1
|
Layunta E, Latorre E, Grasa L, Arruebo MP, Buey B, Alcalde AI, Mesonero JE. Intestinal serotonergic system is modulated by Toll-like receptor 9. J Physiol Biochem 2022; 78:689-701. [PMID: 35670957 PMCID: PMC9381617 DOI: 10.1007/s13105-022-00897-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/14/2022] [Indexed: 12/03/2022]
Abstract
Intestinal serotonergic system is a key modulator of intestinal homeostasis; however, its regulation is still unclear. Toll-like receptor 9 (TLR9), an innate immune receptor, detects different external agents in the intestine, preserving intestinal integrity. Since little is known about TLR9 role in the intestine, our aim was to address the potential regulation between TLR9 and intestinal serotonergic system. Caco-2/TC7 cell line and intestinal tract of Tlr9−/− mice were used in this study. Serotonin uptake studies were performed, and molecular expression of different serotonergic components was analyzed by western blot and real-time PCR. Our results show that TLR9 activation inhibits serotonin transporter activity and expression, involving p38/MAPK and ERK/MAPK intracellular pathways, and reciprocally, serotonin increases TLR9 expression. Supporting this interaction, serotonin transporter, serotonin receptors and serotonin producer enzymes were found altered in intestinal tract of Tlr9−/− mice. We conclude that TLR9 could contribute to intestinal homeostasis by modulation of intestinal serotonergic system.
Collapse
Affiliation(s)
- Elena Layunta
- Institute of Biomedicine, Department of Medical Biochemistry and Cell Biology, University of Gothenburg, Gothenburg, Sweden.,Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Spain
| | - Eva Latorre
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Spain. .,Instituto Agroalimentario de Aragón - IA2- (Universidad de Zaragoza - CITA), Zaragoza, Spain. .,Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Pedro Cerbuna 12, 50009, Zaragoza, Spain.
| | - Laura Grasa
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Spain.,Instituto Agroalimentario de Aragón - IA2- (Universidad de Zaragoza - CITA), Zaragoza, Spain.,Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
| | - María Pilar Arruebo
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Spain.,Instituto Agroalimentario de Aragón - IA2- (Universidad de Zaragoza - CITA), Zaragoza, Spain.,Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
| | - Berta Buey
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
| | - Ana I Alcalde
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
| | - José E Mesonero
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Spain.,Instituto Agroalimentario de Aragón - IA2- (Universidad de Zaragoza - CITA), Zaragoza, Spain.,Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
| |
Collapse
|
2
|
Su X, Wei J, Qi H, Jin M, Zhang Q, Zhang Y, Zhang C, Yang R. LRRC19 Promotes Permeability of the Gut Epithelial Barrier Through Degrading PKC-ζ and PKCι/λ to Reduce Expression of ZO1, ZO3, and Occludin. Inflamm Bowel Dis 2021; 27:1302-1315. [PMID: 33501933 DOI: 10.1093/ibd/izaa354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND A dysfunctional gut epithelial barrier allows the augmented permeation of endotoxins, luminal antigens, and bacteria into the bloodstream, causing disease. The maintenance of gut epithelial barrier integrity may be regulated by multiple factors. Herein we analyze the role of leucine-rich repeat-containing protein 19 (LRRC19) in regulating the permeability of the gut epithelial barrier. METHODS We utilized Lrrc19 knockout (KO) mice and clinical samples through transmission electron, intestinal permeability assay, Western blot, and immunofluorescence staining to characterize the role of LRRC19 in the permeability of the gut epithelial barrier. RESULTS We found that LRRC19, which is expressed in gut epithelial cells, impairs gut barrier function. Transmission electron micrographs revealed a tighter junction and narrower gaps in the colon epithelium cells in LRRC19 KO mice. There were lower levels of serum lipopolysaccharide and 4 kDa-fluorescein isothiocyanate-dextran after gavage in LRRC19 KO mice than in wild-type mice. We found that LRRC19 could reduce the expression of zonula occludens (ZO)-1, ZO-3, and occludin in the colonic epithelial cells. The decreased expression of ZO-1, ZO-3, and occludin was dependent on degrading protein kinase C (PKC) ζ and PKCι/λ through K48 ubiquitination by LRRC19. The expression of LRRC19 was also negatively correlated with ZO-1, ZO-3, occludin, PKCζ, and PKCι/λ in human colorectal cancers. CONCLUSIONS The protein LRRC19 can promote the permeability of the gut epithelial barrier through degrading PKC ζ and PKCι/λ to reduce the expression of ZO-1, ZO-3, and occludin.
Collapse
Affiliation(s)
- Xiaomin Su
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China.,Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, China.,Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Jianmei Wei
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China.,Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, China.,Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Houbao Qi
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China.,Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, China.,Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Mengli Jin
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China.,Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, China.,Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Qianjing Zhang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China.,Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, China.,Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Yuan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China.,Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, China.,Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Chunze Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Rongcun Yang
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China.,Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, China.,Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
3
|
Cao W, Li RW, Chin Y, Wang Y, Xue C, Tang Q. Transcriptome analysis reveals the protective role of fructo-oligosaccharide in colonic mucosal barriers in exercise-induced stressed mice. Food Funct 2021; 12:4484-4495. [PMID: 33885098 DOI: 10.1039/d0fo02556a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Most athletes continually endure mental and physical stress from intense exercise. Fructo-oligosaccharide (FOS) can reduce physical exhaustion, but the concrete mechanism behind it still needs further research. In this study, the effect of FOS on colonic mucosal barriers was investigated using an exercise-induced stress mouse model. Except for control individuals, mice were subject to cycles of 2-day exercise (at 20 rpm) interleaved by 5-day rest. The mice experienced a total of 6 days of exercise during the feeding period. FOS improved common indicators of exhaustion, such as glycogen storage in muscle. 16S rRNA data supported that changes in the gut microbiome were also closely related to stress status. Notably, Anaerotruncus was increased in mice under stress, while FOS facilitated the growth of Dorea, which is negatively associated with exhaustion. The RNA-seq analysis revealed that FOS could maintain the integrity of colonic epithelial barriers. For example, FOS significantly restored the expression of tight junctions (Occludin and Zonula occludens-1) in the colon, which was impaired under a stress state. Besides, the NOD-like receptor family pyrin domain containing 6 (NLRP6) inflammasome might contribute to the protection of the colonic mucosa by promoting the secretion of IL-18, Mucin2 (Muc2) and intestine lectin 1 (Itln1) in FOS-treated individuals. In short, FOS administration attenuated the damage of colonic mucosal barriers in exercise-induced stressed mice.
Collapse
Affiliation(s)
- Wanxiu Cao
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, China.
| | | | | | | | | | | |
Collapse
|
4
|
Sun L, Yang S, Deng Q, Dong K, Li Y, Wu S, Huang R. Salmonella Effector SpvB Disrupts Intestinal Epithelial Barrier Integrity for Bacterial Translocation. Front Cell Infect Microbiol 2020; 10:606541. [PMID: 33392110 PMCID: PMC7773751 DOI: 10.3389/fcimb.2020.606541] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/19/2020] [Indexed: 01/13/2023] Open
Abstract
Salmonella are common enteric bacterial pathogens that infect both humans and animals. Intestinal epithelial barrier, formed by a single layer of epithelial cells and apical junctional complex (AJC), plays a crucial role in host defense against enteric pathogens to prevent bacterial translocation. However, the underlying mechanisms of intestinal epithelial barrier dysfunction caused by Salmonella are poorly understood. It is found that a locus termed Salmonella plasmid virulence (spv) gene exists extensively in clinically important Salmonella serovars. SpvB is a key effector encoded within this locus, and closely related to Salmonella pathogenicity such as interfering with autophagy and iron homeostasis. To investigate the interaction between SpvB and intestinal epithelial barrier and elucidate the underlying molecular mechanism, we used the typical foodborne disease agent Salmonella enterica serovar Typhimurium (Salmonella typhimurium) carrying spvB or not to construct infection models in vivo and in vitro. C57BL/6 mice were orally challenged with S. typhimurium wild-type strain SL1344 or spvB-deficient mutant strain SL1344-ΔspvB. Caco-2 cell monolayer model, as a widely used model to mimic the human intestinal epithelium in vitro, was infected with SL1344, SL1344-ΔspvB, or spvB complementary strain SL1344-c-ΔspvB, respectively. The results showed that SpvB enhanced bacterial pathogenicity during S. typhimurium infection in vivo, and contributed to intestinal epithelial barrier dysfunction in both infection systems. This SpvB-mediated barrier dysfunction was attributed to the cellular redistribution of Claudin-1, Occludin, and E-cadherin junctional proteins. Moreover, by using pharmacological inhibitors, we found that F-actin rearrangement and suppression of protein kinase C (PKC) signaling pathway were involved in SpvB-mediated barrier dysfunction. In conclusion, the study reveals the contribution of Salmonella effector SpvB to the dysfunction of intestinal epithelial barrier integrity, which facilitates bacterial translocation via the paracellular route to promote Salmonella systemic dissemination. Our findings broaden the understanding of host–pathogen interactions in salmonellosis, and provide new strategies for the therapy in limiting bacterial dissemination during infection.
Collapse
Affiliation(s)
- Lanqing Sun
- Department of Medical Microbiology, School of Biology and Basic Medical Sciences, Medical College of Soochow University, Suzhou, China
| | - Sidi Yang
- Department of Medical Microbiology, School of Biology and Basic Medical Sciences, Medical College of Soochow University, Suzhou, China
| | - Qifeng Deng
- Department of Medical Microbiology, School of Biology and Basic Medical Sciences, Medical College of Soochow University, Suzhou, China
| | - Kedi Dong
- Department of Medical Microbiology, School of Biology and Basic Medical Sciences, Medical College of Soochow University, Suzhou, China
| | - Yuanyuan Li
- Department of Medical Microbiology, School of Biology and Basic Medical Sciences, Medical College of Soochow University, Suzhou, China
| | - Shuyan Wu
- Department of Medical Microbiology, School of Biology and Basic Medical Sciences, Medical College of Soochow University, Suzhou, China
| | - Rui Huang
- Department of Medical Microbiology, School of Biology and Basic Medical Sciences, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
5
|
Ren C, Zhang Q, de Haan BJ, Faas MM, Zhang H, de Vos P. Protective effects of lactic acid bacteria on gut epithelial barrier dysfunction are Toll like receptor 2 and protein kinase C dependent. Food Funct 2020; 11:1230-1234. [PMID: 32043507 DOI: 10.1039/c9fo02933h] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Lactic acid bacteria (LAB) are recognized for support of host gut homeostasis but the precise mechanisms remain to be identified. LABs interact with Toll-like receptors (TLRs) which might stimulate barrier function of gut epithelial cells. We previously identified six TLR2-signalling LAB strains. As TLR2 is involved in barrier-function enhancement in gut-epithelium, the epithelial barrier-protective effect of these TLR2-signalling strains was studied by using T84 human colorectal cancer cell monolayer as an in vitro gut epithelial barrier model. The protein kinase C (PKC) dependent barrier disruptor A23187 and mitogen-activated protein kinase dependent barrier stressor deoxynivalenol were tested to determine which pathways LAB influenced. We found that exclusively the PKC dependent disruption was prevented by the selected TLR2-signalling LAB strains. This study suggests that TLR2 is a pivotal epithelial barrier modulator, and provides novel insight in the molecular mechanisms by which LAB contribute to intestinal health.
Collapse
Affiliation(s)
- Chengcheng Ren
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands. and School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
| | - Qiuxiang Zhang
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
| | - Bart J de Haan
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands.
| | - Marijke M Faas
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands.
| | - Hao Zhang
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, China
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands.
| |
Collapse
|
6
|
Paveljšek D, Ivičak-Kocjan K, Treven P, Benčina M, Jerala R, Rogelj I. Distinctive probiotic features share common TLR2-dependent signalling in intestinal epithelial cells. Cell Microbiol 2020; 23:e13264. [PMID: 32945079 PMCID: PMC7757178 DOI: 10.1111/cmi.13264] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/11/2020] [Accepted: 09/13/2020] [Indexed: 12/14/2022]
Abstract
The underlying mechanisms of probiotics and postbiotics are not well understood, but it is known that both affect the adaptive and innate immune responses. In addition, there is a growing concept that some probiotic strains have common core mechanisms that provide certain health benefits. Here, we aimed to elucidate the signalization of the probiotic bacterial strains Lactobacillus paragasseri K7, Limosilactobacillus fermentum L930BB, Bifidobacterium animalis subsp. animalis IM386 and Lactiplantibacillus plantarum WCFS1. We showed in in vitro experiments that the tested probiotics exhibit common TLR2- and TLR10-dependent downstream signalling cascades involving inhibition of NF-κB signal transduction. Under inflammatory conditions, the probiotics activated phosphatidylinositol 3-kinase (PI3K)/Akt anti-apoptotic pathways and protein kinase C (PKC)-dependent pathways, which led to regulation of the actin cytoskeleton and tight junctions. These pathways contribute to the regeneration of the intestinal epithelium and modulation of the mucosal immune system, which, together with the inhibition of canonical TLR signalling, promote general immune tolerance. With this study we identified shared probiotic mechanisms and were the first to pinpoint the role of anti-inflammatory probiotic signalling through TLR10.
Collapse
Affiliation(s)
- Diana Paveljšek
- Biotechnical Faculty, Department of Animal Science, University of Ljubljana, Domžale, Slovenia
| | - Karolina Ivičak-Kocjan
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Primož Treven
- Biotechnical Faculty, Department of Animal Science, University of Ljubljana, Domžale, Slovenia
| | - Mojca Benčina
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, Slovenia
| | - Irena Rogelj
- Biotechnical Faculty, Department of Animal Science, University of Ljubljana, Domžale, Slovenia
| |
Collapse
|
7
|
Belhaouane I, Hoffmann E, Chamaillard M, Brodin P, Machelart A. Paradoxical Roles of the MAL/Tirap Adaptor in Pathologies. Front Immunol 2020; 11:569127. [PMID: 33072109 PMCID: PMC7544743 DOI: 10.3389/fimmu.2020.569127] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/24/2020] [Indexed: 12/14/2022] Open
Abstract
Toll-like receptors (TLRs) are at the forefront of pathogen recognition ensuring host fitness and eliciting protective cellular and humoral responses. Signaling pathways downstream of TLRs are tightly regulated for preventing collateral damage and loss of tolerance toward commensals. To trigger effective intracellular signaling, these receptors require the involvement of adaptor proteins. Among these, Toll/Interleukin-1 receptor domain containing adaptor protein (Tirap or MAL) plays an important role in establishing immune responses. Loss of function of MAL was associated with either disease susceptibility or resistance. These opposite effects reveal paradoxical functions of MAL and their importance in containing infectious or non-infectious diseases. In this review, we summarize the current knowledge on the signaling pathways involving MAL in different pathologies and their impact on inducing protective or non-protective responses.
Collapse
Affiliation(s)
- Imène Belhaouane
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Eik Hoffmann
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Mathias Chamaillard
- Laboratory of Cell Physiology, INSERM U1003, University of Lille, Lille, France
| | - Priscille Brodin
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Arnaud Machelart
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
8
|
Artificial Sweeteners Disrupt Tight Junctions and Barrier Function in the Intestinal Epithelium through Activation of the Sweet Taste Receptor, T1R3. Nutrients 2020; 12:nu12061862. [PMID: 32580504 PMCID: PMC7353258 DOI: 10.3390/nu12061862] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/18/2020] [Accepted: 06/20/2020] [Indexed: 12/20/2022] Open
Abstract
The breakdown of the intestinal epithelial barrier and subsequent increase in intestinal permeability can lead to systemic inflammatory diseases and multiple-organ failure. Nutrition impacts the intestinal barrier, with dietary components such as gluten increasing permeability. Artificial sweeteners are increasingly consumed by the general public in a range of foods and drinks. The sweet taste receptor (T1R3) is activated by artificial sweeteners and has been identified in the intestine to play a role in incretin release and glucose transport; however, T1R3 has not been previously linked to intestinal permeability. Here, the intestinal epithelial cell line, Caco-2, was used to study the effect of commonly-consumed artificial sweeteners, sucralose, aspartame and saccharin, on permeability. At high concentrations, aspartame and saccharin were found to induce apoptosis and cell death in intestinal epithelial cells, while at low concentrations, sucralose and aspartame increased epithelial barrier permeability and down-regulated claudin 3 at the cell surface. T1R3 knockdown was found to attenuate these effects of artificial sweeteners. Aspartame induced reactive oxygen species (ROS) production to cause permeability and claudin 3 internalization, while sweetener-induced permeability and oxidative stress was rescued by the overexpression of claudin 3. Taken together, our findings demonstrate that the artificial sweeteners sucralose, aspartame, and saccharin exert a range of negative effects on the intestinal epithelium through the sweet taste receptor T1R3.
Collapse
|
9
|
Garg M, Royce SG, Tikellis C, Shallue C, Batu D, Velkoska E, Burrell LM, Patel SK, Beswick L, Jackson A, Britto K, Lukies M, Sluka P, Wardan H, Hirokawa Y, Tan CW, Faux M, Burgess AW, Hosking P, Monagle S, Thomas M, Gibson PR, Lubel J. Imbalance of the renin-angiotensin system may contribute to inflammation and fibrosis in IBD: a novel therapeutic target? Gut 2020; 69:841-851. [PMID: 31409604 DOI: 10.1136/gutjnl-2019-318512] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 07/10/2019] [Accepted: 08/03/2019] [Indexed: 12/18/2022]
Abstract
OBJECTIVE We evaluated the influence of the renin-angiotensin system (RAS) on intestinal inflammation and fibrosis. DESIGN Cultured human colonic myofibroblast proliferation and collagen secretion were assessed following treatment with angiotensin (Ang) II and Ang (1-7), their receptor antagonists candesartan and A779, and the ACE inhibitor captopril. Circulating and intestinal RAS components were evaluated in patients with and without IBD. Disease outcomes in patients with IBD treated with ACE inhibitors and angiotensin receptor blockers (ARBs) were assessed in retrospective studies. RESULTS Human colonic myofibroblast proliferation was reduced by Ang (1-7) in a dose-dependent manner (p<0.05). Ang II marginally but not significantly increased proliferation, an effect reversed by candesartan (p<0.001). Colonic myofibroblast collagen secretion was reduced by Ang (1-7) (p<0.05) and captopril (p<0.001), and was increased by Ang II (p<0.001). Patients with IBD had higher circulating renin (mean 25.4 vs 18.6 mIU/L, p=0.026) and ACE2:ACE ratio (mean 0.92 vs 0.69, p=0.015) than controls without IBD. RAS gene transcripts and peptides were identified in healthy and diseased bowels. Colonic mucosal Masson's trichrome staining correlated with Ang II (r=0.346, p=0.010) and inversely with ACE2 activity (r=-0.373, p=0.006). Patients with IBD who required surgery (1/37 vs 12/75, p=0.034) and hospitalisation (0/34 vs 8/68, p=0.049) over 2 years were less often treated with ACE inhibitors and ARBs than patients not requiring surgery or hospitalisation. CONCLUSIONS The RAS mediates fibrosis in human cell cultures, is expressed in the intestine and perturbed in intestinal inflammation, and agents targeting this system are associated with improved disease outcomes.
Collapse
Affiliation(s)
- Mayur Garg
- Eastern Health Clinical School, Monash University, Box Hill, Victoria, Australia .,Gastroenterology and Hepatology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Simon G Royce
- Medicine, Monash University Central Clinical School, Melbourne, Victoria, Australia
| | - Chris Tikellis
- Diabetes, Monash University Central Clinical School, Melbourne, Victoria, Australia
| | - Claire Shallue
- Eastern Health Clinical School, Monash University, Box Hill, Victoria, Australia
| | - Duygu Batu
- Diabetes, Monash University Central Clinical School, Melbourne, Victoria, Australia
| | - Elena Velkoska
- Department of Medicine, University of Melbourne, Heidelberg, Victoria, Australia
| | - Louise M Burrell
- Department of Medicine, University of Melbourne, Heidelberg, Victoria, Australia
| | - Sheila K Patel
- Department of Medicine, University of Melbourne, Heidelberg, Victoria, Australia
| | - Lauren Beswick
- Eastern Health Clinical School, Monash University, Box Hill, Victoria, Australia
| | - Anvesh Jackson
- Eastern Health Clinical School, Monash University, Box Hill, Victoria, Australia
| | - Kaushali Britto
- Gastroenterology, Alfred Health, Melbourne, Victoria, Australia
| | - Matthew Lukies
- Eastern Health Clinical School, Monash University, Box Hill, Victoria, Australia
| | - Pavel Sluka
- Eastern Health Clinical School, Monash University, Box Hill, Victoria, Australia
| | - Hady Wardan
- Eastern Health Clinical School, Monash University, Box Hill, Victoria, Australia
| | - Yumiko Hirokawa
- Structural Biology, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Chin Wee Tan
- Structural Biology, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Maree Faux
- Structural Biology, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Antony W Burgess
- Structural Biology, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Patrick Hosking
- Pathology, Box Hill Hospital, Eastern Health, Box Hill, Victoria, Australia
| | - Shaun Monagle
- Pathology, Box Hill Hospital, Eastern Health, Box Hill, Victoria, Australia
| | - Merlin Thomas
- Diabetes, Monash University Central Clinical School, Melbourne, Victoria, Australia
| | - Peter R Gibson
- Gastroenterology, Alfred Health, Melbourne, Victoria, Australia
| | - John Lubel
- Eastern Health Clinical School, Monash University, Box Hill, Victoria, Australia
| |
Collapse
|
10
|
Bi J, Zhang J, Ren Y, Du Z, Li T, Wang T, Zhang L, Wang M, Wu Z, Lv Y, Wu R. Irisin reverses intestinal epithelial barrier dysfunction during intestinal injury via binding to the integrin αVβ5 receptor. J Cell Mol Med 2020; 24:996-1009. [PMID: 31701659 PMCID: PMC6933384 DOI: 10.1111/jcmm.14811] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 09/30/2019] [Accepted: 10/19/2019] [Indexed: 02/06/2023] Open
Abstract
Disruption of the gut barrier results in severe clinical outcomes with no specific treatment. Metabolic disorders and destruction of enterocytes play key roles in gut barrier dysfunction. Irisin is a newly identified exercise hormone that regulates energy metabolism. However, the effect of irisin on gut barrier function remains unknown. The therapeutic effect of irisin on gut barrier dysfunction was evaluated in gut ischemia reperfusion (IR). The direct effect of irisin on gut barrier function was studied in Caco-2 cells. Here, we discovered that serum and gut irisin levels were decreased during gut IR and that treatment with exogenous irisin restored gut barrier function after gut IR in mice. Meanwhile, irisin decreased oxidative stress, calcium influx and endoplasmic reticulum (ER) stress after gut IR. Moreover, irisin protected mitochondrial function and reduced enterocyte apoptosis. The neutralizing antibody against irisin significantly aggravated gut injury, oxidative stress and enterocyte apoptosis after gut IR. Further studies revealed that irisin activated the AMPK-UCP 2 pathway via binding to the integrin αVβ5 receptor. Inhibition of integrin αVβ5, AMPK or UCP 2 abolished the protective role of irisin in gut barrier function. In conclusion, exogenous irisin restores gut barrier function after gut IR via the integrin αVβ5-AMPK-UCP 2 pathway.
Collapse
Affiliation(s)
- Jianbin Bi
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative MedicineShaanxi Provincial Center for Regenerative Medicine and Surgical EngineeringInstitute of Advanced Surgical Technology and EngineeringFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
- Department of Hepatobiliary SurgeryFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
| | - Jia Zhang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative MedicineShaanxi Provincial Center for Regenerative Medicine and Surgical EngineeringInstitute of Advanced Surgical Technology and EngineeringFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
- Department of Hepatobiliary SurgeryFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
| | - Yifan Ren
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative MedicineShaanxi Provincial Center for Regenerative Medicine and Surgical EngineeringInstitute of Advanced Surgical Technology and EngineeringFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
- Department of Hepatobiliary SurgeryFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
| | - Zhaoqing Du
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative MedicineShaanxi Provincial Center for Regenerative Medicine and Surgical EngineeringInstitute of Advanced Surgical Technology and EngineeringFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
- Department of Hepatobiliary SurgeryFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
| | - Teng Li
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative MedicineShaanxi Provincial Center for Regenerative Medicine and Surgical EngineeringInstitute of Advanced Surgical Technology and EngineeringFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
- Department of Hepatobiliary SurgeryFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
| | - Tao Wang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative MedicineShaanxi Provincial Center for Regenerative Medicine and Surgical EngineeringInstitute of Advanced Surgical Technology and EngineeringFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
- Department of Hepatobiliary SurgeryFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
| | - Lin Zhang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative MedicineShaanxi Provincial Center for Regenerative Medicine and Surgical EngineeringInstitute of Advanced Surgical Technology and EngineeringFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
- Department of Hepatobiliary SurgeryFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
| | - Mengzhou Wang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative MedicineShaanxi Provincial Center for Regenerative Medicine and Surgical EngineeringInstitute of Advanced Surgical Technology and EngineeringFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
- Department of Hepatobiliary SurgeryFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
| | - Zheng Wu
- Department of Hepatobiliary SurgeryFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
| | - Yi Lv
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative MedicineShaanxi Provincial Center for Regenerative Medicine and Surgical EngineeringInstitute of Advanced Surgical Technology and EngineeringFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
- Department of Hepatobiliary SurgeryFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
| | - Rongqian Wu
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative MedicineShaanxi Provincial Center for Regenerative Medicine and Surgical EngineeringInstitute of Advanced Surgical Technology and EngineeringFirst Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
| |
Collapse
|
11
|
Schistosoma mansoni Worm Infection Regulates the Intestinal Microbiota and Susceptibility to Colitis. Infect Immun 2019; 87:IAI.00275-19. [PMID: 31138616 DOI: 10.1128/iai.00275-19] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 05/22/2019] [Indexed: 02/08/2023] Open
Abstract
Infection with parasite helminths induces potent modulation of the immune system of the host. Epidemiological and animal studies have shown that helminth infections can suppress or exacerbate unrelated autoimmune, allergic, and other inflammatory disorders. There is growing evidence that helminth infection-mediated suppression of bystander inflammatory responses is influenced by alterations in the intestinal microbiome modulating metabolic and immune functions of the infected host. We analyzed the fecal microbiota of mice infected with adult male Schistosoma mansoni worms, which are less susceptible to experimental colitis, and male- and female-worm-infected mice, which are highly sensitive to colitis. While both groups of infected mice developed a disrupted microbiota, there were marked alterations in mice with male and female worm infections. Antibiotic-treated recipients that were cohoused with both types of S. mansoni worm-infected mice acquired a colitogenic microbiome, leading to increased susceptibility to experimental colitis. Following anthelmintic treatment to remove worms from worm-only-infected mice, the mice developed exacerbated colitis. This study provides evidence that adult male S. mansoni worm infection modulates the host's immune system and suppresses bystander colitis while limiting dysbiosis of the host's intestinal microbiome during infection.
Collapse
|
12
|
Xing S, Zhang X, Liu JH, Huang X, Zhou P. Host MyD88 signaling protects against acute graft-versus-host disease after allogeneic bone marrow transplantation. Clin Exp Immunol 2018; 195:121-131. [PMID: 30317551 DOI: 10.1111/cei.13215] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 08/05/2018] [Accepted: 09/02/2018] [Indexed: 11/26/2022] Open
Abstract
Recent experimental strategies to reduce graft-versus-host disease (GVHD) have focused largely on modifying innate immunity. Toll-like receptor (TLR)-driven myeloid differentiation primary response 88 (MyD88)-dependent signalling pathways that initiate adaptive immune function are also critical for the pathogenesis of GVHD. This study aimed to delineate the role of host MyD88 in the development of acute GVHD following fully major histocompatibility complex-mismatched allogeneic bone marrow transplantation (BMT). When myeloablated BALB/c MyD88 knock-out recipients were transplanted with C57BL/6 (B6) donor cells, they developed significantly more severe GVHD than wild-type (WT) BALB/c hosts. The increased morbidity and mortality in MyD88-/- mice correlated with increased serum levels of lipopolysaccharide and elevated inflammatory cytokines in GVHD target organs. Additionally, MyD88 deficiency in BMT recipients led to increased donor T cell expansion and more donor CD11c+ cell intestinal infiltration with apoptotic cells but reduced proliferation of intestinal epithelial cells compared with that in WT BMT recipients. Decreased expression of tight junction mRNA in epithelial cells of MyD88-/- mice suggested that MyD88 contributes to intestinal integrity. Cox-2 expression in the GVHD-targeted organs of WT mice is increased upon GVHD induction, but this enhanced expression was obviously inhibited by MyD88 deficiency. The present findings demonstrate an unexpected role for host MyD88 in preventing GVHD after allogeneic BMT.
Collapse
Affiliation(s)
- S Xing
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology,Key Laboratory of Organ Transplantation, Ministry of Health and Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China
| | - X Zhang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology,Key Laboratory of Organ Transplantation, Ministry of Health and Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Department of Breast Surgery, Renmin hospital of Wuhan University, Wuhan University, Wuhan, China
| | - J H Liu
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology,Key Laboratory of Organ Transplantation, Ministry of Health and Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China
| | - X Huang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology,Key Laboratory of Organ Transplantation, Ministry of Health and Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China
| | - P Zhou
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology,Key Laboratory of Organ Transplantation, Ministry of Health and Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China
| |
Collapse
|
13
|
Jevnikar Z, Östling J, Ax E, Calvén J, Thörn K, Israelsson E, Öberg L, Singhania A, Lau LCK, Wilson SJ, Ward JA, Chauhan A, Sousa AR, De Meulder B, Loza MJ, Baribaud F, Sterk PJ, Chung KF, Sun K, Guo Y, Adcock IM, Payne D, Dahlen B, Chanez P, Shaw DE, Krug N, Hohlfeld JM, Sandström T, Djukanovic R, James A, Hinks TSC, Howarth PH, Vaarala O, van Geest M, Olsson H. Epithelial IL-6 trans-signaling defines a new asthma phenotype with increased airway inflammation. J Allergy Clin Immunol 2018; 143:577-590. [PMID: 29902480 DOI: 10.1016/j.jaci.2018.05.026] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 04/15/2018] [Accepted: 05/04/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Although several studies link high levels of IL-6 and soluble IL-6 receptor (sIL-6R) to asthma severity and decreased lung function, the role of IL-6 trans-signaling (IL-6TS) in asthmatic patients is unclear. OBJECTIVE We sought to explore the association between epithelial IL-6TS pathway activation and molecular and clinical phenotypes in asthmatic patients. METHODS An IL-6TS gene signature obtained from air-liquid interface cultures of human bronchial epithelial cells stimulated with IL-6 and sIL-6R was used to stratify lung epithelial transcriptomic data (Unbiased Biomarkers in Prediction of Respiratory Disease Outcomes [U-BIOPRED] cohorts) by means of hierarchical clustering. IL-6TS-specific protein markers were used to stratify sputum biomarker data (Wessex cohort). Molecular phenotyping was based on transcriptional profiling of epithelial brushings, pathway analysis, and immunohistochemical analysis of bronchial biopsy specimens. RESULTS Activation of IL-6TS in air-liquid interface cultures reduced epithelial integrity and induced a specific gene signature enriched in genes associated with airway remodeling. The IL-6TS signature identified a subset of patients with IL-6TS-high asthma with increased epithelial expression of IL-6TS-inducible genes in the absence of systemic inflammation. The IL-6TS-high subset had an overrepresentation of frequent exacerbators, blood eosinophilia, and submucosal infiltration of T cells and macrophages. In bronchial brushings Toll-like receptor pathway genes were upregulated, whereas expression of cell junction genes was reduced. Sputum sIL-6R and IL-6 levels correlated with sputum markers of remodeling and innate immune activation, in particular YKL-40, matrix metalloproteinase 3, macrophage inflammatory protein 1β, IL-8, and IL-1β. CONCLUSIONS Local lung epithelial IL-6TS activation in the absence of type 2 airway inflammation defines a novel subset of asthmatic patients and might drive airway inflammation and epithelial dysfunction in these patients.
Collapse
Affiliation(s)
- Zala Jevnikar
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden.
| | - Jörgen Östling
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Elisabeth Ax
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden; Department of Internal Medicine and Clinical Nutrition, Krefting Research Centre, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Jenny Calvén
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Kristofer Thörn
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Elisabeth Israelsson
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Lisa Öberg
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Akul Singhania
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton University Hospital, Southampton, United Kingdom
| | - Laurie C K Lau
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton University Hospital, Southampton, United Kingdom
| | - Susan J Wilson
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton University Hospital, Southampton, United Kingdom; Histochemistry Research Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Jonathan A Ward
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton University Hospital, Southampton, United Kingdom; Histochemistry Research Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Anoop Chauhan
- Portsmouth Hospitals NHS Trust, Portsmouth, United Kingdom
| | - Ana R Sousa
- Discovery Medicine, GlaxoSmithKline, Brentford, United Kingdom
| | - Bertrand De Meulder
- European Institute for Systems Biology and Medicine, CIRI UMR5308, CNRS-ENS-UCBL-INSERM, Université de Lyon, Lyon, France
| | | | | | - Peter J Sterk
- Department of Respiratory Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London UK & Royal Brompton Biomedical Research Unit at Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, United Kingdom
| | - Kai Sun
- Department of Computing & Data Science Institute, Imperial College London, London, United Kingdom
| | - Yike Guo
- Department of Computing & Data Science Institute, Imperial College London, London, United Kingdom
| | - Ian M Adcock
- National Heart and Lung Institute, Imperial College London, London UK & Royal Brompton Biomedical Research Unit at Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, United Kingdom
| | - Debbie Payne
- Centre for Integrated Genomic Medical Research, University of Manchester, Manchester, United Kingdom
| | - Barbro Dahlen
- Karolinska University Hospital & Centre for Allergy Research, Karolinska Institute, Stockholm, Sweden
| | | | - Dominick E Shaw
- Respiratory Biomedical Research Unit, University of Nottingham, Nottingham, United Kingdom
| | - Norbert Krug
- Fraunhofer Institute of Toxicology and Experimental Medicine, Member of the German Center for Lung Research, Hannover, Germany
| | - Jens M Hohlfeld
- Fraunhofer Institute of Toxicology and Experimental Medicine, Member of the German Center for Lung Research, Hannover, Germany; Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
| | - Thomas Sandström
- Department of Public Health and Clinical Medicine, Medicine, Umeå University, Umeå, Sweden
| | - Ratko Djukanovic
- NIHR Southampton Respiratory Biomedical Research Unit, Southampton University Hospital, Southampton, United Kingdom
| | - Anna James
- Experimental Asthma and Allergy Research, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Timothy S C Hinks
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton University Hospital, Southampton, United Kingdom; NIHR Southampton Respiratory Biomedical Research Unit, Southampton University Hospital, Southampton, United Kingdom; Respiratory Medicine Unit, NDM Experimental Medicine, University of OxfordJohn Radcliffe Hospital, Oxford, United Kingdom
| | - Peter H Howarth
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton University Hospital, Southampton, United Kingdom; NIHR Southampton Respiratory Biomedical Research Unit, Southampton University Hospital, Southampton, United Kingdom
| | - Outi Vaarala
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Marleen van Geest
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Henric Olsson
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | | |
Collapse
|
14
|
Paveljšek D, Juvan P, Košir R, Rozman D, Hacin B, Ivičak-Kocjan K, Rogelj I. Lactobacillus fermentum L930BB and Bifidobacterium animalis subsp. animalis IM386 initiate signalling pathways involved in intestinal epithelial barrier protection. Benef Microbes 2018; 9:515-525. [PMID: 29633647 DOI: 10.3920/bm2017.0107] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The manipulation of intestinal microbiota with beneficial microbes represents a promising alternative or adjunct therapy in gastrointestinal disorders and inflammation. The current study aims to clarify the signalling pathways and evaluate the possible beneficial effects of the combination of two strains. We used a dextran sulphate sodium (DSS)-induced mouse model of colitis. RNA extracted from the middle part of the colon tissue was used for examination of the global gene expression with Affymetrix microarrays. An enrichment analysis of the KEGG pathways was performed, and a subset of genes associated with intestinal epithelial barrier function was verified with qPCR. A clinical condition assessment of the differently treated mice revealed that the combination of these two bacterial strains was safe for use as a dietary supplement. All animals treated with DSS had affected colons and suffered weight loss. There were very small differences between the diseased groups, although the depth of inflammation was lower when cyclosporine A or the strain mixture was used. We discovered that the prophylactic administration of the Lactobacillus fermentum L930BB (L930BB) and Bifidobacterium animalis subsp. animalis IM386 (IM386) strains led to an anti-apoptotic pathway through phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/Akt and to the activation of pathways involved in the regulation of actin cytoskeleton via protein kinase C and GTPases. Reorganisation of actin cytoskeleton and decreased apoptosis are both helpful in intestinal epithelial cell reconstitution. We confirm important previous observations, showing that these pathways are downstream targets of Toll-like receptor 2 and fibroblast growth factor initiated signalling. Taken together, these results suggest that the combination of L930BB and IM386 could aid in the regeneration of the intestinal epithelium during pathogenesis via pattern recognition receptors and the stimulation of growth factor synthesis.
Collapse
Affiliation(s)
- D Paveljšek
- 1 Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Groblje 3, 1230 Domžale, Slovenia
| | - P Juvan
- 2 Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
| | - R Košir
- 2 Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia.,3 BIA Separations CRO, Labena d.o.o., Verovškova 64, 1000 Ljubljana, Slovenia
| | - D Rozman
- 2 Centre for Functional Genomics and Bio-Chips, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
| | - B Hacin
- 4 National Veterinary Institute, Veterinary Faculty, University of Ljubljana, Pri Hrastu 18, 5000 Nova Gorica, Slovenia
| | - K Ivičak-Kocjan
- 5 Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova ulica 19, 1000 Ljubljana, Slovenia
| | - I Rogelj
- 1 Department of Animal Science, Biotechnical Faculty, University of Ljubljana, Groblje 3, 1230 Domžale, Slovenia
| |
Collapse
|
15
|
Chelakkot C, Choi Y, Kim DK, Park HT, Ghim J, Kwon Y, Jeon J, Kim MS, Jee YK, Gho YS, Park HS, Kim YK, Ryu SH. Akkermansia muciniphila-derived extracellular vesicles influence gut permeability through the regulation of tight junctions. Exp Mol Med 2018; 50:e450. [PMID: 29472701 PMCID: PMC5903829 DOI: 10.1038/emm.2017.282] [Citation(s) in RCA: 526] [Impact Index Per Article: 75.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 09/09/2017] [Accepted: 09/11/2017] [Indexed: 12/18/2022] Open
Abstract
The gut microbiota has an important role in the gut barrier, inflammation and metabolic functions. Studies have identified a close association between the intestinal barrier and metabolic diseases, including obesity and type 2 diabetes (T2D). Recently, Akkermansia muciniphila has been reported as a beneficial bacterium that reduces gut barrier disruption and insulin resistance. Here we evaluated the role of A. muciniphila-derived extracellular vesicles (AmEVs) in the regulation of gut permeability. We found that there are more AmEVs in the fecal samples of healthy controls compared with those of patients with T2D. In addition, AmEV administration enhanced tight junction function, reduced body weight gain and improved glucose tolerance in high-fat diet (HFD)-induced diabetic mice. To test the direct effect of AmEVs on human epithelial cells, cultured Caco-2 cells were treated with these vesicles. AmEVs decreased the gut permeability of lipopolysaccharide-treated Caco-2 cells, whereas Escherichia coli-derived EVs had no significant effect. Interestingly, the expression of occludin was increased by AmEV treatment. Overall, these results imply that AmEVs may act as a functional moiety for controlling gut permeability and that the regulation of intestinal barrier integrity can improve metabolic functions in HFD-fed mice.
Collapse
Affiliation(s)
- Chaithanya Chelakkot
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Youngwoo Choi
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Dae-Kyum Kim
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Hyun T Park
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Jaewang Ghim
- NovaCell Technology Inc., Pohang, Republic of Korea
| | - Yonghoon Kwon
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Jinseong Jeon
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Min-Seon Kim
- Asan Institute of Life Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young-Koo Jee
- Department of Internal Medicine, Dankook University College of Medicine, Cheonan, Republic of Korea
| | - Yong S Gho
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon-si, Republic of Korea
| | | | - Sung H Ryu
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Republic of Korea.,Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| |
Collapse
|
16
|
Yu SX, Chen W, Liu ZZ, Zhou FH, Yan SQ, Hu GQ, Qin XX, Zhang J, Ma K, Du CT, Gu JM, Deng XM, Han WY, Yang YJ. Non-Hematopoietic MLKL Protects Against Salmonella Mucosal Infection by Enhancing Inflammasome Activation. Front Immunol 2018; 9:119. [PMID: 29456533 PMCID: PMC5801401 DOI: 10.3389/fimmu.2018.00119] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/15/2018] [Indexed: 12/21/2022] Open
Abstract
The intestinal mucosal barrier is critical for host defense against pathogens infection. Here, we demonstrate that the mixed lineage kinase-like protein (MLKL), a necroptosis effector, promotes intestinal epithelial barrier function by enhancing inflammasome activation. MLKL−/− mice were more susceptible to Salmonella infection compared with wild-type counterparts, with higher mortality rates, increased body weight loss, exacerbated intestinal inflammation, more bacterial colonization, and severe epithelial barrier disruption. MLKL deficiency promoted early epithelial colonization of Salmonella prior to developing apparent intestinal pathology. Active MLKL was predominantly expressed in crypt epithelial cells, and experiments using bone marrow chimeras found that the protective effects of MLKL were dependent on its expression in non-hematopoietic cells. Intestinal mucosa of MLKL−/− mice had impaired caspase-1 and gasdermin D cleavages and decreased interleukin (IL)-18 release. Moreover, administration of exogenous recombinant IL-18 rescued the phenotype of increased bacterial colonization in MLKL−/− mice. Thus, our results uncover the role of MLKL in enhancing inflammasome activation in intestinal epithelial cells to inhibit early bacterial colonization.
Collapse
Affiliation(s)
- Shui-Xing Yu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Wei Chen
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zhen-Zhen Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Feng-Hua Zhou
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Shi-Qing Yan
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Gui-Qiu Hu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiao-Xia Qin
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jie Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ke Ma
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Chong-Tao Du
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jing-Min Gu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xu-Ming Deng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Wen-Yu Han
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yong-Jun Yang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
17
|
Hughes KR, Harnisch LC, Alcon-Giner C, Mitra S, Wright CJ, Ketskemety J, van Sinderen D, Watson AJM, Hall LJ. Bifidobacterium breve reduces apoptotic epithelial cell shedding in an exopolysaccharide and MyD88-dependent manner. Open Biol 2017; 7:rsob.160155. [PMID: 28123052 PMCID: PMC5303268 DOI: 10.1098/rsob.160155] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 12/15/2016] [Indexed: 12/14/2022] Open
Abstract
Certain members of the microbiota genus Bifidobacterium are known to positively influence host well-being. Importantly, reduced bifidobacterial levels are associated with inflammatory bowel disease (IBD) patients, who also have impaired epithelial barrier function, including elevated rates of apoptotic extrusion of small intestinal epithelial cells (IECs) from villi—a process termed ‘cell shedding’. Using a mouse model of pathological cell shedding, we show that mice receiving Bifidobacterium breve UCC2003 exhibit significantly reduced rates of small IEC shedding. Bifidobacterial-induced protection appears to be mediated by a specific bifidobacterial surface exopolysaccharide and interactions with host MyD88 resulting in downregulation of intrinsic and extrinsic apoptotic responses to protect epithelial cells under highly inflammatory conditions. Our results reveal an important and previously undescribed role for B. breve, in positively modulating epithelial cell shedding outcomes via bacterial- and host-dependent factors, supporting the notion that manipulation of the microbiota affects intestinal disease outcomes.
Collapse
Affiliation(s)
- K R Hughes
- The Gut Health and Food Safety Programme, Institute of Food Research, Norwich Research Park, Colney, Norwich NR4 7UA, UK .,Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK
| | - L C Harnisch
- The Gut Health and Food Safety Programme, Institute of Food Research, Norwich Research Park, Colney, Norwich NR4 7UA, UK.,Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK
| | - C Alcon-Giner
- The Gut Health and Food Safety Programme, Institute of Food Research, Norwich Research Park, Colney, Norwich NR4 7UA, UK
| | - S Mitra
- The Gut Health and Food Safety Programme, Institute of Food Research, Norwich Research Park, Colney, Norwich NR4 7UA, UK
| | - C J Wright
- The Gut Health and Food Safety Programme, Institute of Food Research, Norwich Research Park, Colney, Norwich NR4 7UA, UK.,Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK
| | - J Ketskemety
- The Gut Health and Food Safety Programme, Institute of Food Research, Norwich Research Park, Colney, Norwich NR4 7UA, UK
| | - D van Sinderen
- APC Microbiome Institute, University College Cork, Cork, Ireland.,School of Microbiology, University College Cork, Cork, Ireland
| | - A J M Watson
- The Gut Health and Food Safety Programme, Institute of Food Research, Norwich Research Park, Colney, Norwich NR4 7UA, UK .,Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, UK
| | - L J Hall
- The Gut Health and Food Safety Programme, Institute of Food Research, Norwich Research Park, Colney, Norwich NR4 7UA, UK
| |
Collapse
|
18
|
Duijvis NW, Moerland PD, Kunne C, Slaman MMW, van Dooren FH, Vogels EW, de Jonge WJ, Meijer SL, Fluiter K, te Velde AA. Inhibition of miR-142-5P ameliorates disease in mouse models of experimental colitis. PLoS One 2017; 12:e0185097. [PMID: 29059189 PMCID: PMC5653202 DOI: 10.1371/journal.pone.0185097] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 09/06/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are epigenetically involved in regulating gene expression. They may be of importance in the pathogenesis of inflammatory bowel disease (IBD). The aim of this study was to determine the role of miRNAs by their specific blocking in the CD4+CB45RBhi T-cell transfer model of chronic experimental colitis. METHODS Colitis caused by transfer of WT CD4+CD45RBhi T cells in severe combined immunodeficiency (SCID) mice shares many features with human IBD. Colonic miRNA expression levels were measured at three time points in colitic mice, where a time-dependent upregulation of multiple miRNAs was seen. To inhibit these miRNAs, specific locked-nucleic-acid-modified (LNA) oligonucleotides were administered in further experiments at the moment the mice demonstrated the first signs of colitis. As controls, PBS and a scrambled sequence of anti-miRNA were used. Genome-wide expression analyses were also performed in order to detect candidate target genes of miR-142-5p, of which inhibition resulted in most effective amelioration of colitis. RESULTS Anti-miR-142-5p reduced colitis and related wasting disease when administered in the T-cell transfer model, reflected in reduced weight loss and a lower disease activity index (DAI). In further validation experiments we also observed a higher survival rate and less colonic histological inflammation in the antagomir-treated mice. Moreover, by genome-wide expression analyses, we found downstream activation of the anti-inflammatory IL10RA pathway, including three genes also found in the top-20 candidate target genes of miR-142-5p. CONCLUSION In conclusion, CD4+CD45RBhi-transfer colitis induces miR-142-5p. Blocking miR-142-5p reduced colitis and prevented wasting disease, possibly by activation of the IL10RA pathway.
Collapse
Affiliation(s)
- Nicolette W. Duijvis
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), Amsterdam, the Netherlands
- * E-mail: (NWD); (AAV)
| | - Perry D. Moerland
- Bioinformatics Laboratory, Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Academic Medical Center, Amsterdam, the Netherlands
| | - Cindy Kunne
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), Amsterdam, the Netherlands
| | - Monique M. W. Slaman
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), Amsterdam, the Netherlands
| | - Faas H. van Dooren
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), Amsterdam, the Netherlands
| | - Esther W. Vogels
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), Amsterdam, the Netherlands
| | - Wouter J. de Jonge
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), Amsterdam, the Netherlands
| | - Sybren L. Meijer
- Department of Pathology, Academic Medical Center (AMC), Amsterdam, the Netherlands
| | - Kees Fluiter
- Department of Clinical Genetics, Academic Medical Center (AMC), Amsterdam, the Netherlands
| | - Anje A. te Velde
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center (AMC), Amsterdam, the Netherlands
- * E-mail: (NWD); (AAV)
| |
Collapse
|
19
|
Shao YX, Lei Z, Wolf PG, Gao Y, Guo YM, Zhang BK. Zinc Supplementation, via GPR39, Upregulates PKCζ to Protect Intestinal Barrier Integrity in Caco-2 Cells Challenged bySalmonella entericaSerovar Typhimurium. J Nutr 2017; 147:1282-1289. [DOI: 10.3945/jn.116.243238] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 11/23/2016] [Accepted: 04/13/2017] [Indexed: 12/26/2022] Open
Affiliation(s)
- Yu-Xin Shao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhao Lei
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Patricia G Wolf
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL
| | - Yan Gao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yu-Ming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Bing-Kun Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
20
|
Intestinal Epithelial Cell-Specific Deletion of PLD2 Alleviates DSS-Induced Colitis by Regulating Occludin. Sci Rep 2017; 7:1573. [PMID: 28484281 PMCID: PMC5431506 DOI: 10.1038/s41598-017-01797-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 04/04/2017] [Indexed: 02/08/2023] Open
Abstract
Ulcerative colitis is a multi-factorial disease involving a dysregulated immune response. Disruptions to the intestinal epithelial barrier and translocation of bacteria, resulting in inflammation, are common in colitis. The mechanisms underlying epithelial barrier dysfunction or regulation of tight junction proteins during disease progression of colitis have not been clearly elucidated. Increase in phospholipase D (PLD) activity is associated with disease severity in colitis animal models. However, the role of PLD2 in the maintenance of intestinal barrier integrity remains elusive. We have generated intestinal-specific Pld2 knockout mice (Pld2 IEC-KO) to investigate the mechanism of intestinal epithelial PLD2 in colitis. We show that the knockout of Pld2 confers protection against dextran sodium sulphate (DSS)-induced colitis in mice. Treatment with DSS induced the expression of PLD2 and downregulated occludin in colon epithelial cells. PLD2 was shown to mediate phosphorylation of occludin and induce its proteasomal degradation in a c-Src kinase-dependent pathway. Additionally, we have shown that treatment with an inhibitor of PLD2 can rescue mice from DSS-induced colitis. To our knowledge, this is the first report showing that PLD2 is pivotal in the regulation of the integrity of epithelial tight junctions and occludin turn over, thereby implicating it in the pathogenesis of colitis.
Collapse
|
21
|
Hu GQ, Song PX, Li N, Chen W, Lei QQ, Yu SX, Zhang XJ, Du CT, Deng XM, Han WY, Yang YJ. AIM2 contributes to the maintenance of intestinal integrity via Akt and protects against Salmonella mucosal infection. Mucosal Immunol 2016; 9:1330-9. [PMID: 26838050 DOI: 10.1038/mi.2015.142] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 11/16/2015] [Indexed: 02/04/2023]
Abstract
The mechanism regulating the gastrointestinal epithelial barrier remains poorly understood. We herein demonstrate that Absent in melanoma-2 (AIM2) contributes to the maintenance of intestinal barrier integrity and defense against bacterial infection. AIM2-deficient mice displayed an increased susceptibility to mucosal but not systemic infection by Salmonella typhimurium, indicating a protective role for AIM2 in the gastrointestinal tract. In a Salmonella colitis model, compared with wild-type mice, AIM2(-/-) mice exhibited more severe body weight loss, intestinal damage, intestinal inflammation, and disruption of basal and activated epithelial cell turnover. In vivo and in vitro data showed that AIM2 restricted the early epithelial paracellular invasion of Salmonella and decreased epithelial permeability. The decreased epithelial barrier in AIM2(-/-) mice might be attributed to the altered expression of tight junction proteins that contribute to epithelial integrity. AIM2 promoted the expression of tight junction proteins through Akt activation. Together, these results suggest that AIM2 is required for maintaining the integrity of the epithelial barrier.
Collapse
Affiliation(s)
- G-Q Hu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine & The First Hospital, Jilin University, Changchun, China
| | - P-X Song
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine & The First Hospital, Jilin University, Changchun, China
| | - N Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine & The First Hospital, Jilin University, Changchun, China
| | - W Chen
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine & The First Hospital, Jilin University, Changchun, China
| | - Q-Q Lei
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine & The First Hospital, Jilin University, Changchun, China
| | - S-X Yu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine & The First Hospital, Jilin University, Changchun, China
| | - X-J Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine & The First Hospital, Jilin University, Changchun, China
| | - C-T Du
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine & The First Hospital, Jilin University, Changchun, China
| | - X-M Deng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine & The First Hospital, Jilin University, Changchun, China
| | - W-Y Han
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine & The First Hospital, Jilin University, Changchun, China
| | - Y-J Yang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine & The First Hospital, Jilin University, Changchun, China
| |
Collapse
|
22
|
Gu MJ, Song SK, Lee IK, Ko S, Han SE, Bae S, Ji SY, Park BC, Song KD, Lee HK, Han SH, Yun CH. Barrier protection via Toll-like receptor 2 signaling in porcine intestinal epithelial cells damaged by deoxynivalnol. Vet Res 2016; 47:25. [PMID: 26857454 PMCID: PMC4746821 DOI: 10.1186/s13567-016-0309-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 01/05/2016] [Indexed: 12/29/2022] Open
Abstract
Intestinal barrier is the first line of defense inside the body and comprises intercellular tight junction (TJ) proteins that regulate paracellular permeability. Deoxynivalenol (DON), a fungal metabolite often found in the contaminated food of domestic animals, is known to impair intestinal barrier function and may be involved in intestinal inflammation. Unlike in humans and mice, the importance of Toll-like receptor (TLR) 2 expressed in porcine intestinal epithelial cells is largely unclear. Therefore, the aim of the present study was to investigate whether TLR2 stimulation enhances intestinal barrier function and protects against DON exposure. We found that the cells treated with TLR2 ligands decreased the epithelial barrier permeability and enhanced TJ protein expression in intestinal porcine epithelial cells (IPEC-J2). In addition, pretreatment with TLR2 ligand, including Pam3CSK4 (PCSK) and lipoteichoic acid from Bacillus subtilis, prevented DON-induced barrier dysfunction by increasing the expression of TJ proteins via the PI3K-Akt-dependent pathway. It is likely that the DON-disrupted intestinal barrier caused biological changes of immune cells in the lamina propria. Thus, we conducted co-culture of differentiated IPEC-J2 cells in the upper well together with peripheral blood mononuclear cells in the bottom well and found that apical TLR2 stimulation of IPEC-J2 cells could alleviate the reduction in cell survival and proliferation of immune cells. Conclusively, TLR2 signaling on intestinal epithelial cells may enhance intestinal barrier function and prevent DON-induced barrier dysfunction of epithelial cells.
Collapse
Affiliation(s)
- Min Jeong Gu
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921, Republic of Korea. .,Biomodulation Major and Center for Food Bioconvergence, Seoul National University, Seoul, 151-921, Republic of Korea.
| | - Sun Kwang Song
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921, Republic of Korea.
| | - In Kyu Lee
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921, Republic of Korea.
| | - Seongyeol Ko
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921, Republic of Korea. .,Biomodulation Major and Center for Food Bioconvergence, Seoul National University, Seoul, 151-921, Republic of Korea.
| | - Seung Eun Han
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921, Republic of Korea. .,Biomin Korea Ltd., Seoul, 153-714, Republic of Korea.
| | - Suhan Bae
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921, Republic of Korea. .,Seoulfeed Co., Ltd., Incheon, 405-819, Republic of Korea.
| | - Sang Yun Ji
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921, Republic of Korea. .,National Institute of Animal Science, Rural Development Administration, Jeonju, 565-851, Republic of Korea.
| | - Byung-Chul Park
- Institute of Green Bio Science Technology, Seoul National University, Pyeongchang, 232-916, Republic of Korea.
| | - Ki-Duk Song
- Department of Animal Biotechnology, Chonbuk National University, Jeonju, 561-756, Republic of Korea.
| | - Hak-Kyo Lee
- Department of Animal Biotechnology, Chonbuk National University, Jeonju, 561-756, Republic of Korea.
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, School of Dentistry, Seoul National University, Seoul, 110-749, Republic of Korea.
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921, Republic of Korea. .,Biomodulation Major and Center for Food Bioconvergence, Seoul National University, Seoul, 151-921, Republic of Korea. .,Institute of Green Bio Science Technology, Seoul National University, Pyeongchang, 232-916, Republic of Korea.
| |
Collapse
|
23
|
Johnston DGW, Corr SC. Toll-Like Receptor Signalling and the Control of Intestinal Barrier Function. Methods Mol Biol 2016; 1390:287-300. [PMID: 26803636 DOI: 10.1007/978-1-4939-3335-8_18] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Epithelial barrier function and innate immunity are fundamental to the pathogenesis of inflammatory and infectious disease. Along with plasma membranes, epithelial cells are the primary cellular determinant of epithelial barrier function. The mechanism by which polarized epithelia form a permeability barrier is of fundamental importance to the prevention of many infectious and inflammatory diseases. Moreover, epithelial cells express Toll-like receptors (TLRs) which upon recognition of conserved microbial factors such as lipopolysaccharide (LPS) induce epithelial responses including epithelial cell proliferation, secretion of secretory IgA into the lumen and production mucins and antimicrobial peptides, thereby promoting intestinal barrier function. Understanding gut barrier integrity and regulation of permeability is crucial to increase our understanding of the pathogenesis of intestinal disease. A variety of tests have been developed to assess this barrier, including assessing intestinal epithelial cell proliferation or death, intestinal tight junction status and the consequence of intestinal barrier integrity loss such as increased intestinal permeability and susceptibility to bacterial infection. Using a mouse model, this chapter describes some of the methods to assess the functional integrity of this epithelial barrier and the part played by a TLR signalling pathway.
Collapse
Affiliation(s)
- Daniel G W Johnston
- Trinity Biomedical Sciences Institute, School of Biochemistry and Immunology, Trinity College Dublin, Pearse Street, Dublin 2, Ireland
| | - Sinéad C Corr
- Trinity Biomedical Sciences Institute, School of Biochemistry and Immunology, Trinity College Dublin, Pearse Street, Dublin 2, Ireland.
| |
Collapse
|
24
|
A critical role for the TLR signaling adapter Mal in alveolar macrophage-mediated protection against Bordetella pertussis. Mucosal Immunol 2015; 8:982-92. [PMID: 25515629 DOI: 10.1038/mi.2014.125] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 11/06/2014] [Indexed: 02/07/2023]
Abstract
Bordetella pertussis causes whooping cough, an infectious disease of the respiratory tract that is re-emerging despite high vaccine coverage. Here we examined the role of Toll-like receptor (TLR) adapter protein Mal in the control of B. pertussis infection in the lungs. We found that B. pertussis bacterial load in the lungs of Mal-defective (Mal(-/-)) mice exceeded that of wild-type (WT) mice by up to 100-fold and bacteria disseminated to the liver in Mal(-/-) mice and 50% of these mice died from the infection. Macrophages from Mal(-/-) mice were defective in an early burst of pro-inflammatory cytokine production and in their ability to kill or constrain intracellular growth of B. pertussis. Importantly, the B. pertussis bacterial load in the lungs inversely correlated with the number of alveolar macrophages. Despite the maintenance and expansion of other cell populations, alveolar macrophages were completely depleted from the lungs of infected Mal(-/-) mice, but not from infected WT mice. Our findings define for the first time a role for a microbial pattern-recognition pathway in the survival of alveolar macrophages and uncover a mechanism of macrophage-mediated immunity to B. pertussis in which Mal controls intracellular survival and dissemination of bacteria from the lungs.
Collapse
|
25
|
Zhang W, Zhu YH, Yang JC, Yang GY, Zhou D, Wang JF. A Selected Lactobacillus rhamnosus Strain Promotes EGFR-Independent Akt Activation in an Enterotoxigenic Escherichia coli K88-Infected IPEC-J2 Cell Model. PLoS One 2015; 10:e0125717. [PMID: 25915861 PMCID: PMC4411159 DOI: 10.1371/journal.pone.0125717] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 03/17/2015] [Indexed: 12/24/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) are important intestinal pathogens that cause diarrhea in humans and animals. Although probiotic bacteria may protect against ETEC-induced enteric infections, the underlying mechanisms are unknown. In this study, porcine intestinal epithelial J2 cells (IPEC-J2) were pre-incubated with and without Lactobacillus rhamnosus ATCC 7469 and then exposed to F4+ ETEC. Increases in TLR4 and NOD2 mRNA expression were observed at 3 h after F4+ ETEC challenge, but these increases were attenuated by L. rhamnosus treatment. Expression of TLR2 and NOD1 mRNA was up-regulated in cells pre-treated with L. rhamnosus. Pre-treatment with L. rhamnosus counteracted F4+ ETEC-induced increases in TNF-α concentration. Increased PGE2. concentrations were observed in cells infected with F4+ ETEC and in cells treated with L. rhamnosus only. A decrease in phosphorylated epidermal growth factor receptor (EGFR) was observed at 3 h after F4+ ETEC challenge in cells treated with L. rhamnosus. Pre-treatment with L. rhamnosus enhanced Akt phosphorylation and increased ZO-1 and occludin protein expression. Our findings suggest that L. rhamnosus protects intestinal epithelial cells from F4+ ETEC-induced damage, partly through the anti-inflammatory response involving synergism between TLR2 and NOD1. In addition, L. rhamnosus promotes EGFR-independent Akt activation, which may activate intestinal epithelial cells in response to bacterial infection, in turn increasing tight junction integrity and thus enhancing the barrier function and restricting pathogen invasion. Pre-incubation with L. rhamnosus was superior to co-incubation in reducing the adhesion of F4+ ETEC to IPEC-J2 cells and subsequently attenuating F4+ ETEC-induced mucin layer destruction and suppressing apoptosis. Our data indicate that a selected L. rhamnosus strain interacts with porcine intestinal epithelial cells to maintain the epithelial barrier and promote intestinal epithelial cell activation in response to bacterial infection, thus protecting cells from the deleterious effects of F4+ ETEC.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yao-Hong Zhu
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jin-Cai Yang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Gui-Yan Yang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Dong Zhou
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jiu-Feng Wang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
26
|
Yang L, Yan Y. Protein kinases are potential targets to treat inflammatory bowel disease. World J Gastrointest Pharmacol Ther 2014; 5:209-217. [PMID: 25374761 PMCID: PMC4218950 DOI: 10.4292/wjgpt.v5.i4.209] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Revised: 04/05/2014] [Accepted: 07/29/2014] [Indexed: 02/06/2023] Open
Abstract
Protein kinases play a crucial role in the pathogenesis of inflammatory bowel disease (IBD), the two main forms of which are ulcerative colitis and Crohn’s disease. In this article, we will review the mechanisms of involvement of protein kinases in the pathogenesis of and intervention against IBD, in terms of their effects on genetics, microbiota, mucous layer and tight junction, and the potential of protein kinases as therapeutic targets against IBD.
Collapse
|
27
|
Epithelial barrier function: at the front line of asthma immunology and allergic airway inflammation. J Allergy Clin Immunol 2014; 134:509-20. [PMID: 25085341 DOI: 10.1016/j.jaci.2014.05.049] [Citation(s) in RCA: 333] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 05/30/2014] [Accepted: 05/30/2014] [Indexed: 02/08/2023]
Abstract
Airway epithelial cells form a barrier to the outside world and are at the front line of mucosal immunity. Epithelial apical junctional complexes are multiprotein subunits that promote cell-cell adhesion and barrier integrity. Recent studies in the skin and gastrointestinal tract suggest that disruption of cell-cell junctions is required to initiate epithelial immune responses, but how this applies to mucosal immunity in the lung is not clear. Increasing evidence indicates that defective epithelial barrier function is a feature of airway inflammation in asthmatic patients. One challenge in this area is that barrier function and junctional integrity are difficult to study in the intact lung, but innovative approaches should provide new knowledge in this area in the near future. In this article we review the structure and function of epithelial apical junctional complexes, emphasizing how regulation of the epithelial barrier affects innate and adaptive immunity. We discuss why defective epithelial barrier function might be linked to TH2 polarization in asthmatic patients and propose a rheostat model of barrier dysfunction that implicates the size of inhaled allergen particles as an important factor influencing adaptive immunity.
Collapse
|
28
|
Aviello G, Corr SC, Johnston DGW, O'Neill LAJ, Fallon PG. MyD88 adaptor-like (Mal) regulates intestinal homeostasis and colitis-associated colorectal cancer in mice. Am J Physiol Gastrointest Liver Physiol 2014; 306:G769-78. [PMID: 24603458 DOI: 10.1152/ajpgi.00399.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Toll-like receptors (TLRs) play a central role in the recognition and response to microbial pathogens and in the maintenance and function of the epithelial barrier integrity in the gut. The protein MyD88 adaptor-like (Mal/TIRAP) serves as a bridge between TLR2/TLR4- and MyD88-mediated signaling to orchestrate downstream inflammatory responses. Whereas MyD88 has an essential function in the maintenance of intestinal homeostasis, a role for Mal in this context is less well described. Colitis was induced in wild-type (WT) and Mal-deficient (Mal(-/-)) mice by administration of dextran sodium sulfate (DSS). Colitis-associated cancer was induced by DSS and azoxymethane (AOM) treatment. Chimeric mice were generated by total body gamma irradiation followed by transplantation of bone marrow cells. In the DSS model of colon epithelial injury, Mal(-/-) mice developed increased inflammation and severity of colitis relative to WT mice. Mal(-/-) mice demonstrated the presence of inflammatory cell infiltrates, increased crypt proliferation, and presence of neoformations. Furthermore, in the AOM/DSS model, Mal(-/-) mice had greater incidence of tumors. Mal(-/-) and WT bone marrow chimeras demonstrated that nonhematopoietic cell expression of Mal had an important protective role in the control of intestinal inflammation and inflammation-associated cancer. Mal is essential for the maintenance of intestinal homeostasis and expression of Mal in nonhematopoietic cells prevents chronic intestinal inflammation that may predispose to colon neoplasia.
Collapse
Affiliation(s)
- Gabriella Aviello
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | | | | |
Collapse
|
29
|
Bernard NJ, O'Neill LA. Mal, more than a bridge to MyD88. IUBMB Life 2014; 65:777-86. [PMID: 23983209 DOI: 10.1002/iub.1201] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 07/16/2013] [Indexed: 01/17/2023]
Abstract
The family of type 1 transmembrane proteins known as Toll-like receptors (TLRs) provide early immune system recognition and response to infection. In order to transmit their signal to the nucleus and initiate activation of pro-inflammatory and anti-microbial genes, TLRs must initiate a cytoplasmic signalling cascade, which is alternately controlled by 6 known signalling adaptors. These signaling adaptors are crucial for activating the correct immune response to any given TLR / pathogen interaction. This review will focus on one of those adaptors, MyD88 adaptor-like (Mal), also known as TIRAP. Mal is critical for signalling by the best studied of the TLRs, the Gram negative bacterial lipopolysaccharide (LPS) sensor, TLR4. Mal's role in TLR2 signalling in response to activation of the bacterial lipopeptide receptor, TLR2, is more contentious. Mal is a component of the so-called 'MyD88-dependent pathway' in TLR4 signalling. Recent advances in our understanding of the signalling pathways downstream of Mal highlight MyD88-indpendent roles, thus positioning Mal as multifunctional and integral for the molecular control of bacterial infections as well as inflammatory diseases. Here we describe the sequence of molecular events involved in the signalling pathways controlled by Mal, and the importance of Mal in driving host protection against a variety of bacteria, with specific attention to the evidence for Mal's role in TLR2 signalling, recent structural findings that have altered our understanding of Mal signalling, and evidence that single nucleotide polymorphisms (SNPs) of Mal are responsible for variations in population level resistance and susceptibility to bacterial infection.
Collapse
Affiliation(s)
- Nicholas J Bernard
- Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland.
| | | |
Collapse
|