1
|
Kaak JL, Lobo de Sá FD, Turner JR, Schulzke JD, Bücker R. Unraveling the intestinal epithelial barrier in cyanotoxin microcystin-treated Caco-2 cell monolayers. Ann N Y Acad Sci 2022; 1516:188-196. [PMID: 35883254 PMCID: PMC9588585 DOI: 10.1111/nyas.14870] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Microcystin is a widespread cyanobacterial toxin that affects the intestine to produce diarrheal symptoms after ingestion of freshwater blue-green algae. Our study aimed to characterize the mechanism by which the toxin leads to diarrhea via epithelial barrier dysfunction in a small intestine Caco-2 cell model. Microcystin-treated human Caco-2 epithelial monolayers were functionally and molecularly analyzed for barrier dysfunction. Tight junctions (TJs) and cell damage were analyzed in relation to transepithelial electrical resistance (TER) changes. TER of microcystin-treated Caco-2 cells was reduced by 65% of the initial value after 24 h; concomitantly, permeability for fluorescein increased 2.6-fold. Western blot analysis showed reduced claudin-1 expression, while expression of claudin-3 and -4 remained unchanged. Super-resolution stimulated emission depletion microscopy revealed that TJ integrity was compromised by fraying and splitting of the TJ domain of the epithelial cells. Epithelial apoptosis did not significantly contribute to epithelial barrier dysfunction, while cytoskeletal actomyosin constriction was associated with TJ disintegration and the barrier defect. Our results indicate that microcystin causes intestinal barrier leakiness, which helps to explain the leak flux type of diarrhea as the main pathomechanism after ingestion of cyanobacterial toxin.
Collapse
Affiliation(s)
- Jan-Leo Kaak
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Clinical Physiology/Nutritional Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Fábia D. Lobo de Sá
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Clinical Physiology/Nutritional Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Jerrold R. Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jörg-Dieter Schulzke
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Clinical Physiology/Nutritional Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Roland Bücker
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Clinical Physiology/Nutritional Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
2
|
Inflammatory cytokines directly disrupt the bovine intestinal epithelial barrier. Sci Rep 2022; 12:14578. [PMID: 36028741 PMCID: PMC9418144 DOI: 10.1038/s41598-022-18771-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/18/2022] [Indexed: 11/08/2022] Open
Abstract
The small intestinal mucosa constitutes a physical barrier separating the gut lumen from sterile internal tissues. Junctional complexes between cells regulate transport across the barrier, preventing water loss and the entry of noxious molecules or pathogens. Inflammatory diseases in cattle disrupt this barrier; nonetheless, mechanisms of barrier disruption in cattle are poorly understood. We investigated the direct effects of three inflammatory cytokines, TNFα, IFNγ, and IL-18, on the bovine intestinal barrier utilizing intestinal organoids. Flux of fluorescein isothiocyanate (FITC)-labeled dextran was used to investigate barrier permeability. Immunocytochemistry and transmission electron microscopy were used to investigate junctional morphology, specifically tortuosity and length/width, respectively. Immunocytochemistry and flow cytometry was used to investigate cellular turnover via proliferation and apoptosis. Our study shows that 24-h cytokine treatment with TNFα or IFNγ significantly increased dextran permeability and tight junctional tortuosity, and reduced cellular proliferation. TNFα reduced the percentage of G2/M phase cells, and IFNγ treatment increased cell apoptotic rate. IL-18 did not directly induce significant changes to barrier permeability or cellular turnover. Our study concludes that the inflammatory cytokines, TNFα and IFNγ, directly induce intestinal epithelial barrier dysfunction and alter the tight junctional morphology and rate of cellular turnover in bovine intestinal epithelial cells.
Collapse
|
3
|
Fink M, Wrana JL. Regulation of homeostasis and regeneration in the adult intestinal epithelium by the TGF-β superfamily. Dev Dyn 2022; 252:445-462. [PMID: 35611490 DOI: 10.1002/dvdy.500] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 11/09/2022] Open
Abstract
The delicate balance between the homeostatic maintenance and regenerative capacity of the intestine makes this a fascinating tissue of study. The intestinal epithelium undergoes continuous homeostatic renewal but is also exposed to a diverse array of stresses that can range from physiological processes such as digestion, to exposure to infectious agents, drugs, radiation therapy, and inflammatory stimuli. The intestinal epithelium has thus evolved to efficiently maintain and reinstate proper barrier function that is essential for intestinal integrity and function. Factors governing homeostatic epithelial turnover are well described, however, the dynamic regenerative mechanisms that occur following injury are the subject of intense ongoing investigations. The TGF-β superfamily is a key regulator of both homeostatic renewal and regenerative processes of the intestine. Here we review the roles of TGF-β and BMP on the adult intestinal epithelium during self-renewal and injury to provide a framework for understanding how this major family of morphogens can tip the scale between intestinal health and disease. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Mardi Fink
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Jeffrey L Wrana
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Lobo de Sá FD, Schulzke JD, Bücker R. Diarrheal Mechanisms and the Role of Intestinal Barrier Dysfunction in Campylobacter Infections. Curr Top Microbiol Immunol 2021; 431:203-231. [PMID: 33620653 DOI: 10.1007/978-3-030-65481-8_8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Campylobacter enteritis is the most common cause of foodborne bacterial diarrhea in humans. Although various studies have been performed to clarify the pathomechanism in Campylobacter infection, the mechanism itself and bacterial virulence factors are yet not completely understood. The purpose of this chapter is to (i) give an overview on Campylobacter-induced diarrheal mechanisms, (ii) illustrate underlying barrier defects, (iii) explain the role of the mucosal immune response and (iv) weigh preventive and therapeutic approaches. Our present knowledge of pathogenetic and diarrheal mechanisms of Campylobacter jejuni is explained in the first part of this chapter. In the second part, the molecular basis for the Campylobacter-induced barrier dysfunction is compared with that of other species in the Campylobacter genus. The bacteria are capable of overcoming the intestinal epithelial barrier. The invasion into the intestinal mucosa is the initial step of the infection, followed by a second step, the epithelial barrier impairment. The extent of the impairment depends on various factors, including tight junction dysregulation and epithelial apoptosis. The disturbed intestinal epithelium leads to a loss of water and solutes, the leak flux type of diarrhea, and facilitates the uptake of harmful antigens, the leaky gut phenomenon. The barrier dysfunction is accompanied by increased pro-inflammatory cytokine secretion, which is partially responsible for the dysfunction. Moreover, cytokines also mediate ion channel dysregulation (e.g., epithelial sodium channel, ENaC), leading to another diarrheal mechanism, which is sodium malabsorption. Future perspectives of Campylobacter research are the clarification of molecular pathomechanisms and the characterization of therapeutic and preventive compounds to combat and prevent Campylobacter infections.
Collapse
Affiliation(s)
- Fábia Daniela Lobo de Sá
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité - University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Jörg-Dieter Schulzke
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité - University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Roland Bücker
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité - University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany.
| |
Collapse
|
5
|
Sun W, Zhang J, Zhou C, Yan B, Cai Q, He H, Duan X, Fan H. Differential Analysis of Serum Principal Components Treated with Compound Sophora Decoction and Related Compounds Based on High-Resolution Mass Spectrometry (HRMS). EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:7518479. [PMID: 33062021 PMCID: PMC7545453 DOI: 10.1155/2020/7518479] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/11/2020] [Accepted: 09/16/2020] [Indexed: 01/14/2023]
Abstract
OBJECTIVE To compare the differences in the serum principal components in ulcerative colitis- (UC-) induced rats, treated with compound Sophora decoction, matrine, oxymatrine monomer mixture, and indirubin monomer, and to provide a modern scientific basis for elucidating the clinical efficacy of compound Sophora decoction for the treatment of UC. METHODS The serum samples of rats from each group were obtained after drug administration, and the serum principal components of each group were analyzed by high-resolution mass spectrometry. Agilent Eclipse XDB C18 chromatographic column (100 mm × 2.1 mm, 3.5 m) was used for separation. The mobile phase was water (A) and methanol (B) (0.1% formic acid) gradient elution, 0-3 min (B: 20%-40%), 3-10 min (B: 40%-54%), 10-25 min (B: 54%), 25-35 min (B: 54%-70%), 35-45 min (B: 70%-80%), 45-50 min (B: 80%), 50-60 min (B: 80%-100%), 70-72 min (B: 100%-20%), and 72-77 min (B: 20%); flow rate, 300 μL/min; column temperature, 40°C; and injection volume, 10 μL. ESI source was selected and scanned in the positive and negative ion modes. The scanning range was 70-1500 m/z; ion-source gas 1 (GS1): 55 psi; ion-source gas 2 (GS2): 60 psi; CUR: 30 psi; ion-source temperature (TEM): 550°C; ion-source voltage (ISVF) : 5500 V/-4500 V; decluster voltage (DP): 100 V; collision energy (CE): 35 V/-35 V; collision energy gain (CES) : 15 V/-15 V; and data acquisition mode: IDA. After the data from each group were imported into MarkView 1.3, the molecular weights and retention times of different substances were obtained and qualitatively analyzed by ChemSpider and PeakView 2.0. RESULTS In the negative ion mode, 26 differential compounds were identified in the compound Sophora decoction group (FFKST) compared to the model group (M), and 18 differential compounds were identified in the matrine and oxymatrine group (KST) compared to the model group (M). In the positive ion mode, 11 and 7 differential compounds were identified in the compound Sophora decoction group (FFKST) and the matrine and oxymatrine group (KST) compared to the model group (M), respectively. The responses of all compounds in each group were compared with each other. As the different principal component substances in the indirubin group (DYH) displayed little correlation with other groups, the different components in this group were not researched thoroughly. CONCLUSION By comparing the differences in the serum principal components from each administration group, we found that the FFKST group exhibited enhanced synthesis of the serum principal components; however, the compound doses of matrine and oxymatrine monomers did not exhibit the same changes in the serum principal components of UC-induced rats. Finally, the traditional Chinese medicine compound is more advantageous than monomers.
Collapse
Affiliation(s)
- Wanjin Sun
- Department of Pharmacy, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430061, China
- Department of Pharmacy, Hubei Province Academy of Traditional Chinese Medicine, Wuhan 430074, China
| | - Junjie Zhang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 300000, China
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Conghui Zhou
- Department of Pharmacy, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430061, China
- Department of Pharmacy, Hubei Province Academy of Traditional Chinese Medicine, Wuhan 430074, China
| | - Bin Yan
- Department of Pharmacy, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430061, China
- Department of Pharmacy, Hubei Province Academy of Traditional Chinese Medicine, Wuhan 430074, China
| | - Quan Cai
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongxia He
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xueyun Duan
- Department of Pharmacy, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430061, China
- Department of Pharmacy, Hubei Province Academy of Traditional Chinese Medicine, Wuhan 430074, China
| | - Heng Fan
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
6
|
Butkevych E, Lobo de Sá FD, Nattramilarasu PK, Bücker R. Contribution of Epithelial Apoptosis and Subepithelial Immune Responses in Campylobacter jejuni- Induced Barrier Disruption. Front Microbiol 2020; 11:344. [PMID: 32210941 PMCID: PMC7067706 DOI: 10.3389/fmicb.2020.00344] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/17/2020] [Indexed: 12/17/2022] Open
Abstract
Campylobacter jejuni is a widespread zoonotic pathogen and the leading bacterial cause of foodborne gastroenteritis in humans. Previous infection studies showed disruption of intercellular contacts, induction of epithelial apoptosis, and immune activation, all three contributing to intestinal barrier dysfunction leading to diarrhea. The present study aims to determine the impact of subepithelial immune cells on intestinal barrier dysfunction during Campylobacter jejuni infection and the underlying pathological mechanisms. Infection was performed in a co-culture of confluent monolayers of the human colon cell line HT-29/B6-GR/MR and THP-1 immune cells. Twenty-two hours after infection, transepithelial electrical resistance (TER) was decreased by 58 ± 6% compared to controls. The infection resulted in an increase in permeability for fluorescein (332 Da; 4.5-fold) and for FITC-dextran (4 kDa; 3.5-fold), respectively. In contrast, incubation of the co-culture with the pan-caspase inhibitor Q-VD-OPh during the infection resulted in a complete recovery of the decrease in TER and a normalization of flux values. Fluorescence microscopy showed apoptotic fragmentation in infected cell monolayers resulting in a 5-fold increase of the apoptotic ratio, accompanied by an increased caspase-3 cleavage and caspase-3/7 activity, which both were not present after Q-VD-OPh treatment. Western blot analysis revealed increased claudin-1 and claudin-2 protein expression. Inhibition of apoptosis induction did not normalize these tight junction changes. TNFα concentration was increased during the infection in the co-culture. In conclusion, Campylobacter jejuni infection and the consequent subepithelial immune activation cause intestinal barrier dysfunction mainly through caspase-3-dependent epithelial apoptosis. Concomitant tight junction changes were caspase-independent. Anti-apoptotic and immune-modulatory substances appear to be promising agents for treatment of campylobacteriosis.
Collapse
Affiliation(s)
- Eduard Butkevych
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Fábia Daniela Lobo de Sá
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Praveen Kumar Nattramilarasu
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Roland Bücker
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
7
|
Bücker R, Zakrzewski SS, Wiegand S, Pieper R, Fromm A, Fromm M, Günzel D, Schulzke JD. Zinc prevents intestinal epithelial barrier dysfunction induced by alpha-hemolysin-producing Escherichia coli 536 infection in porcine colon. Vet Microbiol 2020; 243:108632. [PMID: 32273011 DOI: 10.1016/j.vetmic.2020.108632] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/10/2020] [Accepted: 03/02/2020] [Indexed: 11/19/2022]
Abstract
Zinc treatment is beneficial for infectious diarrhea or colitis. This study aims to characterize the pathomechanisms of the epithelial barrier dysfunction caused by alpha-hemolysin (HlyA)-expressing Escherichia coli in the colon mucosa and the mitigating effects of zinc ions. We performed Ussing chamber experiments on porcine colon epithelium and infected the tissues with HlyA-producing E. coli. Colon mucosa from piglets was obtained from a feeding trial with defined normal or high dose zinc feeding (pre-conditioning). Additional to the zinc feeding, zinc was added to the luminal compartment of the Ussing chamber. Transepithelial electrical resistance (TER) was measured during the infection of the living tissue and subsequently the tissues were immuno-stained for confocal microscopy. Zinc applied to the luminal compartment was effective in preventing from E. coli-induced epithelial barrier dysfunction in Ussing chamber experiments. In contrast, zinc pre-conditioning of colon mucosae when zinc ions were missing subsequently in the luminal compartment was not sufficient to prevent epithelial barrier impairment during E. coli infection. The pathological changes caused by E. coli HlyA were alterations of tight junction proteins claudin-4 and claudin-5, focal leak formation, and cell exfoliation which reflected the paracellular barrier defect measured by a reduced TER. In microscopic analysis of luminal zinc-treated mucosae these changes were absent. In conclusion, continuous presence of unbound zinc ions in the luminal compartment is essential for the protective action of zinc against E. coli HlyA. This suggests the usage of zinc as therapeutic regimen, while prophylactic intervention by high dietary zinc loads may be less useful.
Collapse
Affiliation(s)
- Roland Bücker
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department Division of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Silke S Zakrzewski
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department Division of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stephanie Wiegand
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department Division of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Robert Pieper
- Institute of Animal Nutrition, Freie Universität Berlin, Berlin, Germany
| | - Anja Fromm
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department Division of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Fromm
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department Division of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dorothee Günzel
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department Division of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jörg-Dieter Schulzke
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department Division of Gastroenterology, Infectiology and Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
8
|
Kahlert S, Renner L, Kluess J, Frahm J, Tesch T, Bannert E, Kersten S, Dänicke S, Rothkötter HJ. Effects of deoxynivalenol-feed contamination on circulating LPS in pigs. Innate Immun 2019; 25:168-175. [PMID: 30760085 PMCID: PMC6830939 DOI: 10.1177/1753425919829552] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Low concentration of LPS can be detected in healthy mammals without triggering
systemic inflammation. Here we analysed the influence of the mycotoxin
deoxynivalenol (DON) on very low LPS concentrations and the role of DON in the
physiology of pigs challenged with high artificial LPS dosage mimicking septic
shock. Pigs were fed for 29 d with DON-contaminated (4.59 mg/kg feed) or control
feed. Samples of control animals showed 6.6 ± 13.5 pg/ml LPS in portal and
3.1 ± 7.6 pg/ml LPS in jugular serum samples. In the DON fed group,
3.4 ± 7.2 pg/ml and 0.6 ± 0.8 pg/ml were detected. The differences were
statistically not significant, indicating that DON is not a trigger for enhanced
LPS transfer into the blood circulation. Next, pigs were challenged with 7.5 µg
LPS/kg body mass via portal or jugular route. The application route did not
significantly influence the LPS concentration. We expected higher circulating
LPS concentrations in the presence of DON due to the additional stress of liver
metabolism and reduced liver capacity to remove LPS from circulation. This
scenario is supported by tendency. In summary, we found that DON is unlikely to
influence LPS transfer in the gut; DON likely reduces the capacity for LPS
removal in septic shock conditions.
Collapse
Affiliation(s)
- Stefan Kahlert
- 1 Institute of Anatomy, Otto von Guericke University Magdeburg, Germany
| | - Lydia Renner
- 1 Institute of Anatomy, Otto von Guericke University Magdeburg, Germany
| | - Jeannette Kluess
- 2 Institute of Animal Nutrition, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Braunschweig, Germany
| | - Jana Frahm
- 2 Institute of Animal Nutrition, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Braunschweig, Germany
| | - Tanja Tesch
- 2 Institute of Animal Nutrition, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Braunschweig, Germany
| | - Erik Bannert
- 2 Institute of Animal Nutrition, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Braunschweig, Germany
| | - Susanne Kersten
- 2 Institute of Animal Nutrition, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Braunschweig, Germany
| | - Sven Dänicke
- 2 Institute of Animal Nutrition, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Braunschweig, Germany
| | | |
Collapse
|
9
|
Karsch S, Kong D, Großhans J, Janshoff A. Single-Cell Defects Cause a Long-Range Mechanical Response in a Confluent Epithelial Cell Layer. Biophys J 2017; 113:2601-2608. [PMID: 29129266 DOI: 10.1016/j.bpj.2017.10.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 10/05/2017] [Accepted: 10/16/2017] [Indexed: 02/06/2023] Open
Abstract
Epithelial cells are responsible for tissue homeostasis and form a barrier to maintain chemical gradients and mechanical integrity. Therefore, rapid wound closure is crucial for proper tissue function and restoring homeostasis. In this study, the mechanical properties of cells surrounding a single-cell wound are investigated during closure of the defect. The single-cell wound is induced in an intact layer using micropipette action and responses in neighboring cells are monitored with atomic force microscopy. Direct neighbors reveal a rise in the apparent pretension, which is dominated by cortical tension. The same effect was observed for a single-cell wound induced by laser ablation and during closure of a not fully confluent layer. Moreover, changes in the apparent pretension are far reaching and persist even in cells separated by three cell widths from the defect. This shows that epithelial cells respond to minimal wounds in a collective fashion by increased contractility with substantial reach.
Collapse
Affiliation(s)
- Susanne Karsch
- Institute for Physical Chemistry, University of Göttingen, Göttingen, Germany
| | - Deqing Kong
- Institute for Developmental Biochemistry, Medical School, University of Göttingen, Göttingen, Germany
| | - Jörg Großhans
- Institute for Developmental Biochemistry, Medical School, University of Göttingen, Göttingen, Germany
| | - Andreas Janshoff
- Institute for Physical Chemistry, University of Göttingen, Göttingen, Germany.
| |
Collapse
|
10
|
Bücker R, Krug SM, Fromm A, Nielsen HL, Fromm M, Nielsen H, Schulzke JD. Campylobacter fetus impairs barrier function in HT-29/B6 cells through focal tight junction alterations and leaks. Ann N Y Acad Sci 2017; 1405:189-201. [PMID: 28662272 DOI: 10.1111/nyas.13406] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/08/2017] [Accepted: 05/11/2017] [Indexed: 12/12/2022]
Abstract
Infections by Campylobacter species are the most common foodborne zoonotic disease worldwide. Campylobacter jejuni and C. coli are isolated most frequently from human stool samples, but severe infections by C. fetus (Cf), which can cause gastroenteritis, septicemia, and abortion, are also found. This study aims at the characterization of pathological changes in Cf infection using an intestinal epithelial cell model. The Cf-induced epithelial barrier defects appeared earlier than those of avian Campylobacter species like C. jejuni/C. coli. Two-path impedance spectroscopy (2PI) distinguished transcellular and paracellular resistance contributions to the overall epithelial barrier impairment. Both transcellular and paracellular resistance of Cf-infected HT-29/B6 monolayers were reduced. The latter was attributed to activation of active anion secretion. Western blot analysis showed no decrease in tight junction (TJ) protein expression (claudin-1, -2, -3, and -4) but showed redistribution of claudin-1 off the TJ domain. In addition, Cf induced epithelial cell death, cell detachment, and lesions (focal leaks), as the result of which macromolecule flux (10-kDa dextran) was increased in Cf-invaded cell monolayers. In conclusion, barrier dysfunction from Cf infection was due to TJ protein redistribution, cell death induction, and leak formation, resulting in bacterial translocation, ion leak flux, and antigen uptake (leaky gut).
Collapse
Affiliation(s)
- Roland Bücker
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Institute of Clinical Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Susanne M Krug
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Institute of Clinical Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anja Fromm
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Institute of Clinical Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Hans Linde Nielsen
- Department of Clinical Microbiology, Aalborg University Hospital, Aalborg, Denmark
| | - Michael Fromm
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Institute of Clinical Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Henrik Nielsen
- Department of Infectious Diseases, Aalborg University Hospital, Aalborg, Denmark
| | - Jörg-Dieter Schulzke
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Institute of Clinical Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
11
|
Zinc treatment is efficient against Escherichia coli α-haemolysin-induced intestinal leakage in mice. Sci Rep 2017; 7:45649. [PMID: 28361997 PMCID: PMC5374507 DOI: 10.1038/srep45649] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 03/01/2017] [Indexed: 12/17/2022] Open
Abstract
Zinc homoeostasis exerts protective effects in inflammatory intestinal diseases and zinc supplementation has been successfully used for treating infectious diarrhoea. This study aimed at a characterisation of zinc effects on focal leak induction by α-haemolysin (HlyA)-producing Escherichia coli (E. coli) as protective mechanism for colitis. We conducted in vivo experiments by oral challenge of gnotobiotic mice colonised with HlyA-expressing E. coli-536. Mice were either fed a defined normal or high zinc diet to analyse effects of zinc as a therapeutic regimen. HlyA-deficient E. coli-536 mutants were used as controls. Mice infected with HlyA-producing E. coli showed impaired barrier integrity when receiving normal zinc. High zinc supplementation in HlyA-producing E. coli-infected mice reduced epithelial dysfunction as indicated by ameliorated macromolecule permeability. Reduced size of focal leaks with diminished bacterial translocation was observed as inherent mechanisms of this zinc action. In human colon cell monolayers application of zinc rescued the HlyA-dependent decline in transepithelial electrical resistance via reduction of the calcium entry into HlyA-exposed cells. Calcium-dependent cell exfoliation was identified as mechanism for focal leak induction. In conclusion, zinc supplementation protects from HlyA-induced barrier dysfunction in vivo and in vitro, providing an explanation for the protective efficacy of zinc in intestinal disorders.
Collapse
|
12
|
14-3-3 Proteins regulate Akt Thr308 phosphorylation in intestinal epithelial cells. Cell Death Differ 2016; 23:1060-72. [PMID: 26846144 DOI: 10.1038/cdd.2015.163] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 11/14/2015] [Accepted: 11/24/2015] [Indexed: 12/11/2022] Open
Abstract
Akt activation has been associated with proliferation, differentiation, survival and death of epithelial cells. Phosphorylation of Thr308 of Akt by phosphoinositide-dependent kinase 1 (PDK1) is critical for optimal stimulation of its kinase activity. However, the mechanism(s) regulating this process remain elusive. Here, we report that 14-3-3 proteins control Akt Thr308 phosphorylation during intestinal inflammation. Mechanistically, we found that IFNγ and TNFα treatment induce degradation of the PDK1 inhibitor, 14-3-3η, in intestinal epithelial cells. This mechanism requires association of 14-3-3ζ with raptor in a process that triggers autophagy and leads to 14-3-3η degradation. Notably, inhibition of 14-3-3 function by the chemical inhibitor BV02 induces uncontrolled Akt activation, nuclear Akt accumulation and ultimately intestinal epithelial cell death. Our results suggest that 14-3-3 proteins control Akt activation and regulate its biological functions, thereby providing a new mechanistic link between cell survival and apoptosis of intestinal epithelial cells during inflammation.
Collapse
|
13
|
Kim MN, Koh SJ, Kim JM, Im JP, Jung HC, Kim JS. Clostridium difficile infection aggravates colitis in interleukin 10-deficient mice. World J Gastroenterol 2014; 20:17084-17091. [PMID: 25493020 PMCID: PMC4258576 DOI: 10.3748/wjg.v20.i45.17084] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Revised: 06/27/2014] [Accepted: 07/30/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of Clostridium difficile (C. difficile) infection in an interleukin 10-deficient (IL-10-/-) mouse model of inflammatory bowel disease.
METHODS: Bone marrow-derived dendritic cells isolated from wild type (WT) and IL-10-/-mice were stimulated for 4 h with C. difficile toxin A (200 μg/mL), and gene expression of interferon (IFN)-γ, IL-12 and IL-23 was determined by real-time reverse transcription polymerase chain reaction. WT and IL-10-/- mice (n = 20 each) were exposed to an antibiotic cocktail for three days and then were injected with clindamycin (i.p.). Mice (n = 10 WT, 10 IL-10-/-) were then challenged with oral administration of C. difficile (1 × 105 colony forming units of strain VPI 10463). Animals were monitored daily for 7 d for signs of colitis. Colonic tissue samples were evaluated for cytokine gene expression and histopathologic analysis.
RESULTS: C. difficile toxin A treatment induced IFN-γ gene expression to a level that was significantly higher in BDMCs from IL-10-/- compared to those from WT mice (P < 0.05). However, expression of IL-12 and IL-23 was not different among the groups. Following C. difficile administration, mice developed diarrhea and lost weight within 2-3 d. Weight loss was significantly greater in IL-10-/- compared to WT mice (P < 0.05). C. difficile infection induced histopathologic features typical of colitis in both IL-10-/- and WT mice. The histopathologic severity score was significantly higher in the IL-10-/- than in WT mice (mean ± standard error; 5.50 ± 0.53 vs 2.44 ± 0.46; P < 0.05). This was accompanied by a significantly greater increase in IFN-γ gene expression in colonic tissues from IL-10-/- than from WT mice challenged with C. difficile (P < 0.05).
CONCLUSION: These results indicate that colitis is more severe after C. difficile infection in IL-10-/-mice, and that IFN-γ expression is involved in this process.
Collapse
MESH Headings
- Animals
- Bacterial Toxins/pharmacology
- Bone Marrow Cells/drug effects
- Bone Marrow Cells/immunology
- Bone Marrow Cells/metabolism
- Cells, Cultured
- Clostridioides difficile/pathogenicity
- Colitis/genetics
- Colitis/immunology
- Colitis/metabolism
- Colitis/microbiology
- Colitis/pathology
- Colon/immunology
- Colon/metabolism
- Colon/microbiology
- Colon/pathology
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Disease Models, Animal
- Enterocolitis, Pseudomembranous/genetics
- Enterocolitis, Pseudomembranous/immunology
- Enterocolitis, Pseudomembranous/metabolism
- Enterocolitis, Pseudomembranous/microbiology
- Enterocolitis, Pseudomembranous/pathology
- Enterotoxins/pharmacology
- Interferon-gamma/genetics
- Interferon-gamma/metabolism
- Interleukin-10/deficiency
- Interleukin-10/genetics
- Interleukin-12/genetics
- Interleukin-12/metabolism
- Interleukin-23/genetics
- Interleukin-23/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- RNA, Messenger/metabolism
- Severity of Illness Index
- Time Factors
- Weight Loss
Collapse
|
14
|
Acute exercises induce disorders of the gastrointestinal integrity in a murine model. Eur J Appl Physiol 2013; 114:609-17. [PMID: 24352573 DOI: 10.1007/s00421-013-2791-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 12/05/2013] [Indexed: 01/16/2023]
Abstract
PURPOSE Many endurance athletes complain about gastrointestinal (GI) symptoms. It is assumed that exercise-induced shift of perfusion with consecutive hypoperfusion of the enteral vascular system leads to an increased GI permeability and tissue damage. Therefore, the aim of the study was to investigate permeability, apoptosis, electrogenic ion transport (Isc), and tissue conductance (Gt) of the small intestine in a murine exercise model. METHODS After spirometry, male Swiss CD-1 mice were subjected to an intensive treadmill exercise (80% VO2max). Sedentary mice served as controls. The small intestine was removed at several time intervals post-exercise. Apoptotic cells were determined by the TUNEL method, while fluorescein isothiocyanate dextran permeation indicated intestinal permeability. The Gt and Isc measurements were carried out in a modified Ussing chamber. RESULTS Apoptosis of epithelial cells increased continuously until 24 h post exercise (0.8 ± 0.42 versus 39.2 ± 26.0%; p < 0.05). Compared with the control group the permeability increased 2 h after exercise (0.47 ± 0.07 versus 0.67 ± 0.14 FU/min; p < 0.05). Isc measurements of the ileum were augmented after 24 h (3.33 ± 0.56 versus 5.77 ± 1.16 μEq/h/cm(2); p < 0.05). At this time the Gt increased as well (28.8 ± 3.37 versus 32.5 ± 2.59 mS/cm(2); p < 0.05). CONCLUSION In the murine exercise model there is evidence that after intense endurance exercise repair processes occur in small intestinal epithelial cells, which affect permeability, Gt, and Isc. The formation of lamellipodia to close the "leaky" tight junctions caused by apoptosis might be an underlying mechanism.
Collapse
|
15
|
Chen CC, Zhou Y, Morris CA, Hou J, Baker LA. Scanning ion conductance microscopy measurement of paracellular channel conductance in tight junctions. Anal Chem 2013; 85:3621-8. [PMID: 23421780 PMCID: PMC3648657 DOI: 10.1021/ac303441n] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Elucidation of epithelial transport across transcellular or paracellular pathways promises to advance the present understanding of ion transport and enables regulation of cell junctions critical to the cell and molecular biology of the epithelium. Here, we demonstrate a new instrumental technique, potentiometric scanning ion conductance microscopy (P-SICM), that utilizes a nanoscale pipet to differentiate paracellular and transcellular transport processes at high spatial resolution. The technique is validated for well-defined polymer membranes and then employed to study wild type and claudin-deficient mutants of Madin-Darby Canine Kidney strain II (MDCKII) cells. Paracellular permeabilities conferred by claudin-2 are captured by P-SICM which demonstrates the utility to monitor apparent conductance at subcellular levels.
Collapse
Affiliation(s)
- Chiao-Chen Chen
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405
| | - Yi Zhou
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405
| | - Celeste A. Morris
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405
| | - Jianghui Hou
- Renal Division, Washington University Medical School, 660 South Euclid Avenue, St. Louis, Missouri 63110
- Center for Investigation of Membrane Excitability Diseases, Washington University Medical School, 660 South Euclid Avenue, St Louis, Missouri 63110
| | - Lane A. Baker
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405
| |
Collapse
|
16
|
Corridoni D, Pastorelli L, Mattioli B, Locovei S, Ishikawa D, Arseneau KO, Chieppa M, Cominelli F, Pizarro TT. Probiotic bacteria regulate intestinal epithelial permeability in experimental ileitis by a TNF-dependent mechanism. PLoS One 2012; 7:e42067. [PMID: 22848704 PMCID: PMC3405026 DOI: 10.1371/journal.pone.0042067] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2012] [Accepted: 07/02/2012] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND We previously showed that the probiotic mixture, VSL#3, prevents the onset of ileitis in SAMP/YitFc (SAMP) mice, and this effect was associated with stimulation of epithelial-derived TNF. The aim of this study was to determine the mechanism(s) of VSL#3-mediated protection on epithelial barrier function and to further investigate the "paradoxical" effects of TNF in preventing SAMP ileitis. METHODS Permeability was evaluated in SAMP mice prior to the onset of inflammation and during established disease by measuring transepithelial electrical resistance (TEER) on ex vivo-cultured ilea following exposure to VSL#3 conditioned media (CM), TNF or VSL#3-CM + anti-TNF. Tight junction (TJ) proteins were assessed by qRT-PCR, Western blot, and confocal microscopy, and TNFRI/TNFRII expression measured in freshly isolated intestinal epithelial cells (IEC) from SAMP and control AKR mice. RESULTS Culture with either VSL#3-CM or TNF resulted in decreased ileal paracellular permeability in pre-inflamed SAMP, but not SAMP with established disease, while addition of anti-TNF abrogated these effects. Modulation of the TJ proteins, claudin-2 and occludin, occurred with a significant decrease in claudin-2 and increase in occludin following stimulation with VSL#3-CM or TNF. TNF protein levels increased in supernatants of SAMP ilea incubated with VSL#3-CM compared to vehicle, while IEC-derived TNFR mRNA expression decreased in young, and was elevated in inflamed, SAMP versus AKR mice. CONCLUSIONS Our data demonstrate that the previously established efficacy of VSL#3 in preventing SAMP ileitis is due to direct innate and homeostatic effects of TNF on the gut epithelium, modulation of the TJ proteins, claudin-2 and occludin, and overall improvement of intestinal permeability.
Collapse
Affiliation(s)
- Daniele Corridoni
- Division of Gastroenterology and Liver Disease, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Luca Pastorelli
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Benedetta Mattioli
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Silviu Locovei
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Division of Gastroenterology and Liver Disease, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Dai Ishikawa
- Division of Gastroenterology and Liver Disease, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Kristen O. Arseneau
- Division of Gastroenterology and Liver Disease, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Marcello Chieppa
- Lab of Experimental Immunopathology, Instituto di Ricovero e Cura a Carattere Scientifico (IRCCS) “De Bellis”, Castellana Grotte, Bari, Italy
| | - Fabio Cominelli
- Division of Gastroenterology and Liver Disease, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Theresa T. Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| |
Collapse
|
17
|
Kim JM. [Inflammatory bowel diseases and inflammasome]. THE KOREAN JOURNAL OF GASTROENTEROLOGY 2012; 58:300-10. [PMID: 22198227 DOI: 10.4166/kjg.2011.58.6.300] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Inflammatory bowel disease (IBD), the most important entities being ulcerative colitis and Crohn's disease, are chronic, relapsing and remitting inflammatory conditions that result from chronic dysregulation of the mucosal immune system in the intestinal tract. Although the precise pathogenesis of IBD is still incompletely understood, increased levels of proinflammatory cytokines, including interleukin (IL)-1b, IL-18 and tumor necrosis factor-a, are detected in active IBD and correlate with the severity of inflammation, indicating that these cytokines may play a key role in the development of IBD. Recently, the intracellular nucleotide-binding oligomerization domain-like receptor (NLR) family members, including NLRP1, NLRP3, NLRC4 and NLRP6, are emerging as important regulators of intestinal homeostasis. Together, one of those aforementioned molecules or the DNA sensor absent in melanoma 2 (AIM2), apoptosis-associated speck-like protein containing 'a caspase recruitment domain (CARD)' (ASC) and caspase-1 form a large (> 700 kDa) multi-protein complex called the inflammasome. Stimulation with specific microbial and endogenous molecules triggers inflammasome assembly and caspase-1 activation. Activated caspase-1 leads to the secretion of proinflammatory cytokines, including IL-1b and IL-18, and the promotion of pyroptosis, a form of phagocyte cell death induced by bacterial pathogens, in an inflamed tissue. Therefore, inflammasomes are assumed to mediate host defense against microbial pathogens and gut homeostasis, so that their dysregulation might contribute to IBD pathogenesis. This review focuses on recent advances of the role of NLRP3 inflammasome signaling in IBD pathogenesis. Improving knowledge of the inflammasome could provide insights into potential therapeutic targets for patients with IBD.
Collapse
Affiliation(s)
- Jung Mogg Kim
- Department of Microbiology, Hanyang University College of Medicine, Seoul, Korea.
| |
Collapse
|
18
|
Bücker R, Krug SM, Rosenthal R, Günzel D, Fromm A, Zeitz M, Chakraborty T, Fromm M, Epple HJ, Schulzke JD. Aerolysin From Aeromonas hydrophila Perturbs Tight Junction Integrity and Cell Lesion Repair in Intestinal Epithelial HT-29/B6 Cells. J Infect Dis 2011; 204:1283-92. [DOI: 10.1093/infdis/jir504] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
19
|
Wang F, Wang F, Zou Z, Liu D, Wang J, Su Y. Active deformation of apoptotic intestinal epithelial cells with adhesion-restricted polarity contributes to apoptotic clearance. J Transl Med 2011; 91:462-71. [PMID: 21042290 DOI: 10.1038/labinvest.2010.182] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Dying epithelial cells are thought to be squeezed out of the epithelium by the contraction of an actomyosin ring formed in live neighboring cells, which simultaneously closes any potential gap, thereby maintaining the integrity of the epithelial layer. The shrinkage and contraction of apoptotic cells contribute little to the extrusion process. In contrast, the clearance of dying intestinal columnar epithelial cells in vivo usually leaves a transient gap via an unknown mechanism. By using freshly isolated small intestinal villus units with or without basal lamina, we found that the nucleus of apoptotic enterocytes moved apically until they budded off, leaving the cytoplasmic residue in the transient gap. Apical polarity of nucleus movement was restricted unless the basal lamina was artificially removed. F-actin mainly accumulated in apoptotic cells rather than neighboring live cells, even after the addition of resistance force against extrusion. The actin accumulation in apoptotic cells does not depend on the living state of neighboring cells. Apoptotic cells can complete the shedding process when neighboring a goblet cell, as the majority of space is occupied by mucin granules and the cytoplasm consists of intermediate filaments and microtubules, but lacks F-actin. We found that the elongation and deformation of apoptotic cells depend on the stretching force generated inside the cell, rather than the force generated by neighboring cells extending. Our findings clearly demonstrate that intestinal epithelial shedding does not depend on the formation and contraction of an actomyosin ring in live neighboring cells. Apoptotic epithelial cells may undergo an active process of cell deformation with adhesion-restricted polarity, which may contribute to maintaining barrier function during a high rate of cellular turnover.
Collapse
Affiliation(s)
- Fengchao Wang
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | | | | | | | | | | |
Collapse
|
20
|
Hillenbrand B, Günzel D, Richter JF, Höhne M, Schreier E, Schulzke JD, Mankertz J. Norovirus non-structural protein p20 leads to impaired restitution of epithelial defects by inhibition of actin cytoskeleton remodelling. Scand J Gastroenterol 2010; 45:1307-19. [PMID: 20695836 DOI: 10.3109/00365521.2010.483013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Norovirus is the most common cause of acute gastroenteritis in humans worldwide. Typical symptoms are vomiting, nausea and severe watery diarrhea. Because of the lack of cell lines susceptible to human norovirus infection, pathomechanisms and replication cycle are largely unknown. Here, we address the issue of how norovirus infection could lead to epithelial barrier dysfunction. MATERIAL AND METHODS Expression of the non-structural norovirus protein p20 in the epithelial cell line HT-29/B6 was activated through a tetracycline sensitive promoter. Tight junction proteins were studied by Western blot and confocal laser scanning microscopy. Apoptoses were detected in TUNEL stainings. Epithelial restitution was monitored by conductance scanning after induction of single cell lesions. RESULTS Changes in the expression or localization of the tight junction proteins occludin and/or claudin-1, -2,- 3, -4, -5, -7 and -8 could be ruled out to mediate epithelial barrier modulation. Cell motility was also unaltered by p20. Investigation of epithelial apoptosis revealed an accumulation of apoptic cells in epithelial monolayers after induction of p20 expression. In epithelial cell restitution assays, an arrest was identified in p20 expressing cells. Fluorescence microscopy revealed an inability for condensation and redistribution of cellular actin, which led to a reduced transepithelial electrical resistance. CONCLUSIONS Functional data for norovirus protein p20 suggest a role in modulation of the actin cytoskeleton leading to barrier dysfunction through impairment of restitution of epithelial defects.
Collapse
Affiliation(s)
- Bernd Hillenbrand
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
21
|
Seltana A, Basora N, Beaulieu JF. Intestinal epithelial wound healing assay in an epithelial-mesenchymal co-culture system. Wound Repair Regen 2010; 18:114-22. [PMID: 20082684 DOI: 10.1111/j.1524-475x.2009.00554.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Rapid and efficient healing of epithelial damage is critical to the functional integrity of the small intestine. Epithelial repair is a complex process that has largely been studied in cultured epithelium but to a much lesser extent in mucosa. We describe a novel method for the study of wound healing using a co-culture system that combined an intestinal epithelial Caco-2/15 cell monolayer cultured on top of human intestinal myofibroblasts, which together formed a basement membrane-like structure that contained many of the major components found at the epithelial-mesenchymal interface in the human intestine. To investigate the mechanism of restitution, small lesions were generated in epithelial cell monolayers on plastic or in co-cultures without disturbing the underlying mesenchymal layer. Monitoring of wound healing showed that repair was more efficient in Caco-2/15-myofibroblast co-cultures than in Caco-2/15 monolayers and involved the deposition of basement membrane components. Functional experiments showed that the addition of type I collagen or human fibronectin to the culture medium significantly accelerated wound closure on epithelial cell co-cultures. This system may provide a new tool to investigate the mechanisms that regulate wound healing in the intestinal epithelium.
Collapse
Affiliation(s)
- Amira Seltana
- CIHR Team on the Digestive Epithelium, Département d'anatomie et de biologie cellulaire, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | | |
Collapse
|
22
|
Nava P, Koch S, Laukoetter MG, Lee WY, Kolegraff K, Capaldo CT, Beeman N, Addis C, Gerner-Smidt K, Neumaier I, Skerra A, Li L, Parkos CA, Nusrat A. Interferon-gamma regulates intestinal epithelial homeostasis through converging beta-catenin signaling pathways. Immunity 2010; 32:392-402. [PMID: 20303298 PMCID: PMC2859189 DOI: 10.1016/j.immuni.2010.03.001] [Citation(s) in RCA: 269] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Revised: 12/16/2009] [Accepted: 01/15/2010] [Indexed: 12/23/2022]
Abstract
Inflammatory cytokines have been proposed to regulate epithelial homeostasis during intestinal inflammation. We report here that interferon-gamma (IFN-gamma) regulates the crucial homeostatic functions of cell proliferation and apoptosis through serine-threonine protein kinase AKT-beta-catenin and Wingless-Int (Wnt)-beta-catenin signaling pathways. Short-term exposure of intestinal epithelial cells to IFN-gamma resulted in activation of beta-catenin through AKT, followed by induction of the secreted Wnt inhibitor Dkk1. Consequently, we observed an increase in Dkk1-mediated apoptosis upon extended IFN-gamma treatment and reduced proliferation through depletion of the Wnt coreceptor LRP6. These effects were enhanced by tumor necrosis factor-alpha (TNF-alpha), suggesting synergism between the two cytokines. Consistent with these results, colitis in vivo was associated with decreased beta-catenin-T cell factor (TCF) signaling, loss of plasma membrane-associated LRP6, and reduced epithelial cell proliferation. Proliferation was partially restored in IFN-gamma-deficient mice. Thus, we propose that IFN-gamma regulates intestinal epithelial homeostasis by sequential regulation of converging beta-catenin signaling pathways.
Collapse
Affiliation(s)
- Porfirio Nava
- Epithelial Pathobiology Unit, Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Preston P, Wartosch L, Günzel D, Fromm M, Kongsuphol P, Ousingsawat J, Kunzelmann K, Barhanin J, Warth R, Jentsch TJ. Disruption of the K+ channel beta-subunit KCNE3 reveals an important role in intestinal and tracheal Cl- transport. J Biol Chem 2010; 285:7165-75. [PMID: 20051516 DOI: 10.1074/jbc.m109.047829] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The KCNE3 beta-subunit constitutively opens outwardly rectifying KCNQ1 (Kv7.1) K(+) channels by abolishing their voltage-dependent gating. The resulting KCNQ1/KCNE3 heteromers display enhanced sensitivity to K(+) channel inhibitors like chromanol 293B. KCNE3 was also suggested to modify biophysical properties of several other K(+) channels, and a mutation in KCNE3 was proposed to underlie forms of human periodic paralysis. To investigate physiological roles of KCNE3, we now disrupted its gene in mice. kcne3(-/-) mice were viable and fertile and displayed neither periodic paralysis nor other obvious skeletal muscle abnormalities. KCNQ1/KCNE3 heteromers are present in basolateral membranes of intestinal and tracheal epithelial cells where they might facilitate transepithelial Cl(-) secretion through basolateral recycling of K(+) ions and by increasing the electrochemical driving force for apical Cl(-) exit. Indeed, cAMP-stimulated electrogenic Cl(-) secretion across tracheal and intestinal epithelia was drastically reduced in kcne3(-/-) mice. Because the abundance and subcellular localization of KCNQ1 was unchanged in kcne3(-/-) mice, the modification of biophysical properties of KCNQ1 by KCNE3 is essential for its role in intestinal and tracheal transport. Further, these results suggest KCNE3 as a potential modifier gene in cystic fibrosis.
Collapse
Affiliation(s)
- Patricia Preston
- Leibniz-Institut für Molekulare Pharmakologie and Max-Delbrück-Centrum für Molekulare Medizin, 13125 Berlin, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Günzel D, Krug SM, Rosenthal R, Fromm M. Biophysical Methods to Study Tight Junction Permeability. CURRENT TOPICS IN MEMBRANES 2010. [DOI: 10.1016/s1063-5823(10)65003-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
25
|
Edelblum KL, Turner JR. The tight junction in inflammatory disease: communication breakdown. Curr Opin Pharmacol 2009; 9:715-20. [PMID: 19632896 PMCID: PMC2788114 DOI: 10.1016/j.coph.2009.06.022] [Citation(s) in RCA: 229] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Revised: 06/25/2009] [Accepted: 06/25/2009] [Indexed: 12/13/2022]
Abstract
The intestinal epithelium restricts free passage of toxic and infectious molecules from the gut lumen while allowing selective paracellular absorption across the tight junction. Inflammatory bowel disease (IBD) patients demonstrate a loss of tight junction barrier function, increased pro-inflammatory cytokine production, and immune dysregulation; however, the relationship between these events is incompletely understood. Although tight junction barrier defects are insufficient to cause experimental IBD, mucosal immune activation is altered in response to increased epithelial permeability. Thus, an evolving model suggests that barrier dysfunction may predispose or enhance disease progression and therapies targeted to specifically restore the barrier function may provide an alternative or supplement to immunology-based therapies.
Collapse
Affiliation(s)
- Karen L. Edelblum
- Department of Pathology, The University of Chicago, 5841 S. Maryland Ave, MC1089, Chicago, IL USA 60637
| | - Jerrold R. Turner
- Department of Pathology, The University of Chicago, 5841 S. Maryland Ave, MC1089, Chicago, IL USA 60637
| |
Collapse
|
26
|
Fromm M, Krug SM, Zeissig S, Richter JF, Rosenthal R, Schulzke JD, Günzel D. High-Resolution Analysis of Barrier Function. Ann N Y Acad Sci 2009; 1165:74-81. [DOI: 10.1111/j.1749-6632.2009.04047.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
27
|
Abstract
PURPOSE OF REVIEW The epithelium makes numerous important contributions to intestinal function. It acts as a physical barrier to prevent pathogenic infection, but allows nutrient uptake and the bidirectional passage of ions and water to lubricate the intestinal lumen while restricting fluid loss. The epithelium mediates communication between the immune system and the commensal flora, and plays a major role in antigen sampling and development of tolerance. After mucosal injury, the epithelium must reestablish its barrier and transport functions for homeostasis to be restored. Here, we will discuss recent advances in our understanding of the roles of the epithelium in intestinal homeostasis. RECENT FINDINGS Mechanisms responsible for epithelial communication with enteric flora and pathogens include the regulation and function of Toll-like receptors and nucleotide-binding oligomerization domain-2, and maintenance and repair of epithelial barrier properties, including the role of growth factors and bacterial peptides in epithelial repair. SUMMARY Recent advances in our understanding of mechanisms by which the gut epithelium modulates, and is modified by, enteric flora and the mucosal immune system illuminate the importance of the epithelium in gut physiology. The work discussed may also identify novel targets that can potentially be modulated therapeutically, either with existing medications or newer agents in development.
Collapse
Affiliation(s)
- Declan F McCole
- School of Medicine, University of California at San Diego, San Diego, California 92093-0063, USA
| | | |
Collapse
|
28
|
Affiliation(s)
- Holger Scholz
- Institut für Physiologie, Charité-Universitätsmedizin Berlin, Tucholskystrasse 2, 10117 Berlin, Germany.
| |
Collapse
|