter concisus is a common pathogenic epsilonproteobacterium, causing enteritis and diarrhea. It can induce barrier dysfunction in the intestine, but its influence on intestinal transport function is still unknown. Therefore, our study aimed to characterize C. concisus effects on ENaC using the HT-29/B6-GR/MR (epithelial cell line HT-29/B6 transfected with glucocorticoid and mineralocorticoid receptors) cell model and mouse colon. In Ussing chambers, C. concisus infection inhibited ENaC-dependent Na+ transport as indicated by a reduction in amiloride-sensitive short circuit current (−55%, n = 15, p < 0.001). This occurred via down-regulation of β- and γ-ENaC mRNA expression and ENaC ubiquitination due to extracellular signal-regulated kinase (ERK)1/2 activation, predicted by Ingenuity Pathway Analysis (IPA). In parallel, C. concisus reduced the expression of the sealing tight junction (TJ) protein claudin-8 and induced claudin-8 redistribution off the TJ domain of the enterocytes, which facilitates the back leakage of Na+ ions into the intestinal lumen. In conclusion, C. concisus caused ENaC dysfunction via interleukin-32-regulated ERK1/2, as well as claudin-8-dependent barrier dysfunction—both of which contribute to Na+ malabsorption and diarrhea.
Collapse
Affiliation(s)
- Praveen Kumar Nattramilarasu
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Roland Bücker
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Fábia Daniela Lobo de Sá
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Anja Fromm
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Oliver Nagel
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - In-Fah Maria Lee
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Eduard Butkevych
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Soraya Mousavi
- Institute of Microbiology, Infectious Diseases and Immunology, Charité—Universitätsmedizin Berlin, Campus Benjamin Franklin, 14195 Berlin, Germany
| | - Claudia Genger
- Institute of Microbiology, Infectious Diseases and Immunology, Charité—Universitätsmedizin Berlin, Campus Benjamin Franklin, 14195 Berlin, Germany
| | - Sigri Kløve
- Institute of Microbiology, Infectious Diseases and Immunology, Charité—Universitätsmedizin Berlin, Campus Benjamin Franklin, 14195 Berlin, Germany
| | - Markus M. Heimesaat
- Institute of Microbiology, Infectious Diseases and Immunology, Charité—Universitätsmedizin Berlin, Campus Benjamin Franklin, 14195 Berlin, Germany
| | - Stefan Bereswill
- Institute of Microbiology, Infectious Diseases and Immunology, Charité—Universitätsmedizin Berlin, Campus Benjamin Franklin, 14195 Berlin, Germany
| | - Michal R. Schweiger
- Laboratory for Epigenetics and Tumour genetics, University Hospital Cologne and Centre for Molecular Medicine Cologne, 50931 Cologne, Germany
| | - Hans Linde Nielsen
- Department of Clinical Microbiology, Aalborg University Hospital, 9000 Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, 9000 Aalborg, Denmark
| | - Hanno Troeger
- Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Jörg-Dieter Schulzke
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
- Correspondence:
| |
Collapse
|
21
|
Bachinger D, Mayer E, Kaschubek T, Schieder C, König J, Teichmann K. Influence of phytogenics on recovery of the barrier function of intestinal porcine epithelial cells after a calcium switch. J Anim Physiol Anim Nutr (Berl) 2018; 103:210-220. [PMID: 30353576 PMCID: PMC7379982 DOI: 10.1111/jpn.12997] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 08/06/2018] [Accepted: 08/26/2018] [Indexed: 12/13/2022]
Abstract
Background The gut barrier is essential for animal health as it prevents the passage of potentially harmful foreign substances. The epithelial tight junctions support the intestinal barrier and can be disrupted by stress caused, for example, by pathogens or dietary or environmental factors, predisposing the host to disease. In animal husbandry, phytogenics (plant‐derived feed additives) are used to support and maintain growth, feed efficiency and health. Therefore, several phytogenics were tested in vitro for their influence on the barrier function recovery of intestinal porcine epithelial cells (IPEC‐J2) after disruption, particularly on the abundance of tight junction proteins. Results IPEC‐J2 treated with 1,000 µg/ml liquorice root extract, 80 µg/ml plant powder mix, or 80 µg/ml angelica root powder showed significantly higher trans‐epithelial electric resistance (TEER) 24 hr after tight junction disruption via a calcium switch assay than the control. In contrast, cells treated with 1,000 µg/ml oak bark extract showed a significantly lower TEER after 6 hr but not at later time points. The increased TEER caused by the liquorice root extract correlated with an increase in the abundance of the tight junction protein claudin‐4. Conclusions This study suggests potential beneficial effects of liquorice and angelica root extracts on the gut barrier function when used as feed additives for livestock. Further studies, especially in vivo, are necessary to confirm these findings.
Collapse
Affiliation(s)
| | | | - Theresa Kaschubek
- BIOMIN Research Center, Tulln an der Donau, Austria.,Department of Nutritional Science, University of Vienna, Vienna, Austria
| | | | - Jürgen König
- Department of Nutritional Science, University of Vienna, Vienna, Austria
| | | |
Collapse
|
22
|
Li Y, Wu J, Niu Y, Chen H, Tang Q, Zhong Y, Lambers TT, Cai W. Milk Fat Globule Membrane Inhibits NLRP3 Inflammasome Activation and Enhances Intestinal Barrier Function in a Rat Model of Short Bowel. JPEN J Parenter Enteral Nutr 2018; 43:677-685. [PMID: 30144105 DOI: 10.1002/jpen.1435] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/18/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND The milk fat globule membrane (MFGM) contains various bioactive components which have been shown to maintain gut barrier integrity. This study aimed to evaluate the protective effects of MFGM on intestinal barrier function and its possible mechanisms in a rat model of short bowel syndrome (SBS). MATERIALS AND METHODS Five-week-old male Sprague-Dawley rats were divided into 3 groups (n = 8 per group), consisting of Sham group and rats submitted to massive small-bowel resection then supplemented with either water (SBS) or 1.5g/kg/d MFGM (SBS+MFGM) by daily gavage. Rats were sacrificed on day 15 postoperation. Intestinal adaptation, gut permeability, bacterial translocation (BT), expression of tight junction proteins, mucin 1 (MUC1), and nucleotide-binding oligomerization domain leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) pathway in the ileum were evaluated. RESULTS Both SBS+MFGM and SBS groups exhibited lower body weight and higher ileum villus height than Sham group, but no difference was detected between each other. SBS group had significantly higher intestinal permeability and BT rate than other groups (P < .05). Compared with SBS rats, SBS+MFGM group showed higher expression of tight junction proteins and MUC1, lower expression of NLRP3 and caspase-1 in the ileum, as well as lower interleukin (IL)-1β but higher IL-18 levels in ileum tissue. CONCLUSIONS Supplementation of MFGM helps to modulate NLRP3 inflammasome activation and enhances gut barrier integrity in rats after massive small-bowel resection, which provides experimental support for potential applications of MGFM in intestinal barrier dysfunction, although further studies are needed.
Collapse
Affiliation(s)
- Ying Li
- Department of Clinical Nutrition, Xin Hua Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiang Wu
- Department of Clinical Nutrition, Xin Hua Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Yang Niu
- Department of Clinical Nutrition, Xin Hua Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Honghao Chen
- Department of Clinical Nutrition, Xin Hua Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingya Tang
- Department of Clinical Nutrition, Xin Hua Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Yan Zhong
- Mead Johnson Pediatric Nutrition Institute, Nijmegen, the Netherlands
| | - Tim T Lambers
- Mead Johnson Pediatric Nutrition Institute, Nijmegen, the Netherlands
| | - Wei Cai
- Department of Clinical Nutrition, Xin Hua Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.,Department of Pediatric Surgery, Xin Hua Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
23
|
Zhou K, Cheng R, Liu B, Wang L, Xie H, Zhang C. Eupatilin ameliorates dextran sulphate sodium-induced colitis in mice partly through promoting AMPK activation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 46:46-56. [PMID: 30097122 DOI: 10.1016/j.phymed.2018.04.033] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 02/19/2018] [Accepted: 04/15/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Despite the higher morbidity of ulcerative colitis (UC), available treatments remain unsatisfactory in recent years. A natural flavone eupatilin (Eup) is known to inhibit the intestinal contraction. PURPOSE The protective role of Eup in intestinal inflammation remains unclear. This study attempted to determine the bioactivity of Eup against colitis and clarify the mechanism of action. STUDY DESIGN The in vitro effects of Eup on lipopolysaccharide-induced human THP-M macrophage activation and tumour necrosis factor-α (TNF-α)-damaged intestinal epithelial (NCM460) cells were explored to clarify its potential protective effects. Then, the alleviative efficacy of Eup was established in dextran sodium sulphate (DSS)-induced mice colitis. METHODS Pathological diagnosis, immunohistochemical staining, and reverse transcriptase PCR analysis as well as western blot analysis were employed in the current study. RESULTS Eup clearly inhibited inflammatory responses in LPS-stimulated macrophages. Eup also clearly stabilized colonic epithelia by down-regulating overexpression of tight junction proteins and NADPH oxidases 4 (NOX4), and by promoting AMP-activated protein kinase (AMPK) activation in TNF-α-stimulated NCM460 cells. In addition, in vivo study demonstrated that Eup treatment clearly ameliorated the symptoms and pathologic changes of colitis mice. The therapeutic effect of Eup was found to be reduced when compound C (an AMPK pharmacological inhibitor) was given to mice. CONCLUSION The study successfully demonstrated that Eup ameliorated DSS-induced mice colitis by suppressing inflammation and maintaining the integrity of the intestinal epithelial barrier via AMPK activation. The results provide valuable guidance for using Eup in UC treatment.
Collapse
Affiliation(s)
- Kai Zhou
- State Key Laboratory of Natural Medicines, Department of Traditional Chinese Medicines, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Rui Cheng
- State Key Laboratory of Natural Medicines, Department of Traditional Chinese Medicines, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Bei Liu
- State Key Laboratory of Natural Medicines, Department of Traditional Chinese Medicines, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Lei Wang
- State Key Laboratory of Natural Medicines, Department of Traditional Chinese Medicines, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China
| | - Haifeng Xie
- Chengdu Biopurity Phytochemicals Ltd., Chengdu, 611131, PR China
| | - Chaofeng Zhang
- State Key Laboratory of Natural Medicines, Department of Traditional Chinese Medicines, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, PR China.
| |
Collapse
|
24
|
Casselbrant A, Söfteland JM, Hellström M, Malinauskas M, Oltean M. Luminal Polyethylene Glycol Alleviates Intestinal Preservation Injury Irrespective of Molecular Size. J Pharmacol Exp Ther 2018; 366:29-36. [DOI: 10.1124/jpet.117.247023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
25
|
Stefano GB, Pilonis N, Ptacek R, Raboch J, Vnukova M, Kream RM. Gut, Microbiome, and Brain Regulatory Axis: Relevance to Neurodegenerative and Psychiatric Disorders. Cell Mol Neurobiol 2018; 38:1197-1206. [PMID: 29802603 PMCID: PMC6061125 DOI: 10.1007/s10571-018-0589-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 05/07/2018] [Indexed: 12/23/2022]
Abstract
It has become apparent that the molecular and biochemical integrity of interactive families, genera, and species of human gut microflora is critically linked to maintaining complex metabolic and behavioral processes mediated by peripheral organ systems and central nervous system neuronal groupings. Relatively recent studies have established intrinsic ratios of enterotypes contained within the human microbiome across demographic subpopulations and have empirically linked significant alterations in the expression of bacterial enterotypes with the initiation and persistence of several major metabolic and psychiatric disorders. Accordingly, the goal of our review is to highlight potential thematic/functional linkages of pathophysiological alterations in gut microbiota and bidirectional gut-brain signaling pathways with special emphasis on the potential roles of gut dysbiosis on the pathophysiology of psychiatric illnesses. We provide critical discussion of putative thematic linkages of Parkinson's disease (PD) data sets to similar pathophysiological events as potential causative factors in the development and persistence of diverse psychiatric illnesses. Finally, we include a concise review of preclinical paradigms that involve immunologically-induced GI deficits and dysbiosis of maternal microflora that are functionally linked to impaired neurodevelopmental processes leading to affective behavioral syndromes in the offspring.
Collapse
Affiliation(s)
- G B Stefano
- Department of Psychiatry, First Faculty of Medicine Charles University in Prague and General University Hospital in Prague, Center for Cognitive and Molecular Neuroscience, Ke Karlovu 11, 120 00, Prague 2, Czech Republic.
| | - N Pilonis
- Warsaw Medical University, Public Central Teaching Hospital, Warsaw, Poland
| | - R Ptacek
- Department of Psychiatry, First Faculty of Medicine Charles University in Prague and General University Hospital in Prague, Center for Cognitive and Molecular Neuroscience, Ke Karlovu 11, 120 00, Prague 2, Czech Republic
| | - J Raboch
- Department of Psychiatry, First Faculty of Medicine Charles University in Prague and General University Hospital in Prague, Center for Cognitive and Molecular Neuroscience, Ke Karlovu 11, 120 00, Prague 2, Czech Republic
| | - M Vnukova
- Department of Psychiatry, First Faculty of Medicine Charles University in Prague and General University Hospital in Prague, Center for Cognitive and Molecular Neuroscience, Ke Karlovu 11, 120 00, Prague 2, Czech Republic
| | - R M Kream
- Department of Psychiatry, First Faculty of Medicine Charles University in Prague and General University Hospital in Prague, Center for Cognitive and Molecular Neuroscience, Ke Karlovu 11, 120 00, Prague 2, Czech Republic
| |
Collapse
|
26
|
Abstract
With the coming of the "silver tsunami," expanding the knowledge about how various intrinsic and extrinsic factors affect the immune system in the elderly is timely and of immediate clinical need. The global population is increasing in age. By the year 2030, more than 20% of the population of the United States will be older than 65 years of age. This article focuses on how advanced age alters the immune systems and how this, in turn, modulates the ability of the aging lung to deal with infectious challenges from the outside world and from within the host.
Collapse
Affiliation(s)
- Elizabeth J Kovacs
- Division of GI, Trauma and Endocrine Surgery, Department of Surgery, Mucosal Inflammation Program, GILIIP (GI, Liver and Innate Immunity Program), Graduate Program in Immunology, IMAGE (Investigations in Metabolism, Aging, Gender and Exercise), University of Colorado Denver, Anschutz Medical Campus, 12700 East 19th Avenue, Research Complex 2, Mailstop #8620, Aurora, CO 80045, USA.
| | - Devin M Boe
- Division of GI, Trauma and Endocrine Surgery, Department of Surgery, Mucosal Inflammation Program, Graduate Program in Immunology, University of Colorado Denver, Anschutz Medical Campus, 12700 East 19th Avenue, Research Complex 2, Room 6460, Aurora, CO 80045, USA
| | - Lisbeth A Boule
- Division of GI, Trauma and Endocrine Surgery, Department of Surgery, Mucosal Inflammation Program, IMAGE, University of Colorado Denver, Anschutz Medical Campus, 12700 East 19th Avenue, Research Complex 2, Room 6460, Aurora, CO 80045, USA
| | - Brenda J Curtis
- Division of GI, Trauma and Endocrine Surgery, Department of Surgery, Mucosal Inflammation Program, IMAGE, University of Colorado Denver, Anschutz Medical Campus, 12700 East 19th Avenue, Research Complex 2, Room 6018, Aurora, CO 80045, USA
| |
Collapse
|
27
|
Zou Y, Ma L, Zhao Y, Zhang S, Zhou C, Cai Y. Inhibition of Rho kinase protects against colitis in mice by attenuating intestinal epithelial barrier dysfunction via MLC and the NF-κB pathway. Int J Mol Med 2017; 41:430-438. [PMID: 29115372 DOI: 10.3892/ijmm.2017.3197] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 10/09/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the role of Rho kinase (also known as ROCK) inhibitor in 2,4,6-trinitrobenzene sulfonic acid induced mouse colitis; and to elucidate the underlying mechanism of ROCK1/ROCK2 inhibition in enhancing intestinal epithelial barrier (IEB) function. A specific inhibitor of ROCK, Y-27632, was used to examine the role of ROCK in mouse colitis models. ROCK1 and ROCK2 were silenced respectively using RNA interference in Caco-2 cells. The expression of tight junction proteins and the downstream molecules of ROCK were assessed. Y-27632 alleviated colonic inflammation and decreased intestinal permeability. ROCK-myosin light chain (MLC) and ROCK-NF-κB pathway were activated in colitis and inhibited by Y-27632. In vitro, ROCK1 RNAi primarily downregulated the phosphorylation of myosin phosphatase-targeting subunit-1 (MYPT-1) and MLC, while ROCK2 RNAi inhibited phosphorylation of nuclear factor-κB (NF-κB). In conclusion, the results suggested that the ROCK inhibitor alleviated colitis and IEB dysfunction. Inhibition of phospho-MYPT-1 and MLC by ROCK1 knockout or inhibition of NF-κB phosphorylation by ROCK2 knockout may be the underlying mechanisms.
Collapse
Affiliation(s)
- Yanting Zou
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Xuhui, Shanghai 200032, P.R. China
| | - Lili Ma
- Endoscopy Center, Zhongshan Hospital, Fudan University, Xuhui, Shanghai 200032, P.R. China
| | - Yuan Zhao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Xuhui, Shanghai 200032, P.R. China
| | - Shuncai Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Xuhui, Shanghai 200032, P.R. China
| | - Chaohui Zhou
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Xuhui, Shanghai 200032, P.R. China
| | - Yu Cai
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Xuhui, Shanghai 200032, P.R. China
| |
Collapse
|
28
|
Intestinal Epithelial Ecto-5'-Nucleotidase (CD73) Regulates Intestinal Colonization and Infection by Nontyphoidal Salmonella. Infect Immun 2017; 85:IAI.01022-16. [PMID: 28717030 DOI: 10.1128/iai.01022-16] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 07/03/2017] [Indexed: 12/23/2022] Open
Abstract
Ecto-5'-nucleotidase (CD73) is expressed abundantly on the apical surface of intestinal epithelial cells (IECs) and functions as the terminal enzyme in the generation of extracellular adenosine. Previous work demonstrated that adenosine signaling in IECs results in a number of tissue-protective effects during inflammation; however, a rationale for its apical expression has been lacking. We hypothesized that the highly polarized expression of CD73 is indicative of an important role for extracellular adenosine as a mediator of host-microbe interactions. We show that adenosine harbors bacteriostatic activity against Salmonella enterica serovar Typhimurium that is not shared by the related purine metabolite 5'-AMP, inosine, or hypoxanthine. Analysis of Salmonella colonization in IEC-specific CD73 knockout mice (CD73f/fVillinCre ) revealed a nearly 10-fold increase in colonization compared to that in controls. Despite the increased luminal colonization by Salmonella, CD73f/fVillinCre mice were protected against Salmonella colitis and showed reduced Salmonella burdens in viscera, suggesting that adenosine promotes dissemination. The knockdown of CD73 expression in cultured IECs resulted in dramatic defects in intraepithelial localization and replication as well as defective transepithelial translocation by Salmonella In conclusion, we define a novel antimicrobial activity of adenosine in the gastrointestinal tract and unveil an important role for adenosine as a regulator of host-microbe interactions. These findings have broad implications for the development of new therapeutic agents for infectious disease.
Collapse
|
29
|
Jo H, Hwang D, Kim JK, Lim YH. Oxyresveratrol improves tight junction integrity through the PKC and MAPK signaling pathways in Caco-2 cells. Food Chem Toxicol 2017; 108:203-213. [PMID: 28780155 DOI: 10.1016/j.fct.2017.08.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 07/14/2017] [Accepted: 08/01/2017] [Indexed: 12/18/2022]
Abstract
Strengthening intestinal tight junctions (TJ) provides an effective barrier from the external environment and is important for recovery from inflammatory bowel disease. Oxyresveratrol (OXY), an isomer of hydroxylated resveratrol, is isolated from many plants. The aim of this study was to investigate the effect of OXY on intestinal TJ and to elucidate the mechanism underlying the OXY-mediated increase in TJ integrity in human intestinal Caco-2 cells. OXY-treated Caco-2 cell monolayers showed decreased monolayer permeability as evaluated by paracellular transport assay. The results showed that OXY significantly increased the levels of TJ-related genes and proteins (Claudin-1, Occludin and ZO-1) compared with those of the negative control. OXY activated protein kinase C (PKC) and increased expression levels of mitogen-activated protein kinase (MAPK) genes. OXY also increased gene and protein levels of the transcription factor Cdx-2. Expression levels of TJ, PKC and Cdx-2 proteins and transepithelial electrical resistance (TEER) value decreased in OXY-treated Caco-2 cells following treatment with a pan-PKC inhibitor compared with those of the untreated control. In conclusion, OXY strengthens the integrity of the intestinal TJ barrier via activation of the PKC and MAPK pathways.
Collapse
Affiliation(s)
- HyunA Jo
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 136-701, Republic of Korea
| | - Dahyun Hwang
- Department of Biomedical Laboratory Science, College of Life and Health Sciences, Hoseo University, Asan 31499, Republic of Korea
| | - Jeong-Keun Kim
- Department of Chemical Engineering and Biotechnology, Korea Polytechnic University, Shihung-si, Gyeonggi-do 429-793, Republic of Korea
| | - Young-Hee Lim
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 136-701, Republic of Korea; Department of Public Health Science (Brain Korea 21 PLUS Program), Graduate School, Korea University, Seoul 136-701, Republic of Korea; Department of Laboratory Medicine, Korea University Guro Hospital, Seoul 152-703, Republic of Korea.
| |
Collapse
|
30
|
Yang L, Chen X, Simet SM, Hu G, Cai Y, Niu F, Kook Y, Buch SJ. Reactive Oxygen Species/Hypoxia-Inducible Factor-1α/Platelet-Derived Growth Factor-BB Autocrine Loop Contributes to Cocaine-Mediated Alveolar Epithelial Barrier Damage. Am J Respir Cell Mol Biol 2017; 55:736-748. [PMID: 27391108 DOI: 10.1165/rcmb.2016-0096oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Abuse of psychostimulants, such as cocaine, has been shown to be closely associated with complications of the lung, such as pulmonary hypertension, edema, increased inflammation, and infection. However, the mechanism by which cocaine mediates impairment of alveolar epithelial barrier integrity that underlies various pulmonary complications has not been well determined. Herein, we investigate the role of cocaine in disrupting the alveolar epithelial barrier function and the associated signaling cascade. Using the combinatorial electric cell-substrate impedance sensing and FITC-dextran permeability assays, we demonstrated cocaine-mediated disruption of the alveolar epithelial barrier, as evidenced by increased epithelial monolayer permeability with a concomitant loss of the tight junction protein zonula occludens-1 (Zo-1) in both mouse primary alveolar epithelial cells and the alveolar epithelial cell line, L2 cells. To dissect the signaling pathways involved in this process, we demonstrated that cocaine-mediated induction of permeability factors, platelet-derived growth factor (PDGF-BB) and vascular endothelial growth factor, involved reactive oxygen species (ROS)-dependent induction of hypoxia-inducible factor (HIF)-1α. Interestingly, we demonstrated that ROS-dependent induction of another transcription factor, nuclear factor erythroid-2-related factor-2, that did not play a role in cocaine-mediated barrier dysfunction. Importantly, this study identifies, for the first time, that ROS/HIF-1α/PDGF-BB autocrine loop contributes to cocaine-mediated barrier disruption via amplification of oxidative stress and downstream signaling. Corroboration of these cell culture findings in vivo demonstrated increased permeability of the alveolar epithelial barrier, loss of expression of Zo-1, and a concomitantly increased expression of both HIF-1α and PDGF-BB. Pharmacological blocking of HIF-1α significantly abrogated cocaine-mediated loss of Zo-1. Understanding the mechanism(s) by which cocaine mediates barrier dysfunction could provide insights into the development of potential therapeutic targets for cocaine-mediated pulmonary hypertension.
Collapse
Affiliation(s)
- Lu Yang
- 1 School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xufeng Chen
- 2 Department of Emergence, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China; and
| | - Samantha M Simet
- 3 Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska
| | - Guoku Hu
- 1 School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.,3 Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska
| | - Yu Cai
- 3 Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska
| | - Fang Niu
- 3 Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska
| | - Yeonhee Kook
- 3 Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska
| | - Shilpa J Buch
- 3 Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
31
|
Stamatovic SM, Johnson AM, Sladojevic N, Keep RF, Andjelkovic AV. Endocytosis of tight junction proteins and the regulation of degradation and recycling. Ann N Y Acad Sci 2017; 1397:54-65. [PMID: 28415156 DOI: 10.1111/nyas.13346] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/08/2017] [Accepted: 03/09/2017] [Indexed: 12/31/2022]
Abstract
Internalization of tight junction (TJ) proteins from the plasma membrane is a pivotal mechanism regulating TJ plasticity and function in both epithelial and endothelial barrier tissues. Once internalized, the TJ proteins enter complex vesicular machinery, where further trafficking is directly dependent on the initiating stimulus and downstream signaling pathways that regulate the sorting and destiny of TJ proteins, as well as on cell and barrier responses. The destiny of internalized TJ proteins is recycling to the plasma membrane or sorting to late endosomes and degradation. This review highlights recent advances in our knowledge of endocytosis and vesicular trafficking of TJ proteins in both epithelial and endothelial cells. A greater understanding of these processes may allow for the development of methods to modulate barrier permeability for drug delivery or prevent barrier dysfunction in disease states.
Collapse
Affiliation(s)
| | | | | | - Richard F Keep
- Neurosurgery.,Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
| | | |
Collapse
|
32
|
Mend Your Fences: The Epithelial Barrier and its Relationship With Mucosal Immunity in Inflammatory Bowel Disease. Cell Mol Gastroenterol Hepatol 2017; 4:33-46. [PMID: 28560287 PMCID: PMC5439240 DOI: 10.1016/j.jcmgh.2017.03.007] [Citation(s) in RCA: 415] [Impact Index Per Article: 51.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 03/20/2017] [Indexed: 12/12/2022]
Abstract
The intestinal epithelium can be easily disrupted during gut inflammation as seen in inflammatory bowel disease (IBD), such as ulcerative colitis or Crohn's disease. For a long time, research into the pathophysiology of IBD has been focused on immune cell-mediated mechanisms. Recent evidence, however, suggests that the intestinal epithelium might play a major role in the development and perpetuation of IBD. It is now clear that IBD can be triggered by disturbances in epithelial barrier integrity via dysfunctions in intestinal epithelial cell-intrinsic molecular circuits that control the homeostasis, renewal, and repair of intestinal epithelial cells. The intestinal epithelium in the healthy individual represents a semi-permeable physical barrier shielding the interior of the body from invasions of pathogens on the one hand and allowing selective passage of nutrients on the other hand. However, the intestinal epithelium must be considered much more than a simple physical barrier. Instead, the epithelium is a highly dynamic tissue that responds to a plenitude of signals including the intestinal microbiota and signals from the immune system. This epithelial response to these signals regulates barrier function, the composition of the microbiota, and mucosal immune homeostasis within the lamina propria. The epithelium can thus be regarded as a translator between the microbiota and the immune system and aberrant signal transduction between the epithelium and adjacent immune cells might promote immune dysregulation in IBD. This review summarizes the important cellular and molecular barrier components of the intestinal epithelium and emphasizes the mechanisms leading to barrier dysfunction during intestinal inflammation.
Collapse
Key Words
- BMP, bone morphogenic protein
- CD, Crohn's disease
- Fz, frizzled
- HD, humans α-defensin
- IBD, inflammatory bowel disease
- IECs, intestinal epithelial cells
- IL, interleukin
- Immune-Epithelial Crosstalk
- Intestinal Epithelial Barrier
- Intestinal Inflammation
- JAMs, junctional adhesion molecules
- Lgr5, leucine rich repeat containing G-protein coupled receptor 5
- MARVEL, myelin and lymphocyte and related proteins for vesicle trafficking and membrane link
- MLCK, myosin light chain kinase
- NFκB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NOD-2, nucleotide-binding oligomerization domain-containing protein 2
- STAT, signal transducer and activator of transcription
- TAMP, tight junction–associated MARVEL protein
- TJ, tight junction
- TNF, tumor necrosis factor
- TSLP, thymic stromal lymphopoietin
- UC, ulcerative colitis
Collapse
|
33
|
Lechuga S, Ivanov AI. Disruption of the epithelial barrier during intestinal inflammation: Quest for new molecules and mechanisms. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1183-1194. [PMID: 28322932 DOI: 10.1016/j.bbamcr.2017.03.007] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 03/13/2017] [Accepted: 03/15/2017] [Indexed: 02/06/2023]
Abstract
The intestinal epithelium forms a key protective barrier that separates internal organs from the harmful environment of the gut lumen. Increased permeability of the gut barrier is a common manifestation of different inflammatory disorders contributing to the severity of disease. Barrier permeability is controlled by epithelial adherens junctions and tight junctions. Junctional assembly and integrity depend on fundamental homeostatic processes such as cell differentiation, rearrangements of the cytoskeleton, and vesicle trafficking. Alterations of intestinal epithelial homeostasis during mucosal inflammation may impair structure and remodeling of apical junctions, resulting in increased permeability of the gut barrier. In this review, we summarize recent advances in our understanding of how altered epithelial homeostasis affects the structure and function of adherens junctions and tight junctions in the inflamed gut. Specifically, we focus on the transcription reprogramming of the cell, alterations in the actin cytoskeleton, and junctional endocytosis and exocytosis. We pay special attention to knockout mouse model studies and discuss the relevance of these mechanisms to human gastrointestinal disorders.
Collapse
Affiliation(s)
- Susana Lechuga
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Andrei I Ivanov
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA; Virginia Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
34
|
Huang L, Deng M, He Y, Lu S, Ma R, Fang Y. β-asarone and levodopa co-administration increase striatal dopamine level in 6-hydroxydopamine induced rats by modulating P-glycoprotein and tight junction proteins at the blood-brain barrier and promoting levodopa into the brain. Clin Exp Pharmacol Physiol 2016; 43:634-43. [PMID: 26991136 DOI: 10.1111/1440-1681.12570] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 03/09/2016] [Accepted: 03/10/2016] [Indexed: 01/06/2023]
Affiliation(s)
- Liping Huang
- Hainan Medical University; Haikou China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine; Guangzhou China
| | - Minzhen Deng
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine; Guangzhou China
| | - Yuping He
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine; Guangzhou China
| | - Shiyao Lu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine; Guangzhou China
| | - Ruanxin Ma
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine; Guangzhou China
| | - Yongqi Fang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine; Guangzhou China
| |
Collapse
|
35
|
Experimental Colitis Is Attenuated by Cardioprotective Diet Supplementation That Reduces Oxidative Stress, Inflammation, and Mucosal Damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:8473242. [PMID: 26881044 PMCID: PMC4736595 DOI: 10.1155/2016/8473242] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 12/01/2015] [Indexed: 12/19/2022]
Abstract
Inflammatory bowel diseases (IBD) such as ulcerative colitis (UC) and Crohn's disease (CD) are multifactorial, relapsing disorders of the gastrointestinal tract. However, the etiology is still poorly understood but involves altered immune responses, epithelial dysfunction, environmental factors, and nutrition. Recently, we have shown that the diet supplement corabion has cardioprotective effects due to reduction of oxidative stress and inflammation. Since oxidative stress and inflammation are also prominent risk factors in IBD, we speculated that corabion also has beneficial effects on experimental colitis. Colitis was induced in male mice by administration of 3.5% (w/v) dextran sulfate sodium (DSS) in drinking water for a period of 3 or 7 days with or without daily gavage feeding of corabion consisting of vitamin C, vitamin E, L-arginine, and eicosapentaenoic and docosahexaenoic acid. We found that corabion administration attenuated DSS-induced colon shortening, tissue damage, and disease activity index during the onset of colitis. Mechanistically, these effects could be explained by reduced neutrophil recruitment, oxidative stress, production of proinflammatory cytokines, and internalization of the junctional proteins ZO-1 and E-cadherin leading to less edema formation. Thus, corabion may be a useful diet supplement for the management of chronic inflammatory intestinal disorders such as IBD.
Collapse
|
36
|
Zhang S, Zheng S, Wang X, Shi Q, Wang X, Yuan S, Wang G, Ji Z. Carbon Monoxide-Releasing Molecule-2 Reduces Intestinal Epithelial Tight-Junction Damage and Mortality in Septic Rats. PLoS One 2015; 10:e0145988. [PMID: 26720630 PMCID: PMC4697838 DOI: 10.1371/journal.pone.0145988] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 11/13/2015] [Indexed: 02/07/2023] Open
Abstract
Objective Damage to intestinal epithelial tight junctions plays an important role in sepsis. Recently we found that Carbon Monoxide-Releasing Molecule-2 (CORM-2) is able to protect LPS-induced intestinal epithelial tight junction damage and in this study we will investigate if CORM-2 could protect intestinal epithelial tight junctions in the rat cecal ligation and puncture (CLP) model. Materials and Methods The CLP model was generated using male Sprague-Dawley (SD) rats according to standard procedure and treated with CORM-2 or inactive CORM-2 (iCORM-2), 8 mg/kg, i.v. immediately after CLP induction and euthanized after 24h or 72h (for mortality rate only). Morphological changes were investigated using both transmission electron and confocal microscopy. The levels of important TJ proteins and phosphorylation of myosin light chain (MLC) were examined using Western blotting. Cytokines, IL-1β and TNF-α were measured using ELISA kits. The overall intestinal epithelial permeability was evaluated using FD-4 as a marker. Results CORM-2, but not iCORM-2, significantly reduced sepsis-induced damage of intestinal mucosa (including TJ disruption), TJ protein reduction (including zonula occludens-l (ZO-1), claudin-1 and occludin), MLC phosphorylation and proinflammatory cytokine release. The overall outcomes showed that CORM-2 suppressed sepsis-induced intestinal epithelial permeability changes and reduced mortality rate of those septic rats. Conclusions Our data strongly suggest that CORM-2 could be a potential therapeutic reagent for sepsis by suppressing inflammation, restoring intestinal epithelial barrier and reducing mortality.
Collapse
Affiliation(s)
- Shulong Zhang
- Department of General Surgery, Zhongda Hospital, Southeast University Medical School, Nanjing, Jiangsu 210009, China
| | - Shuyun Zheng
- Department of Critical Care Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Xin Wang
- Department of General Surgery, Zhongda Hospital, Southeast University Medical School, Nanjing, Jiangsu 210009, China
| | - Qiankun Shi
- Department of Critical Care Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Xiang Wang
- Department of Critical Care Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Shoutao Yuan
- Department of Critical Care Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Guozheng Wang
- Institute of Infection and Global Health, University of Liverpool, Liverpool, L69 7BE, United Kingdom
| | - Zhenling Ji
- Department of General Surgery, Zhongda Hospital, Southeast University Medical School, Nanjing, Jiangsu 210009, China
- * E-mail: ;
| |
Collapse
|
37
|
Liu W, Hu D, Huo H, Zhang W, Adiliaghdam F, Morrison S, Ramirez JM, Gul SS, Hamarneh SR, Hodin RA. Intestinal Alkaline Phosphatase Regulates Tight Junction Protein Levels. J Am Coll Surg 2015; 222:1009-17. [PMID: 27106638 DOI: 10.1016/j.jamcollsurg.2015.12.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/02/2015] [Accepted: 12/03/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND Intestinal alkaline phosphatase (IAP) plays a pivotal role in maintaining gut health and well-being. Oral supplementation with IAP in mice improves gut barrier function and prevents luminal proinflammatory factors from gaining access to the circulation. In this study, we sought to explore the relationship between IAP and tight junction protein (TJP) expression and function. STUDY DESIGN The effect of IAP deletion on TJP levels was studied in mouse embryonic fibroblasts (MEFs) generated from IAP-knockout and wild type mice. Regulation of TJPs by IAP was assayed in the human colon cancer Caco-2 and T84 cells by overexpressing the human IAP gene. Tight junction protein levels and localization were measured by using RT q-PCR and antibodies targeting the specific TJPs. Finally, the effect of IAP on inflammation-induced intestinal permeability was measured by in vitro trans-well epithelial electrical resistance (TEER). RESULTS Intestinal alkaline phosphatase gene deletion in MEFs resulted in significantly lower levels of ZO-1, ZO-2, and Occludin compared with levels in wild-type control cells; IAP overexpression in Caco-2 and T84 cells resulted in approximate 2-fold increases in the mRNA levels of ZO-1 and ZO-2. The IAP treatment ameliorated lipopolysaccharide-induced increased permeability in the Caco-2 trans-well system. Furthermore, IAP treatment preserved the localization of the ZO-1 and Occludin proteins during inflammation and was also associated with improved epithelial barrier function. CONCLUSIONS Intestinal alkaline phosphatase is a major regulator of gut mucosal permeability and appears to work at least partly through improving TJP levels and localization. These data provide a strong foundation to develop IAP as a novel therapy to maintain gut barrier function.
Collapse
Affiliation(s)
- Wei Liu
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Dong Hu
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Haizhong Huo
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Weifeng Zhang
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Fatemeh Adiliaghdam
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Sarah Morrison
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Juan M Ramirez
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Sarah S Gul
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Sulaiman R Hamarneh
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Richard A Hodin
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
38
|
Saeedi BJ, Kao DJ, Kitzenberg DA, Dobrinskikh E, Schwisow KD, Masterson JC, Kendrick AA, Kelly CJ, Bayless AJ, Kominsky DJ, Campbell EL, Kuhn KA, Furuta GT, Colgan SP, Glover LE. HIF-dependent regulation of claudin-1 is central to intestinal epithelial tight junction integrity. Mol Biol Cell 2015; 26:2252-62. [PMID: 25904334 PMCID: PMC4462943 DOI: 10.1091/mbc.e14-07-1194] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 04/15/2015] [Indexed: 01/30/2023] Open
Abstract
This study demonstrates a critical link between hypoxia-inducible factor (HIF) and claudin-1 (CLDN1). HIF1β-deficient intestinal epithelial cells develop abnormal tight junction (TJ) structure and have striking barrier defects. CLDN1 is an HIF target gene, and overexpression of CLDN1 in HIF1β-deficient cells restores TJ structure and function. Intestinal epithelial cells (IECs) are exposed to profound fluctuations in oxygen tension and have evolved adaptive transcriptional responses to a low-oxygen environment. These adaptations are mediated primarily through the hypoxia-inducible factor (HIF) complex. Given the central role of the IEC in barrier function, we sought to determine whether HIF influenced epithelial tight junction (TJ) structure and function. Initial studies revealed that short hairpin RNA–mediated depletion of the HIF1β in T84 cells resulted in profound defects in barrier and nonuniform, undulating TJ morphology. Global HIF1α chromatin immunoprecipitation (ChIP) analysis identified claudin-1 (CLDN1) as a prominent HIF target gene. Analysis of HIF1β-deficient IEC revealed significantly reduced levels of CLDN1. Overexpression of CLDN1 in HIF1β-deficient cells resulted in resolution of morphological abnormalities and restoration of barrier function. ChIP and site-directed mutagenesis revealed prominent hypoxia response elements in the CLDN1 promoter region. Subsequent in vivo analysis revealed the importance of HIF-mediated CLDN1 expression during experimental colitis. These results identify a critical link between HIF and specific tight junction function, providing important insight into mechanisms of HIF-regulated epithelial homeostasis.
Collapse
Affiliation(s)
- Bejan J Saeedi
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Daniel J Kao
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - David A Kitzenberg
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Evgenia Dobrinskikh
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kayla D Schwisow
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Joanne C Masterson
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 Section of Pediatric Gastroenterology, Hepatology and Nutrition, Gastrointestinal Eosinophilic Diseases Program, Department of Pediatrics, Digestive Health Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Agnieszka A Kendrick
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Caleb J Kelly
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Amanda J Bayless
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Douglas J Kominsky
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 Department of Anesthesiology and Perioperative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Eric L Campbell
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kristine A Kuhn
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Glenn T Furuta
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Sean P Colgan
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Louise E Glover
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
39
|
|
40
|
A novel approach to maintain gut mucosal integrity using an oral enzyme supplement. Ann Surg 2014; 260:706-14; discussion 714-5. [PMID: 25203888 DOI: 10.1097/sla.0000000000000916] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE To determine the role of intestinal alkaline phosphatase (IAP) in enteral starvation-induced gut barrier dysfunction and to study its therapeutic effect as a supplement to prevent gut-derived sepsis. BACKGROUND Critically ill patients are at increased risk for systemic sepsis and, in some cases, multiorgan failure leading to death. Years ago, the gut was identified as a major source for this systemic sepsis syndrome. Previously, we have shown that IAP detoxifies bacterial toxins, prevents endotoxemia, and preserves intestinal microbiotal homeostasis. METHODS WT and IAP-KO mice were used to examine gut barrier function and tight junction protein levels during 48-hour starvation and fed states. Human ileal fluid samples were collected from 20 patients postileostomy and IAP levels were compared between fasted and fed states. To study the effect of IAP supplementation on starvation-induced gut barrier dysfunction, WT mice were fasted for 48 hours +/- IAP supplementation in the drinking water. RESULTS The loss of IAP expression is associated with decreased expression of intestinal junctional proteins and impaired barrier function. For the first time, we demonstrate that IAP expression is also decreased in humans who are deprived of enteral feeding. Finally, our data demonstrate that IAP supplementation reverses the gut barrier dysfunction and tight junction protein losses due to a lack of enteral feeding. CONCLUSIONS IAP is a major regulator of gut mucosal permeability and is able to ameliorate starvation-induced gut barrier dysfunction. Enteral IAP supplementation may represent a novel approach to maintain bowel integrity in critically ill patients.
Collapse
|
41
|
Ivanov AI. Tissue Barriers: Introducing an exciting new journal. Temperature (Austin) 2014; 1:151-3. [PMID: 27626042 PMCID: PMC5008708 DOI: 10.4161/23328940.2014.978716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 10/16/2014] [Accepted: 10/16/2014] [Indexed: 11/19/2022] Open
Abstract
This Editorial is written to introduce Tissue Barriers, a new Taylor & Francis journal, to the readers of Temperature. It describes the role of temperature in the regulation of different tissue barriers under normal and disease conditions. It also highlights the most interesting articles published in the first volume of Tissue Barriers.
Collapse
Affiliation(s)
- Andrei I Ivanov
- Department of Human and Molecular Genetics; Virginia Institute of Molecular Medicine; VCU Massey Cancer Center; Virginia Commonwealth University ; Richmond, VA USA
| |
Collapse
|
42
|
Wan HT, Mruk DD, Tang EI, Xiao X, Cheng YH, Wong EWP, Wong CKC, Cheng CY. Role of non-receptor protein tyrosine kinases in spermatid transport during spermatogenesis. Semin Cell Dev Biol 2014; 30:65-74. [PMID: 24727349 DOI: 10.1016/j.semcdb.2014.04.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Accepted: 04/04/2014] [Indexed: 12/16/2022]
Abstract
Non-receptor protein tyrosine kinases are cytoplasmic kinases that activate proteins by phosphorylating tyrosine residues, which in turn affect multiple functions in eukaryotic cells. Herein, we focus on the role of non-receptor protein tyrosine kinases, most notably, FAK, c-Yes and c-Src, in the transport of spermatids across the seminiferous epithelium during spermatogenesis. Since spermatids, which are formed from spermatocytes via meiosis, are immotile haploid cells, they must be transported by Sertoli cells across the seminiferous epithelium during the epithelial cycle of spermatogenesis. Without the timely transport of spermatids across the epithelium, the release of sperms at spermiation fails to occur, leading to infertility. Thus, the molecular event pertinent to spermatid transport is crucial to spermatogenesis. We provide a critical discussion based on recent findings in this review. We also provide a hypothetical model on spermatid transport, and the role of non-receptor protein tyrosine kinases in this event. We also highlight areas of research that deserve attention by investigators in the field.
Collapse
Affiliation(s)
- H T Wan
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, United States
| | - Dolores D Mruk
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, United States
| | - Elizabeth I Tang
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, United States
| | - Xiang Xiao
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, United States
| | - Yan-Ho Cheng
- Richmond University Medical Center, Staten Island, NY 10301, United States
| | - Elissa W P Wong
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, United States
| | - Chris K C Wong
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong, China
| | - C Yan Cheng
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, United States.
| |
Collapse
|
43
|
Citalán-Madrid AF, García-Ponce A, Vargas-Robles H, Betanzos A, Schnoor M. Small GTPases of the Ras superfamily regulate intestinal epithelial homeostasis and barrier function via common and unique mechanisms. Tissue Barriers 2013; 1:e26938. [PMID: 24868497 PMCID: PMC3942330 DOI: 10.4161/tisb.26938] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 10/21/2013] [Accepted: 10/24/2013] [Indexed: 12/11/2022] Open
Abstract
The intestinal epithelium forms a stable barrier protecting underlying tissues from pathogens in the gut lumen. This is achieved by specialized integral membrane structures such as tight and adherens junctions that connect neighboring cells and provide stabilizing links to the cytoskeleton. Junctions are constantly remodeled to respond to extracellular stimuli. Assembly and disassembly of junctions is regulated by interplay of actin remodeling, endocytotic recycling of junctional proteins, and various signaling pathways. Accumulating evidence implicate small G proteins of the Ras superfamily as important signaling molecules for the regulation of epithelial junctions. They function as molecular switches circling between an inactive GDP-bound and an active GTP-bound state. Once activated, they bind different effector molecules to control cellular processes required for correct junction assembly, maintenance and remodelling. Here, we review recent advances in understanding how GTPases of the Rho, Ras, Rab and Arf families contribute to intestinal epithelial homeostasis.
Collapse
Affiliation(s)
- Alí Francisco Citalán-Madrid
- Department of Molecular Biomedicine; Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav del IPN); Mexico City, Mexico
| | - Alexander García-Ponce
- Department of Molecular Biomedicine; Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav del IPN); Mexico City, Mexico
| | - Hilda Vargas-Robles
- Department of Molecular Biomedicine; Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav del IPN); Mexico City, Mexico
| | - Abigail Betanzos
- Department of Infectomics and Molecular Pathogenesis; Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav del IPN); Mexico City, Mexico
| | - Michael Schnoor
- Department of Molecular Biomedicine; Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav del IPN); Mexico City, Mexico
| |
Collapse
|
44
|
Xiao X, Mruk DD, Cheng CY. Intercellular adhesion molecules (ICAMs) and spermatogenesis. Hum Reprod Update 2013; 19:167-86. [PMID: 23287428 DOI: 10.1093/humupd/dms049] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND During the seminiferous epithelial cycle, restructuring takes places at the Sertoli-Sertoli and Sertoli-germ cell interface to accommodate spermatogonia/spermatogonial stem cell renewal via mitosis, cell cycle progression and meiosis, spermiogenesis and spermiation since developing germ cells, in particular spermatids, move 'up and down' the seminiferous epithelium. Furthermore, preleptotene spermatocytes differentiated from type B spermatogonia residing at the basal compartment must traverse the blood-testis barrier (BTB) to enter the adluminal compartment to prepare for meiosis at Stage VIII of the epithelial cycle, a process also accompanied by the release of sperm at spermiation. These cellular events that take place at the opposite ends of the epithelium are co-ordinated by a functional axis designated the apical ectoplasmic specialization (ES)-BTB-basement membrane. However, the regulatory molecules that co-ordinate cellular events in this axis are not known. METHODS Literature was searched at http://www.pubmed.org and http://scholar.google.com to identify published findings regarding intercellular adhesion molecules (ICAMs) and the regulation of this axis. RESULTS Members of the ICAM family, namely ICAM-1 and ICAM-2, and the biologically active soluble ICAM-1 (sICAM-1) are the likely regulatory molecules that co-ordinate these events. sICAM-1 and ICAM-1 have antagonistic effects on the Sertoli cell tight junction-permeability barrier, involved in Sertoli cell BTB restructuring, whereas ICAM-2 is restricted to the apical ES, regulating spermatid adhesion during the epithelial cycle. Studies in other epithelia/endothelia on the role of the ICAM family in regulating cell movement are discussed and this information has been evaluated and integrated into studies of these proteins in the testis to create a hypothetical model, depicting how ICAMs regulate junction restructuring events during spermatogenesis. CONCLUSIONS ICAMs are crucial regulatory molecules of spermatogenesis. The proposed hypothetical model serves as a framework in designing functional experiments for future studies.
Collapse
Affiliation(s)
- Xiang Xiao
- The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10065, USA
| | | | | |
Collapse
|