1
|
Mitra D, Armijo GK, Ober EH, Baker SM, Turner HC, Broustas CG. MIIST305 mitigates gastrointestinal acute radiation syndrome injury and ameliorates radiation-induced gut microbiome dysbiosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619652. [PMID: 39484519 PMCID: PMC11526895 DOI: 10.1101/2024.10.22.619652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
High-dose radiation exposure results in gastrointestinal (GI) acute radiation syndrome identified by the destruction of mucosal layer, intestinal epithelial barrier dysfunction, and aberrant inflammatory responses. In addition, radiation causes gut microbiome dysbiosis characterized by diminished microbial diversity, reduction in the abundance of beneficial commensal bacteria, and the spread of bacterial pathogens that trigger the recruitment of immune cells and the production of pro-inflammatory factors that lead to further GI tissue damage. Currently, there are no FDA- approved countermeasures that can treat radiation-induced GI injury. To meet this critical need, Synedgen Inc ., has developed a glycopolymer radiomitigator (MIIST305) that is specifically targeted to the GI tract which acts by intercalating into the mucus layer and the glycocalyx of intestinal epithelial cells that could potentially ameliorate the deleterious effects of radiation. Male C57BL/6J adult mice were exposed to 13 Gy total body X-irradiation with 5% bone marrow shielding and MIIST305 was administered on days 1, 3, and 5 post-irradiation. Approximately 85% of the animals survived the irradiation exposure and were apparently healthy until the end of the 30-day study period. In contrast, no control, vehicle-treated animals survived past day 10 at this radiation dose. We show that MIIST305 improved intestinal epithelial barrier function and suppressed systemic inflammatory response mediated by radiation-induced pro-inflammatory cytokines. Taxonomic profiling and community structure of the fecal and colonic mucosa microbiota demonstrated that MIIST305 treatment increased microbial diversity and restored abundance of beneficial commensal bacteria, including Lactobacillus and Bifidobacterium genera, while suppressing potentially pathogenic bacteria compared with vehicle-treated animals. In summary, MIIST305 is a novel GI-targeted therapeutic that greatly enhances survival in mice exposed to lethal radiation and protects the GI tract from injury by restoring a balanced gut microbiota and effectively reducing proinflammatory responses. Further development of this drug as an FDA-approved medical countermeasure will be of critical importance in the event of a radiation public health emergency.
Collapse
Affiliation(s)
- Debmalya Mitra
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Gabriel K. Armijo
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Elizabeth H. Ober
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Helen C. Turner
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Constantinos G. Broustas
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
2
|
Borowczak J, Łaszczych D, Olejnik K, Michalski J, Gutowska A, Kula M, Bator A, Sekielska-Domanowska M, Makarewicz R, Marszałek A, Szylberg Ł, Bodnar M. Tight Junctions and Cancer: Targeting Claudin-1 and Claudin-4 in Thyroid Pathologies. Pharmaceuticals (Basel) 2024; 17:1304. [PMID: 39458944 PMCID: PMC11509894 DOI: 10.3390/ph17101304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/22/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Purpose: Claudins are tight junction proteins partaking in epithelial-mesenchymal transition and cancer progression. In this study, we investigated the expression patterns of claudin-1 and claudin-4 in thyroid pathologies, discussed their links with the pathogenesis of thyroid cancers, and reviewed the therapeutic potential of targeting claudins in cancers. Methods: The research group 162 cores of thyroid samples from patients (70 female and 11 male) diagnosed with thyroid adenoma, goiter, papillary, medullary, and anaplastic thyroid cancers. All samples were stained for the expression of claudin-1 and claudin-4, and the analysis of IHC was performed. Results: Goiter samples showed negative claudin-1 and mostly positive expression of claudin-4. Papillary thyroid cancer and thyroid adenoma showed positive expression of claudin-1, while claudin-4 was positive in papillary thyroid cancers, goiters, and adenomas. In The Cancer Genome Atlas cohort, claudin-1 and claudin-4 were overexpressed in papillary thyroid cancer compared to normal thyroid tissues. Patients with high claudin-1 expression had significantly lower 5-year overall survival than patients with low claudin-1 levels (86.75% vs. 98.65, respectively). In multivariate analysis, high claudin-1 expression (HR 7.91, CI 95% 1.79-35, p = 0.006) and advanced clinical stage remained statistically significant prognostic factors of poor prognosis in papillary thyroid cancer. Conclusions: The pattern of claudin-1 staining was pathology-specific and changed between cancers of different histology. This phenomenon may be associated with the different pathogenesis of thyroid cancers and early metastasis. The loss of claudin-1 and claudin-4 characterized more aggressive cancers. Several studies have shown the benefits of targeting claudins in cancers, but their implementation into clinical practice requires further trials.
Collapse
Affiliation(s)
- Jędrzej Borowczak
- Department of Clinical Oncology, Oncology Centre Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland;
| | - Dariusz Łaszczych
- Department of Tumor Pathology and Pathomorphology, Oncology Centre Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland; (D.Ł.); (Ł.S.)
| | - Katarzyna Olejnik
- Chair of Pathology, Dr Jan Biziel University Hospital No. 2, 85-168 Bydgoszcz, Poland
| | - Jakub Michalski
- Chair of Pathology, Dr Jan Biziel University Hospital No. 2, 85-168 Bydgoszcz, Poland
| | - Anna Gutowska
- Department of Tumor Pathology and Pathomorphology, Oncology Centre Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland; (D.Ł.); (Ł.S.)
| | - Monika Kula
- Chair of Pathology, Dr Jan Biziel University Hospital No. 2, 85-168 Bydgoszcz, Poland
| | - Anita Bator
- Department of Tumor Pathology and Pathomorphology, Oncology Centre Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland; (D.Ł.); (Ł.S.)
| | - Marta Sekielska-Domanowska
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum, Nicolaus Copernicus University, 85-168 Bydgoszcz, Poland
| | - Roman Makarewicz
- Department of Oncology and Brachytherapy, Collegium Medicum, Nicolaus Copernicus University, 85-796 Bydgoszcz, Poland
| | - Andrzej Marszałek
- Chair of Oncologic Pathology and Prophylaxis, Poznan University of Medical Sciences and Greater Poland Cancer Center, 61-866 Poznan, Poland
| | - Łukasz Szylberg
- Department of Tumor Pathology and Pathomorphology, Oncology Centre Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland; (D.Ł.); (Ł.S.)
- Chair of Pathology, Dr Jan Biziel University Hospital No. 2, 85-168 Bydgoszcz, Poland
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum, Nicolaus Copernicus University, 85-168 Bydgoszcz, Poland
| | - Magdalena Bodnar
- Chair of Pathology, Dr Jan Biziel University Hospital No. 2, 85-168 Bydgoszcz, Poland
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum, Nicolaus Copernicus University, 85-168 Bydgoszcz, Poland
| |
Collapse
|
3
|
Huang Z, Teng W, Yao L, Xie K, Hang S, He R, Li Y. mTOR signaling pathway regulation HIF-1 α effects on LPS induced intestinal mucosal epithelial model damage. BMC Mol Cell Biol 2024; 25:13. [PMID: 38654163 PMCID: PMC11036631 DOI: 10.1186/s12860-024-00509-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 04/05/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Sepsis-induced small-intestinal injury is associated with increased morbidity and mortality. Our previous study and other papers have shown that HIF-1α has a protective effect on intestinal mucosal injury in septic rats. The purpose of this study is to further verify the protective effect of HIF-1α on intestinal mucosa and its molecular mechanism in vitro experiments. METHODS Caco-2 cells were selected and experiment was divided into 2 parts. Part I: HIF-1α activator and inhibitor were used to treat lipopolysacchrides (LPS)-stimulated Caco-2 cells respectively, to explore the effect of HIF-1α on LPS induced Caco-2 cell epithelial model; Part II: mTOR activator or inhibitor combined with or without HIF-1α activator, inhibitor to treat LPS-stimulated Caco-2 cells respectively, and then the molecular mechanism of HIF-1α reducing LPS induced Caco-2 cell epithelial model damage was detected. RESULTS The results showed that HIF-1α activator decreased the permeability and up regulated tight junction (TJ) expression, while HIF-1α inhibitor had the opposite effect with the HIF-1α activator. mTOR activation increased, while mTOR inhibition decreased HIF-1α protein and expression of its downstream target molecules, which can be attenuated by HIF-1α activator or inhibitor. CONCLUSION This study once again confirmed that HIF-1α alleviates LPS-induced mucosal epithelial model damage through P70S6K signalling pathway. It is of great value to explore whether HIF-2α plays crucial roles in the regulation of mucosal epithelial model functions in the future.
Collapse
Affiliation(s)
- Zeyong Huang
- Department of Anesthesiology, Shulan (Hangzhou) Hospital, Shulan International Medical College, Zhejiang Shuren College, 310015, Hangzhou, China
| | - Wenbin Teng
- Department of Anesthesiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, 310001, Hangzhou, China
| | - Liuxu Yao
- Rehabilitation Medicine Center, Department of Anesthesiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 310014, Hangzhou, China
| | - Kai Xie
- Department of Anesthesiology, Shaoxing People's Hospital, Zhejiang University, 312000, Shaoxing, China
| | - Suqin Hang
- Department of Anesthesiology, Shulan (Hangzhou) Hospital, Shulan International Medical College, Zhejiang Shuren College, 310015, Hangzhou, China
| | - Rui He
- Department of Anesthesiology, Shaoxing People's Hospital, Zhejiang University, 312000, Shaoxing, China.
| | - Yuhong Li
- Department of Anesthesiology, Shulan (Hangzhou) Hospital, Shulan International Medical College, Zhejiang Shuren College, 310015, Hangzhou, China.
- Department of Anesthesiology, Shulan (Hangzhou) Hospital, Shulan International Medical College, Shuren University, 848 Dongxin Road, Xiacheng District, 310004, Hangzhou, Zhejiang, China.
| |
Collapse
|
4
|
Li Y, Ma M, Wang X, Li J, Fang Z, Li J, Yang B, Lu Y, Xu X, Li Y. Celecoxib alleviates the DSS-induced ulcerative colitis in mice by enhancing intestinal barrier function, inhibiting ferroptosis and suppressing apoptosis. Immunopharmacol Immunotoxicol 2024; 46:240-254. [PMID: 38156770 DOI: 10.1080/08923973.2023.2300508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 12/17/2023] [Indexed: 01/03/2024]
Abstract
INTRODUCTION Ulcerative colitis (UC) is an inflammatory intestine disease characterized by dysfunction of the intestinal mucosal barrier, ferroptosis, and apoptosis. Previous researches suggest that celecoxib, a nonsteroidal anti-inflammatory drug, holds promise in alleviating inflammation in UC. Therefore, this study aims to investigate the effects and mechanisms of celecoxib in UC. METHODS To identify ferroptosis-related drugs and genes associated with UC, we utilized the Gene Expression Omnibus (GEO), FerrDb databases, and DGIdb database. Subsequently, we established a 2.5% DSS (Dextran sulfate sodium)-induced colitis model in mice and treated them with 10 mg/kg of celecoxib to validate the bioinformatics results. We evaluated histological pathologies, inflammatory response, intestinal barrier function, ferroptosis markers, and apoptosis regulators. RESULTS Celecoxib treatment significantly ameliorated DSS-induced UC in mice, as evidenced by the body weight change curve, colon length change curve, disease activity index (DAI) score, and histological index score. Celecoxib treatment reduced the level of pro-inflammatory factors and promoted the expressions of intestinal tight junction proteins such as Claudin-1 and Occludin, thereby restoring the integrity of the intestinal mucosal barrier. Furthermore, celecoxib treatment reversed the ferroptosis characteristics in DSS-induced mice by increasing glutathione (GSH), decreasing malondialdehyde (MDA), and increasing the expression of GPX-4 and xCT. Additionally, apoptosis was induced in mice with UC, as evidenced by increased Caspase3, BAD, P53, BAX, Caspase9 and Aifm1 production, and decreased expression of BCL-XL and BCL2. Celecoxib treatment significantly reversed the apoptotic changes in DSS-induced mice. CONCLUSION Our findings suggest that celecoxib effectively treats DSS-induced UC in mice by inhibiting ferroptosis and apoptosis.
Collapse
Affiliation(s)
- Yaxian Li
- General Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mengdi Ma
- General Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaodong Wang
- The Robert Bosch Center for Tumor Diseases (RBCT), Stuttgart, Germany
| | - Jing Li
- General Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ziqing Fang
- General Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jianhui Li
- General Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bo Yang
- General Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yida Lu
- General Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xin Xu
- General Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yongxiang Li
- General Surgery Department, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
5
|
Han LL, Lu QQ, Zheng WW, Li YL, Song YY, Zhang XZ, Long SR, Liu RD, Wang ZQ, Cui J. A novel trypsin of Trichinella spiralis mediates larval invasion of gut epithelium via binding to PAR2 and activating ERK1/2 pathway. PLoS Negl Trop Dis 2024; 18:e0011874. [PMID: 38166153 PMCID: PMC10786404 DOI: 10.1371/journal.pntd.0011874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/12/2024] [Accepted: 12/19/2023] [Indexed: 01/04/2024] Open
Abstract
BACKGROUND Proteases secreted by Trichinella spiralis intestinal infective larvae (IIL) play an important role in larval invasion and pathogenesis. However, the mechanism through which proteases mediate larval invasion of intestinal epithelial cells (IECs) remains unclear. A novel T. spiralis trypsin (TsTryp) was identified in IIL excretory/secretory (ES) proteins. It was an early and highly expressed protease at IIL stage, and had the potential as an early diagnostic antigen. The aim of this study was to investigate the biological characteristics of this novel TsTryp, its role in larval invasion of gut epithelium, and the mechanisms involved. METHODOLOGY/PRINCIPAL FINDING TsTryp with C-terminal domain was cloned and expressed in Escherichia coli BL21 (DE3), and the rTsTryp had the enzymatic activity of natural trypsin, but it could not directly degrade gut tight junctions (TJs) proteins. qPCR and western blotting showed that TsTryp was highly expressed at the invasive IIL stage. Immunofluorescence assay (IFA), ELISA and Far Western blotting revealed that rTsTryp specifically bound to IECs, and confocal microscopy showed that the binding of rTsTryp with IECs was mainly localized in the cytomembrane. Co-immunoprecipitation (Co-IP) confirmed that rTsTryp bound to protease activated receptors 2 (PAR2) in Caco-2 cells. rTsTryp binding to PAR2 resulted in decreased expression levels of ZO-1 and occludin and increased paracellular permeability in Caco-2 monolayers by activating the extracellular regulated protein kinases 1/2 (ERK1/2) pathway. rTsTryp decreased TJs expression and increased epithelial permeability, which could be abrogated by the PAR2 antagonist AZ3451 and ERK1/2 inhibitor PD98059. rTsTryp facilitated larval invasion of IECs, and anti-rTsTryp antibodies inhibited invasion. Both inhibitors impeded larval invasion and alleviated intestinal inflammation in vitro and in vivo. CONCLUSIONS TsTryp binding to PAR2 activated the ERK1/2 pathway, decreased the expression of gut TJs proteins, disrupted epithelial integrity and barrier function, and consequently mediated larval invasion of the gut mucosa. Therefore, rTsTryp could be regarded as a potential vaccine target for blocking T. spiralis invasion and infection.
Collapse
Affiliation(s)
- Lu Lu Han
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Qi Qi Lu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Wen Wen Zheng
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Yang Li Li
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Yan Yan Song
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Xin Zhuo Zhang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Shao Rong Long
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Ruo Dan Liu
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Zhong Quan Wang
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| | - Jing Cui
- Department of Parasitology, Medical College, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
6
|
Nakai D, Miyake M. Intestinal Membrane Function in Inflammatory Bowel Disease. Pharmaceutics 2023; 16:29. [PMID: 38258040 PMCID: PMC10820082 DOI: 10.3390/pharmaceutics16010029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
Inflammatory bowel disease is a set of chronic inflammatory diseases that mainly develop in the gastrointestinal mucosa, including ulcerative colitis and Crohn's disease. Gastrointestinal membrane permeability is an important factor influencing the pharmacological effects of pharmaceuticals administered orally for treating inflammatory bowel disease and other diseases. Understanding the presence or absence of changes in pharmacokinetic properties under a disease state facilitates effective pharmacotherapy. In this paper, we reviewed the gastrointestinal membrane function in ulcerative colitis and Crohn's disease from the perspective of in vitro membrane permeability and electrophysiological parameters. Information on in vivo permeability in humans is summarized. We also overviewed the inflammatory bowel disease research using gut-on-a-chip, in which some advances have recently been achieved. It is expected that these findings will be exploited for the development of therapeutic drugs for inflammatory bowel disease and the optimization of treatment options and regimens.
Collapse
Affiliation(s)
- Daisuke Nakai
- Drug Metabolism & Pharmacokinetics Research Laboratory, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Masateru Miyake
- Pharmapack Co., Ltd., 1-27 Nakaokubo, Toyama 939-2243, Japan;
| |
Collapse
|
7
|
Balda MS, Matter K. Tight junctions. Curr Biol 2023; 33:R1135-R1140. [PMID: 37935122 DOI: 10.1016/j.cub.2023.09.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Various functions within our bodies require the generation and maintenance of compartments with distinct compositions, which in turn necessitate the formation of semipermeable cellular diffusion barriers. For example, the blood-brain barrier protects the brain by allowing only specific molecules to pass through. Another instance is the intestinal barrier, which allows the uptake of essential nutrients, while restricting the passage of pathogenic molecules and bacteria. Breakdown of such barriers causes various pathologies, such as brain or retinal edema, or diarrhoea. Epithelia and endothelia are the most common barrier-forming cells. Individual cells in such barriers are held together by cell-cell adhesion structures - also known as intercellular junctions - that are essential for barrier formation and maintenance. Here, we will focus on the structure and assembly of tight junctions (TJs) and their functions as barriers, but will refer to other adhesive structures crucial for barrier regulation such as adherens junctions (AJs) and focal adhesions to the extracellular matrix (ECM) (Figure 1A,B). We will also discuss additional functions of TJs in cell surface polarity and the regulation of gene expression, cell function, and cell behaviour.
Collapse
Affiliation(s)
- Maria S Balda
- UCL Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK.
| | - Karl Matter
- UCL Institute of Ophthalmology, University College London, Bath Street, London EC1V 9EL, UK.
| |
Collapse
|
8
|
Szabó K, Bolla BS, Erdei L, Balogh F, Kemény L. Are the Cutaneous Microbiota a Guardian of the Skin's Physical Barrier? The Intricate Relationship between Skin Microbes and Barrier Integrity. Int J Mol Sci 2023; 24:15962. [PMID: 37958945 PMCID: PMC10647730 DOI: 10.3390/ijms242115962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
The skin is a tightly regulated, balanced interface that maintains our integrity through a complex barrier comprising physical or mechanical, chemical, microbiological, and immunological components. The skin's microbiota affect various properties, one of which is the establishment and maintenance of the physical barrier. This is achieved by influencing multiple processes, including keratinocyte differentiation, stratum corneum formation, and regulation of intercellular contacts. In this review, we summarize the potential contribution of Cutibacterium acnes to these events and outline the contribution of bacterially induced barrier defects to the pathogenesis of acne vulgaris. With the combined effects of a Westernized lifestyle, microbial dysbiosis, epithelial barrier defects, and inflammation, the development of acne is very similar to that of several other multifactorial diseases of barrier organs (e.g., inflammatory bowel disease, celiac disease, asthma, atopic dermatitis, and chronic rhinosinusitis). Therefore, the management of acne requires a complex approach, which should be taken into account when designing novel treatments that address not only the inflammatory and microbial components but also the maintenance and strengthening of the cutaneous physical barrier.
Collapse
Affiliation(s)
- Kornélia Szabó
- HUN-REN-SZTE Dermatological Research Group, 6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary; (B.S.B.)
- HCEMM-USZ Skin Research Group, 6720 Szeged, Hungary
| | - Beáta Szilvia Bolla
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary; (B.S.B.)
- HCEMM-USZ Skin Research Group, 6720 Szeged, Hungary
| | - Lilla Erdei
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary; (B.S.B.)
- HCEMM-USZ Skin Research Group, 6720 Szeged, Hungary
| | - Fanni Balogh
- HUN-REN-SZTE Dermatological Research Group, 6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary; (B.S.B.)
| | - Lajos Kemény
- HUN-REN-SZTE Dermatological Research Group, 6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary; (B.S.B.)
- HCEMM-USZ Skin Research Group, 6720 Szeged, Hungary
| |
Collapse
|
9
|
Nyimanu D, Behm C, Choudhury S, Yu ASL. The role of claudin-2 in kidney function and dysfunction. Biochem Soc Trans 2023; 51:1437-1445. [PMID: 37387353 DOI: 10.1042/bst20220639] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/01/2023]
Abstract
Claudin-2 is a tight junction protein expressed in leaky epithelia where it forms paracellular pores permeable to cations and water. The paracellular pore formed by claudin-2 is important in energy-efficient cation and water transport in the proximal tubules of the kidneys. Mounting evidence now suggests that claudin-2 may modulate cellular processes often altered in disease, including cellular proliferation. Also, dysregulation of claudin-2 expression has been linked to various diseases, including kidney stone disease and renal cell carcinoma. However, the mechanisms linking altered claudin-2 expression and function to disease are poorly understood and require further investigation. The aim of this review is to discuss the current understanding of the role of claudin-2 in kidney function and dysfunction. We provide a general overview of the claudins and their organization in the tight junction, the expression, and function of claudin-2 in the kidney, and the evolving evidence for its role in kidney disease.
Collapse
Affiliation(s)
- Duuamene Nyimanu
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, U.S.A
| | - Christine Behm
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, U.S.A
| | - Sonali Choudhury
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, U.S.A
| | - Alan S L Yu
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160, U.S.A
| |
Collapse
|
10
|
Rollins J, Worthington T, Dransfield A, Whitney J, Stanford J, Hooke E, Hobson J, Wengler J, Hope S, Mizrachi D. Expression of Cell-Adhesion Molecules in E. coli: A High Throughput Screening to Identify Paracellular Modulators. Int J Mol Sci 2023; 24:9784. [PMID: 37372932 DOI: 10.3390/ijms24129784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Cell-adhesion molecules (CAMs) are responsible for cell-cell, cell-extracellular matrix, and cell-pathogen interactions. Claudins (CLDNs), occludin (OCLN), and junctional adhesion molecules (JAMs) are CAMs' components of the tight junction (TJ), the single protein structure tasked with safeguarding the paracellular space. The TJ is responsible for controlling paracellular permeability according to size and charge. Currently, there are no therapeutic solutions to modulate the TJ. Here, we describe the expression of CLDN proteins in the outer membrane of E. coli and report its consequences. When the expression is induced, the unicellular behavior of E. coli is replaced with multicellular aggregations that can be quantified using Flow Cytometry (FC). Our method, called iCLASP (inspection of cell-adhesion molecules aggregation through FC protocols), allows high-throughput screening (HTS) of small-molecules for interactions with CAMs. Here, we focused on using iCLASP to identify paracellular modulators for CLDN2. Furthermore, we validated those compounds in the mammalian cell line A549 as a proof-of-concept for the iCLASP method.
Collapse
Affiliation(s)
- Jay Rollins
- Department of Cell Biology and Physiology, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA
| | - Tyler Worthington
- Department of Cell Biology and Physiology, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA
| | - Allison Dransfield
- Department of Cell Biology and Physiology, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA
| | - Jordan Whitney
- Department of Cell Biology and Physiology, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA
| | - Jordan Stanford
- Department of Cell Biology and Physiology, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA
| | - Emily Hooke
- Department of Cell Biology and Physiology, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA
| | - Joseph Hobson
- Department of Cell Biology and Physiology, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA
| | - Jacob Wengler
- Department of Cell Biology and Physiology, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA
| | - Sandra Hope
- Department of Microbiology and Molecular Biology, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA
| | - Dario Mizrachi
- Department of Cell Biology and Physiology, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA
| |
Collapse
|
11
|
Capaldo CT. Claudin Barriers on the Brink: How Conflicting Tissue and Cellular Priorities Drive IBD Pathogenesis. Int J Mol Sci 2023; 24:8562. [PMID: 37239907 PMCID: PMC10218714 DOI: 10.3390/ijms24108562] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Inflammatory bowel diseases (IBDs) are characterized by acute or chronic recurring inflammation of the intestinal mucosa, often with increasing severity over time. Life-long morbidities and diminishing quality of life for IBD patients compel a search for a better understanding of the molecular contributors to disease progression. One unifying feature of IBDs is the failure of the gut to form an effective barrier, a core role for intercellular complexes called tight junctions. In this review, the claudin family of tight junction proteins are discussed as they are a fundamental component of intestinal barriers. Importantly, claudin expression and/or protein localization is altered in IBD, leading to the supposition that intestinal barrier dysfunction exacerbates immune hyperactivity and disease. Claudins are a large family of transmembrane structural proteins that constrain the passage of ions, water, or substances between cells. However, growing evidence suggests non-canonical claudin functions during mucosal homeostasis and healing after injury. Therefore, whether claudins participate in adaptive or pathological IBD responses remains an open question. By reviewing current studies, the possibility is assessed that with claudins, a jack-of-all-trades is master of none. Potentially, a robust claudin barrier and wound restitution involve conflicting biophysical phenomena, exposing barrier vulnerabilities and a tissue-wide frailty during healing in IBD.
Collapse
Affiliation(s)
- Christopher T Capaldo
- College of Natural and Computer Sciences, Hawai'i Pacific University, Honolulu, HI 96813, USA
| |
Collapse
|
12
|
Structure Composition and Intracellular Transport of Clathrin-Mediated Intestinal Transmembrane Tight Junction Protein. Inflammation 2023; 46:18-34. [PMID: 36050591 DOI: 10.1007/s10753-022-01724-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/20/2022] [Accepted: 07/27/2022] [Indexed: 11/05/2022]
Abstract
Tight junctions (TJs) are located in the apical region of the junctions between epithelial cells and are widely found in organs such as the brain, retina, intestinal epithelium, and endothelial system. As a mechanical barrier of the intestinal mucosa, TJs can not only maintain the integrity of intestinal epithelial cells but also maintain intestinal mucosal permeability by regulating the entry of ions and molecules into paracellular channels. Therefore, the formation disorder or integrity destruction of TJs can induce damage to the intestinal epithelial barrier, ultimately leading to the occurrence of various gastrointestinal diseases, such as inflammatory bowel disease (IBD), gastroesophageal reflux disease (GERD), and irritable bowel syndrome (IBS). However, a large number of studies have shown that TJs protein transport disorder from the endoplasmic reticulum to the apical membrane can lead to TJs formation disorder, in addition to disruption of TJs integrity caused by external pathological factors and reduction of TJs protein synthesis. In this review, we focus on the structural composition of TJs, the formation of clathrin-coated vesicles containing transmembrane TJs from the Golgi apparatus, and the transport process from the Golgi apparatus to the plasma membrane via microtubules and finally fusion with the plasma membrane. At present, the mechanism of the intracellular transport of TJ proteins remains unclear. More studies are needed in the future to focus on the sorting of TJs protein vesicles, regulation of transport processes, and recycling of TJ proteins, etc.
Collapse
|
13
|
Monteiro L, Delgado L, Amaral B, Ricardo S, Fraga M, Lopes C, Warnakulasuryia S. Occludin and Claudin-1 are potential prognostic biomarkers in patients with oral squamous cell carcinomas. An observational study. Oral Surg Oral Med Oral Pathol Oral Radiol 2022; 134:588-598. [DOI: 10.1016/j.oooo.2022.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/21/2022] [Accepted: 06/22/2022] [Indexed: 11/30/2022]
|
14
|
Moonwiriyakit A, Pathomthongtaweechai N, Steinhagen PR, Chantawichitwong P, Satianrapapong W, Pongkorpsakol P. Tight junctions: from molecules to gastrointestinal diseases. Tissue Barriers 2022; 11:2077620. [PMID: 35621376 PMCID: PMC10161963 DOI: 10.1080/21688370.2022.2077620] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Intestinal epithelium functions as a tissue barrier to prevent interaction between the internal compartment and the external milieu. Intestinal barrier function also determines epithelial polarity for the absorption of nutrients and the secretion of waste products. These vital functions require strong integrity of tight junction proteins. In fact, intestinal tight junctions that seal the paracellular space can restrict mucosal-to-serosal transport of hostile luminal contents. Tight junctions can form both an absolute barrier and a paracellular ion channel. Although defective tight junctions potentially lead to compromised intestinal barrier and the development and progression of gastrointestinal (GI) diseases, no FDA-approved therapies that recover the epithelial tight junction barrier are currently available in clinical practice. Here, we discuss the impacts and regulatory mechanisms of tight junction disruption in the gut and related diseases. We also provide an overview of potential therapeutic targets to restore the epithelial tight junction barrier in the GI tract.
Collapse
Affiliation(s)
- Aekkacha Moonwiriyakit
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Nutthapoom Pathomthongtaweechai
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Peter R Steinhagen
- Department of Hepatology and Gastroenterology, Charité Medical School, Berlin, Germany
| | | | | | - Pawin Pongkorpsakol
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| |
Collapse
|
15
|
Kelley CF, Pollack I, Yacoub R, Zhu Z, Van Doren VE, Gumber S, Amara RR, Fedirko V, Kraft CS, de Man TJB, Hu YJ, Grimsley Ackerley C, Sullivan PS, Bostick RM. Condomless receptive anal intercourse is associated with markers of mucosal inflammation in a cohort of men who have sex with men in Atlanta, Georgia. J Int AIDS Soc 2021; 24:e25859. [PMID: 34911162 PMCID: PMC8673926 DOI: 10.1002/jia2.25859] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 11/25/2021] [Indexed: 11/16/2022] Open
Abstract
Introduction We previously showed that the rectal mucosal immune environment among men who have sex with men (MSM) engaging in condomless receptive anal intercourse (CRAI) is immunologically distinct from that of men who do not engage in anal intercourse (AI). Here, we further examined these differences with quantitative immunohistochemistry to better understand the geographic distribution of immune markers of interest. Methods We enrolled a cohort of MSM engaging in CRAI (n = 41) and men who do not engage in AI (n = 21) between October 2013 and April 2015. Participants were healthy, HIV‐negative men aged 18–45 from the metro Atlanta area. We performed rectal mucosal sampling via rigid sigmoidoscopy during two study visits separated by a median of nine weeks and timed with sexual activity for MSM engaging in CRAI. We used standardized, automated immunohistochemistry and quantitative image analysis to investigate the rectal mucosal distribution of neutrophils (MPO), IL‐17‐producing cells (IL‐17) and Tregs (FOXP3) in the lamina propria, and cellular proliferation (Ki67) and adherens junction protein (E‐cadherin) in the epithelium. We examined associations between biomarker expression and the rectal mucosal microbiota composition by 16s rRNA sequencing. Results Relative to the colonic crypt base, IL‐17, FOXP3, and MPO expression increased towards the rectal lumen, while Ki67 decreased and E‐cadherin was more uniformly distributed. Throughout the rectal mucosa distribution examined, MSM engaging in CRAI had higher mean lamina propria MPO expression (p = 0.04) and epithelial Ki67 (p = 0.04) compared to controls. There were no significant differences in IL‐17, FOXP3 or E‐cadherin expression. We found no significant associations of the five biomarkers with the global rectal microbiota composition or the individual taxa examined. Conclusions Understanding the mucosal distribution of inflammatory mediators can enhance our knowledge of the earliest events in HIV transmission. Neutrophil enrichment and crypt epithelial cell proliferation likely represent sub‐clinical inflammation in response to CRAI in the rectal mucosa of MSM, which could increase the risk for HIV acquisition. However, the contributory role of the microbiota in mucosal inflammation among MSM remains unclear. HIV prevention may be enhanced by interventions that reduce inflammation or capitalize on the presence of specific inflammatory mechanisms during HIV exposure.
Collapse
Affiliation(s)
- Colleen F Kelley
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, The Hope Clinic, Atlanta, Georgia, USA.,Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Ilana Pollack
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, The Hope Clinic, Atlanta, Georgia, USA
| | - Rami Yacoub
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Zhengyi Zhu
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Vanessa E Van Doren
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, The Hope Clinic, Atlanta, Georgia, USA
| | - Sanjeev Gumber
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Rama R Amara
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Veronika Fedirko
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Colleen S Kraft
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, The Hope Clinic, Atlanta, Georgia, USA.,Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Yi-Juan Hu
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Cassie Grimsley Ackerley
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, The Hope Clinic, Atlanta, Georgia, USA
| | - Patrick S Sullivan
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Roberd M Bostick
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
16
|
Watson MJ, Berger PL, Banerjee K, Frank SB, Tang L, Ganguly SS, Hostetter G, Winn M, Miranti CK. Aberrant CREB1 activation in prostate cancer disrupts normal prostate luminal cell differentiation. Oncogene 2021; 40:3260-3272. [PMID: 33846571 PMCID: PMC10760404 DOI: 10.1038/s41388-021-01772-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 03/12/2021] [Accepted: 03/29/2021] [Indexed: 02/02/2023]
Abstract
The molecular mechanisms of luminal cell differentiation are not understood well enough to determine how differentiation goes awry during oncogenesis. Using RNA-Seq analysis, we discovered that CREB1 plays a central role in maintaining new luminal cell survival and that oncogenesis dramatically changes the CREB1-induced transcriptome. CREB1 is active in luminal cells, but not basal cells. We identified ING4 and its E3 ligase, JFK, as CREB1 transcriptional targets in luminal cells. During luminal cell differentiation, transient induction of ING4 expression is followed by a peak in CREB1 activity, while JFK increases concomitantly with CREB1 activation. Transient expression of ING4 is required for luminal cell induction; however, failure to properly down-regulate ING4 leads to luminal cell death. Consequently, blocking CREB1 increased ING4 expression, suppressed JFK, and led to luminal cell death. Thus, CREB1 is responsible for the suppression of ING4 required for luminal cell survival and maintenance. Oncogenic transformation by suppressing PTEN resulted in constitutive activation of CREB1. However, the tumor cells could no longer fully differentiate into luminal cells, failed to express ING4, and displayed a unique CREB1 transcriptome. Blocking CREB1 in tumorigenic cells suppressed tumor growth in vivo, rescued ING4 expression, and restored luminal cell formation, but ultimately induced luminal cell death. IHC of primary prostate tumors demonstrated a strong correlation between loss of ING4 and loss of PTEN. This is the first study to define a molecular mechanism whereby oncogenic loss of PTEN, leading to aberrant CREB1 activation, suppresses ING4 expression causing disruption of luminal cell differentiation.
Collapse
Affiliation(s)
- M J Watson
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI, USA
| | - P L Berger
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI, USA
| | - K Banerjee
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | - S B Frank
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | - L Tang
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | - S S Ganguly
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | - G Hostetter
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI, USA
| | - M Winn
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI, USA
| | - C K Miranti
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI, USA.
- Department of Cellular and Molecular Medicine, University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
17
|
The role of mucosal barriers in human gut health. Arch Pharm Res 2021; 44:325-341. [PMID: 33890250 DOI: 10.1007/s12272-021-01327-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/26/2021] [Indexed: 12/15/2022]
Abstract
The intestinal mucosa is continuously exposed to a large number of commensal or pathogenic microbiota and foreign food antigens. The intestinal epithelium forms a dynamic physicochemical barrier to maintain immune homeostasis. To efficiently absorb nutrients from food, the epithelium in the small intestine has thin, permeable layers spread over a vast surface area. Epithelial cells are renewed from the crypt toward the villi, accompanying epithelial cell death and shedding, to control bacterial colonization. Tight junction and adherens junction proteins provide epithelial cell-cell integrity. Microbial signals are recognized by epithelial cells via toll-like receptors. Environmental signals from short-chain fatty acids derived from commensal microbiota metabolites, aryl hydrocarbon receptors, and hypoxia-induced factors fortify gut barrier function. Here we summarize recent findings regarding various environmental factors for gut barrier function. Further, we discuss the role of gut barriers in the pathogenesis of human intestinal disease and the challenges of therapeutic strategies targeting gut barrier restoration.
Collapse
|
18
|
Li C, Bai X, Liu X, Zhang Y, Liu L, Zhang L, Xu F, Yang Y, Liu M. Disruption of Epithelial Barrier of Caco-2 Cell Monolayers by Excretory Secretory Products of Trichinella spiralis Might Be Related to Serine Protease. Front Microbiol 2021; 12:634185. [PMID: 33815318 PMCID: PMC8013981 DOI: 10.3389/fmicb.2021.634185] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/19/2021] [Indexed: 11/16/2022] Open
Abstract
The physical barrier is composed of epithelial cells which are joined together through intercellular connections. It serves to prevent pathogenic microorganisms from departing the intestinal lumen to invade the host. The excretory secretory (ES) products of Trichinella spiralis are critical for invasion. However, whether ES products of T. spiralis can act on the intestinal barrier is still unknown. In this study, the role of ES products of T. spiralis muscle larvae (Ts-ML-ES) in host invasion was studied by establishing an in vitro cell monolayers model. Barrier integrity analysis by a transmembrane resistance test and a paracellular permeability assay revealed that the Ts-ML-ES was able to destroy barrier function. It occurred via a reduction in the expression of tight junction (TJ) proteins, which was induced by serine protease. Furthermore, Western bolt analysis indicated that Ts-ML-ES reduced the expression of TJ proteins via the MAPK signaling pathway. Based on these data, we conclude that serine protease are likely the main factors from Ts-ML-ES that affect host intestinal barrier integrity by reducing the expression of TJs via the P38-MAPK signaling pathway. Serine protease in Ts-ML-ES might be a key invasion factor in T. spiralis.
Collapse
Affiliation(s)
- Chengyao Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Xue Bai
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Xiaolei Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Yuanyuan Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Lei Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Lixiao Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Fengyan Xu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Yong Yang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Mingyuan Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
19
|
Tripathi A, Hazari PP, Mishra AK, Kumar B, Sagi SSK. Quercetin: a savior of alveolar barrier integrity under hypoxic microenvironment. Tissue Barriers 2021; 9:1883963. [PMID: 33632082 DOI: 10.1080/21688370.2021.1883963] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
High altitude pulmonary edema (HAPE) is generally characterized by the loss of alveolar epithelial barrier integrity. The current study was undertaken to assess the noninvasive approaches of HAPE diagnosis and to evaluate the prophylactic potential of quercetin in preventing alveolar junction impairments. Male SD rats fed with quercetin 1 h prior to hypoxia (7,620 m, for 6 h) were selected. PET/CT imaging was performed to visualize the lung uptake of 18F-FDG in animals under hypoxia. Further, oxidant status, catalase activity, hematological & blood gas parameters were evaluated. Moreover, tight junction (TJ) proteins (ZO-1, JAM-C, Claudin-4, and occludin) expression analysis was accomplished using immune-blotting. The structural differences in lung epithelia were noted by TEM imaging. Quercetin prophylaxis has significantly reduced the FDG uptake in rat lungs under hypoxia. It has also dramatically alleviated the protein oxidation followed by an elevation in catalase activity in the lungs under hypoxia. The TJ protein expression in the lungs has also been restored to normal upon quercetin pre-treatment. Concomitantly, the quercetin preconditioning has elicited the stable blood gas and hematological parameters under hypoxia. The observations from TEM imaging have also implicated the normal lung epithelial structures in the quercetin pretreated animals under hypoxia. Quercetin prophylaxis has significantly restored alveolar epithelium integrity by abating oxidative stress in the lungs under hypoxia.Abbreviations: CT- Computed Tomography18F-FDG- Fluorodeoxyglucose (18FHAPE- High Altitude Pulmonary EdemaHb- HemoglobinHCT- HematocritHCO3- BicarbonateJAM- Junctional Adhesion MoleculeKBq- Killo BecquerelPaO2- Partial pressure of arterial oxygenPaCO2- Partial pressure of arterial carbon di-oxidePET- Positron Emission TomographyRBC- Red Blood CorpusclesSD- Sprague DawleyTJ- Tight JunctionsTEM- Transmission Electron MicroscopyWBC- White Blood CorpusclesZO- Zona Occludin.
Collapse
Affiliation(s)
- Ankit Tripathi
- Hematology Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur Delhi-India
| | - Puja P Hazari
- Molecular Imaging and Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi-India
| | - Anil K Mishra
- Molecular Imaging and Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi-India
| | - Bhuvnesh Kumar
- Hematology Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur Delhi-India
| | - Sarada S K Sagi
- Hematology Division, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur Delhi-India
| |
Collapse
|
20
|
Anwer S, Branchard E, Dan Q, Dan A, Szászi K. Tumor necrosis factor-α induces claudin-3 upregulation in kidney tubular epithelial cells through NF-κB and CREB1. Am J Physiol Cell Physiol 2021; 320:C495-C508. [PMID: 33439776 DOI: 10.1152/ajpcell.00185.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Claudins are essential for tight junction formation and paracellular transport, and they affect key cellular events including proliferation and migration. The properties of tight junctions are dynamically modulated by a variety of inputs. We previously showed that the inflammatory cytokine tumor necrosis factor-α (TNFα), a major pathogenic factor in kidney disease, alters epithelial permeability by affecting the expression of claudin-1, -2, and -4 in kidney tubular cells. Here, we explored the effect of TNFα on claudin-3 (Cldn-3), a ubiquitous barrier-forming protein. We found that TNFα elevated Cldn-3 protein expression in tubular epithelial cells (LLC-PK1 and IMCD3) as early as 3 h post treatment. Bafilomycin A and bortezomib, inhibitors of lysosomal and proteasomes, respectively, reduced Cldn-3 degradation. However, TNFα caused a strong upregulation of Cldn-3 in the presence of bafilomycin, suggesting an effect independent from lysosomes. Blocking protein synthesis using cycloheximide prevented Cldn-3 upregulation by TNFα, verifying the contribution of de novo Cldn-3 synthesis. Indeed, TNFα elevated Cldn-3 mRNA levels at early time points. Using pharmacological inhibitors and siRNA-mediated silencing, we determined that the effect of TNFα on Cldn-3 was mediated by extracellular signal regulated kinase (ERK)-dependent activation of NF-κB and PKA-induced activation of CREB1. These two pathways were turned on by TNFα in parallel and both were required for the upregulation of Cldn-3. Because Cldn-3 was suggested to modulate cell migration and epithelial-mesenchymal transition (EMT), and TNFα was shown to affect these processes, Cldn-3 upregulation may modulate regeneration of the tubules following injury.
Collapse
Affiliation(s)
- Shaista Anwer
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario, Canada
| | - Emily Branchard
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario, Canada
| | - Qinghong Dan
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario, Canada
| | - Angela Dan
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario, Canada
| | - Katalin Szászi
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
21
|
Hopkins EGD, Frankel G. Overview of the Effect of Citrobacter rodentium Infection on Host Metabolism and the Microbiota. Methods Mol Biol 2021; 2291:399-418. [PMID: 33704766 DOI: 10.1007/978-1-0716-1339-9_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Citrobacter rodentium is a natural enteric mouse pathogen that models human intestinal diseases, such as pathogenic E. coli infections, ulcerative colitis, and colon cancer. Upon reaching the monolayer of intestinal epithelial cells (IECs) lining the gut, a complex web of interactions between the host, the pathogen, and the microbiota ensues. A number of studies revealed surprisingly rapid changes in IEC bioenergetics upon infection, involving a switch from oxidative phosphorylation to aerobic glycolysis, leading to mucosal oxygenation and subsequent changes in microbiota composition. Microbiome studies have revealed a bloom in Enterobacteriaceae during C. rodentium infection in both resistant (i.e., C57BL/6) and susceptible (i.e., C3H/HeN) strains of mice concomitant with a depletion of butyrate-producing Clostridia. The emerging understanding that dysbiosis of cholesterol metabolism is induced by enteric infection further confirms the pivotal role immunometabolism plays in disease outcome. Inversely, the host and microbiota also impact upon the progression of infection, from the susceptibility of the distal colon to C. rodentium colonization to clearance of the pathogen, both via opsonization from the host adaptive immune system and out competition by the resident microbiota. Further complicating this compendium of interactions, C. rodentium exploits microbiota metabolites to fine-tune virulence gene expression and promote colonization. This chapter summarizes the current knowledge of the myriad of pathogen-host-microbiota interactions that occur during the progression of C. rodentium infection in mice and the broader implications of these findings on our understanding of enteric disease.
Collapse
Affiliation(s)
- Eve G D Hopkins
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College London, London, UK
| | - Gad Frankel
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Imperial College London, London, UK.
| |
Collapse
|
22
|
Noninvasive Biomarkers of Gut Barrier Function in Patients Suffering from Diarrhea Predominant-IBS: An Update. DISEASE MARKERS 2020; 2020:2886268. [PMID: 33110455 PMCID: PMC7582069 DOI: 10.1155/2020/2886268] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 09/23/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022]
Abstract
The intestinal barrier plays a crucial role in the absorption of nutrients and in preventing the entry of pathogenic microorganisms and toxic molecules. Several studies have shown a compromised intestinal barrier associated with low-grade inflammation in the small intestinal mucosa in celiac disease, inflammatory bowel disease, and irritable bowel syndrome (IBS), particularly in IBS with diarrhea (IBS-D). In light of these new data, IBS is no longer considered a functional disease but rather a heterogeneous syndrome that has yet to be carefully studied. Therefore, investigating the integrity and function of the intestinal barrier is now essential to improving knowledge of the pathophysiology of IBS-D and to improving the management of IBS-D patients. However, the study of the intestinal barrier must clarify some still unsolved methodological aspects and propose standardised assays before becoming a useful diagnostic tool. In this framework, this review will discuss data about the tests that noninvasively evaluate the integrity and functionality of the human intestinal barrier, paying particular attention to patients with IBS-D, in both clinical and research situations.
Collapse
|
23
|
The Compromised Intestinal Barrier Induced by Mycotoxins. Toxins (Basel) 2020; 12:toxins12100619. [PMID: 32998222 PMCID: PMC7600953 DOI: 10.3390/toxins12100619] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/23/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022] Open
Abstract
Mycotoxins are fungal metabolites that occur in human foods and animal feeds, potentially threatening human and animal health. The intestine is considered as the first barrier against these external contaminants, and it consists of interconnected physical, chemical, immunological, and microbial barriers. In this context, based on in vitro, ex vivo, and in vivo models, we summarize the literature for compromised intestinal barrier issues caused by various mycotoxins, and we reviewed events related to disrupted intestinal integrity (physical barrier), thinned mucus layer (chemical barrier), imbalanced inflammatory factors (immunological barrier), and dysfunctional bacterial homeostasis (microbial barrier). We also provide important information on deoxynivalenol, a leading mycotoxin implicated in intestinal dysfunction, and other adverse intestinal effects induced by other mycotoxins, including aflatoxins and ochratoxin A. In addition, intestinal perturbations caused by mycotoxins may also contribute to the development of mycotoxicosis, including human chronic intestinal inflammatory diseases. Therefore, we provide a clear understanding of compromised intestinal barrier induced by mycotoxins, with a view to potentially develop innovative strategies to prevent and treat mycotoxicosis. In addition, because of increased combinatorial interactions between mycotoxins, we explore the interactive effects of multiple mycotoxins in this review.
Collapse
|
24
|
Kim JM, Kim DH, Park HJ, Ma HW, Park IS, Son M, Ro SY, Hong S, Han HK, Lim SJ, Kim SW, Cheon JH. Nanocomposites-based targeted oral drug delivery systems with infliximab in a murine colitis model. J Nanobiotechnology 2020; 18:133. [PMID: 32933548 PMCID: PMC7493402 DOI: 10.1186/s12951-020-00693-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/09/2020] [Indexed: 02/08/2023] Open
Abstract
Background Infliximab (IFX), a TNF-α blocking chimeric monoclonal antibody, induces clinical response and mucosal healing in patients with inflammatory bowel disease (IBD). However, systemic administration of this agent causes unwanted side effects. Oral delivery of antibody therapeutics might be an effective treatment strategy for IBD compared to intravenous administration. Results All three carriers had a high encapsulation efficiency, narrow size distribution, and minimal systemic exposure. There was a higher interaction between nanocomposite carriers and monocytes compared to lymphocytes in the PBMC of IBD patients. Orally administered nanocomposite carriers targeted to inflamed colitis minimized systemic exposure. All IFX delivery formulations with nanocomposite carriers had a significantly less colitis-induced body weight loss, colon shortening and histomorphological score, compared to the DSS-treated group. AC-IFX-L and EAC-IFX-L groups showed significantly higher improvement of the disease activity index, compared to the DSS-treated group. In addition, AC-IFX-L and EAC-IFX-L alleviated pro-inflammatory cytokine expressions (Tnfa, Il1b, and Il17). Conclusion We present orally administered antibody delivery systems which improved efficacy in murine colitis while reducing systemic exposure. These oral delivery systems suggest a promising therapeutic approach for treating IBD.![]()
Collapse
Affiliation(s)
- Jung Min Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Da Hye Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyo Jeong Park
- Department of Integrated Bioscience and Biotechnology, Sejong University, 209 Neungdong‑ro, Gwangjin‑gu, Seoul, South Korea
| | - Hyun Woo Ma
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - I Seul Park
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Mijeong Son
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - So Youn Ro
- Department of Integrated Bioscience and Biotechnology, Sejong University, 209 Neungdong‑ro, Gwangjin‑gu, Seoul, South Korea
| | - Seokmann Hong
- Department of Integrated Bioscience and Biotechnology, Sejong University, 209 Neungdong‑ro, Gwangjin‑gu, Seoul, South Korea.,Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, 05006, Republic of Korea
| | - Hyo Kyung Han
- College of Pharmacy, Dongguk University-Seoul, Dongguk‑ro‑32, Ilsan‑donggu, Goyang, South Korea
| | - Soo Jeong Lim
- Department of Integrated Bioscience and Biotechnology, Sejong University, 209 Neungdong‑ro, Gwangjin‑gu, Seoul, South Korea
| | - Seung Won Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea. .,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea. .,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
| | - Jae Hee Cheon
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea. .,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea. .,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
| |
Collapse
|
25
|
Hall CHT, Lee JS, Murphy EM, Gerich ME, Dran R, Glover LE, Abdulla ZI, Skelton MR, Colgan SP. Creatine Transporter, Reduced in Colon Tissues From Patients With Inflammatory Bowel Diseases, Regulates Energy Balance in Intestinal Epithelial Cells, Epithelial Integrity, and Barrier Function. Gastroenterology 2020; 159:984-998.e1. [PMID: 32433978 PMCID: PMC7891846 DOI: 10.1053/j.gastro.2020.05.033] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 05/04/2020] [Accepted: 05/11/2020] [Indexed: 01/03/2023]
Abstract
BACKGROUND & AIMS Patients with inflammatory bowel diseases (IBDs) have intestinal barrier dysfunction. Creatine regulates energy distribution within cells and reduces the severity of colitis in mice. We studied the functions of the creatine transporter solute carrier family 6 member 8 (SLC6A8, also called CRT) in intestinal epithelial cells (IECs) and mice, and we measured levels in mucosal biopsies from patients with IBD. METHODS Colon biopsy specimens from patients with IBD (30 with Crohn's disease and 27 with ulcerative colitis) and 30 patients without IBD (control individuals) and colon tissues from mice (with and without disruption of Crt) were analyzed by immunofluorescence, immunoblots, and/or quantitative reverse-transcription polymerase chain reaction (qRT-PCR). CRT was knocked down or overexpressed in T84 cells, which were analyzed by immunofluorescence, immunoblots, high-performance liquid chromatography (to measure creatine levels), qRT-PCR, transepithelial electrical resistance, barrier function, actin localization, wound healing, mitochondrial oxygen consumption, and glycolysis extracellular acidification rate assays. Organoids from colon cells of CRT-knockout mice and control mice were analyzed by qRT-PCR, immunoblot, and transepithelial electrical resistance. RESULTS CRT localized around tight junctions (TJs) of T84 IECs. In analyses of IECs with CRT knockdown or overexpression, we found that CRT regulates intracellular creatine, barrier formation, and wound healing. CRT-knockout organoids also had diminished barrier formation. In the absence of adequate creatine, IECs transition toward a stressed, glycolysis-predominant form of metabolism; this resulted in leaky TJs and mislocalization of actin and TJ proteins. Colon tissues from patients with IBD had reduced levels of CRT messenger RNA compared with those from control individuals. CONCLUSIONS In an analysis of IEC cell lines and colonoids derived from CRT-knockout mice, we found that CRT regulates energy balance in IECs and thereby epithelial integrity and barrier function. Mucosal biopsy specimens from patients with ulcerative colitis and inactive Crohn's disease have lower levels of CRT, which might contribute to the reduced barrier function observed in patients with IBD.
Collapse
Affiliation(s)
- Caroline H T Hall
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital Colorado, Aurora, Colorado; Mucosal Inflammation Program, University of Colorado, Aurora, Colorado
| | - J Scott Lee
- Mucosal Inflammation Program, University of Colorado, Aurora, Colorado; Division of Gastroenterology and Hepatology, University of Colorado, Aurora, Colorado
| | - Emily M Murphy
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital Colorado, Aurora, Colorado; Mucosal Inflammation Program, University of Colorado, Aurora, Colorado; Division of Gastroenterology and Hepatology, University of Colorado, Aurora, Colorado
| | - Mark E Gerich
- Mucosal Inflammation Program, University of Colorado, Aurora, Colorado; Division of Gastroenterology and Hepatology, University of Colorado, Aurora, Colorado
| | - Rachael Dran
- Mucosal Inflammation Program, University of Colorado, Aurora, Colorado; Division of Gastroenterology and Hepatology, University of Colorado, Aurora, Colorado
| | - Louis E Glover
- Mucosal Inflammation Program, University of Colorado, Aurora, Colorado; Division of Gastroenterology and Hepatology, University of Colorado, Aurora, Colorado; School of Biochemistry and Immunology, Trinity College Dublin, Ireland
| | - Zuhair I Abdulla
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio; Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, Ohio
| | - Matthew R Skelton
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio; Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, Ohio
| | - Sean P Colgan
- Mucosal Inflammation Program, University of Colorado, Aurora, Colorado; Division of Gastroenterology and Hepatology, University of Colorado, Aurora, Colorado.
| |
Collapse
|
26
|
Pérez AG, Andrade-Da-Costa J, De Souza WF, De Souza Ferreira M, Boroni M, De Oliveira IM, Freire-Neto CA, Fernandes PV, De Lanna CA, Souza-Santos PT, Morgado-Díaz JA, De-Freitas-Junior JCM. N‑glycosylation and receptor tyrosine kinase signaling affect claudin‑3 levels in colorectal cancer cells. Oncol Rep 2020; 44:1649-1661. [PMID: 32945502 PMCID: PMC7448416 DOI: 10.3892/or.2020.7727] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 07/14/2020] [Indexed: 12/14/2022] Open
Abstract
Changes in protein levels in different components of the apical junctional complex occur in colorectal cancer (CRC). Claudin-3 is one of the main constituents of tight junctions, and its overexpression can increase the paracellular flux of macromolecules, as well as the malignant potential of CRC cells. The aim of this study was to investigate the molecular mechanisms involved in the regulation of claudin-3 and its prognostic value in CRC. In silico evaluation in each of the CRC consensus molecular subtypes (CMSs) revealed that high expression levels of CLDN3 (gene encoding claudin-3) in CMS2 and CMS3 worsened the patients' long-term survival, whereas a decrease in claudin-3 levels concomitant with a reduction in phosphorylation levels of epidermal growth factor receptor (EGFR) and insulin-like growth factor 1 receptor (IGF1R) could be achieved by inhibiting N-glycan biosynthesis in CRC cells. We also observed that specific inactivation of these receptor tyrosine kinases (RTKs) led to a decrease in claudin-3 levels, and this regulation seems to be mediated by phospholipase C (PLC) and signal transducer and activator of transcription 3 (STAT3) in CRC cells. RTKs are modulated by their N-linked glycans, and inhibition of N-glycan biosynthesis decreased the claudin-3 levels; therefore, we evaluated the correlation between N-glycogenes and CLDN3 expression levels in each of the CRC molecular subtypes. The CMS1 (MSI immune) subtype concomitantly exhibited low expression levels of CLDN3 and N-glycogenes (MGAT5, ST6GAL1, and B3GNT8), whereas CMS2 (canonical) exhibited high gene expression levels of CLDN3 and N-glycogenes (ST6GAL1 and B3GNT8). A robust positive correlation was also observed between CLDN3 and B3GNT8 expression levels in all CMSs. These results support the hypothesis of a mechanism integrating RTK signaling and N-glycosylation for the regulation of claudin-3 levels in CRC, and they suggest that CLDN3 expression can be used to predict the prognosis of patients identified as CMS2 or CMS3.
Collapse
Affiliation(s)
- Amelia G Pérez
- Cellular and Molecular Oncobiology Program, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Jéssica Andrade-Da-Costa
- Cellular and Molecular Oncobiology Program, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Waldemir F De Souza
- Cellular and Molecular Oncobiology Program, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Michelle De Souza Ferreira
- Cellular and Molecular Oncobiology Program, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Mariana Boroni
- Bioinformatics and Computational Biology Laboratory, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Ivanir M De Oliveira
- Pathology Division, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Carlos A Freire-Neto
- Cellular and Molecular Oncobiology Program, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Priscila V Fernandes
- Pathology Division, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | - Cristóvão A De Lanna
- Bioinformatics and Computational Biology Laboratory, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | | | - José A Morgado-Díaz
- Cellular and Molecular Oncobiology Program, National Cancer Institute (INCA), Rio de Janeiro, RJ 20231‑050, Brazil
| | | |
Collapse
|
27
|
Bolla BS, Erdei L, Urbán E, Burián K, Kemény L, Szabó K. Cutibacterium acnes regulates the epidermal barrier properties of HPV-KER human immortalized keratinocyte cultures. Sci Rep 2020; 10:12815. [PMID: 32733073 PMCID: PMC7393503 DOI: 10.1038/s41598-020-69677-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/13/2020] [Indexed: 12/20/2022] Open
Abstract
Our skin provides a physical barrier to separate the internal part of our body from the environment. Maintenance of complex barrier functions is achieved through anatomical structures in the skin, the stratified squamous epithelium specialized junctional organelles, called tight junctions (TJs). Several members of our microbial communities are known to affect the differentiation state and function of the colonized organ. Whether and how interactions between skin cells and cutaneous microbes, including Cutibacterium acnes (C. acnes), modify the structure and/or function of our skin is currently only partly understood. Thus, in our studies, we investigated whether C. acnes may affect the epidermal barrier using in vitro model systems. Real-time cellular analysis showed that depending on the keratinocyte differentiation state, the applied C. acnes strains and their dose, the measured impedance values change, together with the expression of selected TJ proteins. These may reflect barrier alterations, which can be partially restored upon antibiotic–antimycotic treatment. Our findings suggest that C. acnes can actively modify the barrier properties of cultured keratinocytes, possibly through alteration of tight cell-to-cell contacts. Similar events may play important roles in our skin, in the maintenance of cutaneous homeostasis.
Collapse
Affiliation(s)
- Beáta Szilvia Bolla
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary.,HCEMM-SZTE Skin Research Group, Szeged, Hungary
| | - Lilla Erdei
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary.,HCEMM-SZTE Skin Research Group, Szeged, Hungary
| | - Edit Urbán
- Department of Public Health, University of Szeged, Szeged, Hungary
| | - Katalin Burián
- Institute of Clinical Microbiology, University of Szeged, Szeged, Hungary
| | - Lajos Kemény
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary.,HCEMM-SZTE Skin Research Group, Szeged, Hungary.,MTA-SZTE Dermatological Research Group, Szeged, Hungary
| | - Kornélia Szabó
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary. .,MTA-SZTE Dermatological Research Group, Szeged, Hungary.
| |
Collapse
|
28
|
Ono K, Sogawa C, Kawai H, Tran MT, Taha EA, Lu Y, Oo MW, Okusha Y, Okamura H, Ibaragi S, Takigawa M, Kozaki KI, Nagatsuka H, Sasaki A, Okamoto K, Calderwood SK, Eguchi T. Triple knockdown of CDC37, HSP90-alpha and HSP90-beta diminishes extracellular vesicles-driven malignancy events and macrophage M2 polarization in oral cancer. J Extracell Vesicles 2020; 9:1769373. [PMID: 33144925 PMCID: PMC7580842 DOI: 10.1080/20013078.2020.1769373] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Evidence has been accumulating to indicate that extracellular vesicles (EVs), including exosomes, released by cancer cells can foster tumour progression. The molecular chaperones – CDC37, HSP90α and HSP90β play key roles in cancer progression including epithelial-mesenchymal transition (EMT), although their contribution to EVs-mediated cell–cell communication in tumour microenvironment has not been thoroughly examined. Here we show that triple depletion of the chaperone trio attenuates numerous cancer malignancy events exerted through EV release. Metastatic oral cancer-derived EVs (MEV) were enriched with HSP90α HSP90β and cancer-initiating cell marker CD326/EpCAM. Depletion of these chaperones individually induced compensatory increases in the other chaperones, whereas triple siRNA targeting of these molecules markedly diminished the levels of the chaperone trio and attenuated EMT. MEV were potent agents in initiating EMT in normal epithelial cells, a process that was attenuated by the triple chaperone depletion. The migration, invasion, and in vitro tumour initiation of oral cancer cells were significantly promoted by MEV, while triple depletion of CDC37/HSP90α/β reversed these MEV-driven malignancy events. In metastatic oral cancer patient-derived tumours, HSP90β was significantly accumulated in infiltrating tumour-associated macrophages (TAM) as compared to lower grade oral cancer cases. HSP90-enriched MEV-induced TAM polarization to an M2 phenotype, a transition known to support cancer progression, whereas the triple chaperone depletion attenuated this effect. Mechanistically, the triple chaperone depletion in metastatic oral cancer cells effectively reduced MEV transmission into macrophages. Hence, siRNA-mediated knockdown of the chaperone trio (CDC37/HSP90α/HSP90β) could potentially be a novel therapeutic strategy to attenuate several EV-driven malignancy events in the tumour microenvironment. Abbreviations CDC37: cell division control 37; EMT: epithelial-mesenchymal transmission; EV: extracellular vesicles; HNSCC: head and neck squamous cell carcinoma; HSP90: heat shock protein 90; TAM: tumour-associated macrophage
Collapse
Affiliation(s)
- Kisho Ono
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan.,Department of Oral and Maxillofacial Surgery, Okayama University Hospital, Okayama, Japan
| | - Chiharu Sogawa
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hotaka Kawai
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Manh Tien Tran
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Eman A Taha
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yanyin Lu
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - May Wathone Oo
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yuka Okusha
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hirohiko Okamura
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Soichiro Ibaragi
- Department of Oral and Maxillofacial Surgery, Okayama University Hospital, Okayama, Japan
| | - Masaharu Takigawa
- Advanced Research Center for Oral and Craniofacial Sciences, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ken-Ichi Kozaki
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hitoshi Nagatsuka
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.,Advanced Research Center for Oral and Craniofacial Sciences, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Akira Sasaki
- Department of Oral and Maxillofacial Surgery, Okayama University Hospital, Okayama, Japan.,Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kuniaki Okamoto
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Stuart K Calderwood
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Takanori Eguchi
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan.,Advanced Research Center for Oral and Craniofacial Sciences, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
29
|
Liu P, Bian Y, Fan Y, Zhong J, Liu Z. Protective Effect of Naringin on In Vitro Gut-Vascular Barrier Disruption of Intestinal Microvascular Endothelial Cells Induced by TNF-α. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:168-175. [PMID: 31850758 DOI: 10.1021/acs.jafc.9b06347] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Naringin is a polymethoxylated flavonoid commonly found in citrus species and has therapeutic potential in intestinal disorders. However, the effect and mechanism of naringin on gut-vascular barrier disruption has not yet been reported. This study aimed to investigate the distinguishing and selectively protective effects of naringin on tumor necrosis factor (TNF)-α-induced gut-vascular barrier disruption and elucidate the potential mechanism. In the present study, an in vitro gut-vascular barrier model composed of rat intestinal microvascular endothelial cells (RIMVECs) was studied. Evans blue-albumin efflux assay showed that naringin (50 μM) evidently protected the integrity of RIMVEC monolayer barriers against TNF-α-induced disruption. Naringin maintained the expression and distribution of tight junction proteins including zona occludin-1, occludin, claudin-1, and claudin-2. Additionally, naringin protected RIMVECs from TNF-α-induced apoptosis and cell migration suppression (41.1 ± 2.2 vs 51.1 ± 3.5%; 61.0 ± 5.1 vs 72.2 ± 6.2%). Our results indicate that naringin effectively ameliorates gut-vascular barrier disruption.
Collapse
Affiliation(s)
| | | | - Yingsai Fan
- College of Traditional Chinese Veterinary Medicine , Hebei Agricultural University , No. 289 Lingyusi Street , Baoding , Heibei 071001 , People's Republic of China
| | | | | |
Collapse
|
30
|
Wang X, Wang H, Zhang R, Li D, Gao MQ. LRRC75A antisense lncRNA1 knockout attenuates inflammatory responses of bovine mammary epithelial cells. Int J Biol Sci 2020; 16:251-263. [PMID: 31929753 PMCID: PMC6949150 DOI: 10.7150/ijbs.38214] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 10/14/2019] [Indexed: 12/12/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) play multiple key roles during inflammatory processes. In this study, a novel lncRNA identified by the high-throughput sequencing analysis was found significantly down-regulated in Escherichia coli-introduced cell model of bovine mastitis. Given that this lncRNA consists of the antisense of leucine-rich repeat-containing protein 75A (LRRC75A), it was named LRRC75A antisense lncRNA1 (LRRC75A-AS1). The expression of LRRC75A-AS1 was down-regulated in bovine mammary epithelial cells and mammary tissues under inflammatory condition. Knockout (KO) of LRRC75A-AS1 by CRISPR-Cas9 system in bovine mammary alveolar cell-T (MAC-T) cell line could enhance expressions of tight junction (TJ) proteins Claudin-1, Occludin and ZO-1, reduce cell monolayer permeability, and inhibit Staphylococcus aureus adhesion and invasion. Meanwhile, it also down-regulated expressions of inflammatory factors and attenuated activation of NF-κB pathway. Similarly, knockdown of LRRC75A caused the changes as LRRC75A-AS1 KO did, while overexpression of LRRC75A enabled the opposite effects. TJ of epithelioid cells barriers the pathogenic microorganisms outside during inflammation, in which LRRC75A-AS1 can regulate the expression of TJ proteins through LRRC75A, affecting the development of inflammation.
Collapse
Affiliation(s)
- Xixi Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Hao Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Ruiqi Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Dan Li
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Ming-Qing Gao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
31
|
Claudin-1 upregulation is associated with favorable tumor features and a reduced risk for biochemical recurrence in ERG-positive prostate cancer. World J Urol 2019; 38:2185-2196. [PMID: 31745645 DOI: 10.1007/s00345-019-03017-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/07/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Claudin-1 is a membrane-tight junction protein and important for the sealing of the paracellular cleft in epithelial and endothelial cells. Differential expression of Claudin-1 is linked to disease outcome in various cancers. MATERIAL AND METHODS To evaluate the potential relevance of Claudin-1 expression in prostate cancer, a tissue microarray containing samples of 17,747 tumors with annotated clinico-pathological and molecular data was immunohistochemically analyzed for Claudin-1 expression. RESULTS In normal prostate, glandular cells were always Claudin-1-negative while there was a strong staining of gland-surrounding basal cells. In contrast to normal prostatic glands, a positive Claudin-1 immunostaining, was found, however, in 38.7% of 12,441 interpretable cancers and was considered weak in 12.7%, moderate in 13.2%, and strong in 12.8% of cases. Positive Claudin-1 immunostaining was associated with favorable tumor features like low pT (p = 0.0032), low Gleason grade (p< 0.0001), and a reduced risk of PSA recurrence (p = 0.0005). A positive Claudin-1 staining was markedly more frequent in ERG-positive (63%) than in ERG-negative cancers (23%; p < 0.0001). Subset analyses revealed that all associations of Claudin-1 expression and favorable phenotype and prognosis were driven by ERG-positive cancers. Multivariate analyses revealed, however, that even in ERG-positive cancers, the prognostic impact of high Claudin-1 expression was not independent of established clinico-pathological parameters. Comparison with 12 previously analyzed chromosomal deletions identified conspicuous associations with PTEN and 12p13 deletions potentially indicating functional interactions. CONCLUSION These data identify a peculiar role for Claudin-1 in prostate cancer. The protein is overexpressed in a fraction of prostate cancers and increased Claudin-1 expression levels predict a favorable prognosis in ERG-positive cancer.
Collapse
|
32
|
Venugopal S, Anwer S, Szászi K. Claudin-2: Roles beyond Permeability Functions. Int J Mol Sci 2019; 20:ijms20225655. [PMID: 31726679 PMCID: PMC6888627 DOI: 10.3390/ijms20225655] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 11/08/2019] [Accepted: 11/09/2019] [Indexed: 12/12/2022] Open
Abstract
Claudin-2 is expressed in the tight junctions of leaky epithelia, where it forms cation-selective and water permeable paracellular channels. Its abundance is under fine control by a complex signaling network that affects both its synthesis and turnover in response to various environmental inputs. Claudin-2 expression is dysregulated in many pathologies including cancer, inflammation, and fibrosis. Claudin-2 has a key role in energy-efficient ion and water transport in the proximal tubules of the kidneys and in the gut. Importantly, strong evidence now also supports a role for this protein as a modulator of vital cellular events relevant to diseases. Signaling pathways that are overactivated in diseases can alter claudin-2 expression, and a good correlation exists between disease stage and claudin-2 abundance. Further, loss- and gain-of-function studies showed that primary changes in claudin-2 expression impact vital cellular processes such as proliferation, migration, and cell fate determination. These effects appear to be mediated by alterations in key signaling pathways. The specific mechanisms linking claudin-2 to these changes remain poorly understood, but adapters binding to the intracellular portion of claudin-2 may play a key role. Thus, dysregulation of claudin-2 may contribute to the generation, maintenance, and/or progression of diseases through both permeability-dependent and -independent mechanisms. The aim of this review is to provide an overview of the properties, regulation, and functions of claudin-2, with a special emphasis on its signal-modulating effects and possible role in diseases.
Collapse
|
33
|
Soroosh A, Rankin CR, Polytarchou C, Lokhandwala ZA, Patel A, Chang L, Pothoulakis C, Iliopoulos D, Padua DM. miR-24 Is Elevated in Ulcerative Colitis Patients and Regulates Intestinal Epithelial Barrier Function. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1763-1774. [PMID: 31220450 PMCID: PMC6723227 DOI: 10.1016/j.ajpath.2019.05.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/01/2019] [Accepted: 05/28/2019] [Indexed: 02/07/2023]
Abstract
Inflammatory bowel disease is characterized by high levels of inflammation and loss of barrier integrity in the colon. The intestinal barrier is a dynamic network of proteins that encircle intestinal epithelial cells. miRNAs regulate protein-coding genes. In this study, miR-24 was found to be elevated in colonic biopsies and blood samples from ulcerative colitis (UC) patients compared with healthy controls. In the colon of UC patients, miR-24 is localized to intestinal epithelial cells, which prompted an investigation of intestinal epithelial barrier function. Two intestinal epithelial cell lines were used to study the effect of miR-24 overexpression on barrier integrity. Overexpression of miR-24 in both cell lines led to diminished transepithelial electrical resistance and increased dextran flux, suggesting an effect on barrier integrity. Overexpression of miR-24 did not induce apoptosis or affect cell proliferation, suggesting that the effect of miR-24 on barrier function was due to an effect on cell-cell junctions. Although the tight junctions in cells overexpressing miR-24 appeared normal, miR-24 overexpression led to a decrease in the tight junction-associated protein cingulin. Loss of cingulin compromised barrier formation; cingulin levels negatively correlated with disease severity in UC patients. Together, these data suggest that miR-24 is a significant regulator of intestinal barrier that may be important in the pathogenesis of UC.
Collapse
Affiliation(s)
- Artin Soroosh
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Carl R Rankin
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Christos Polytarchou
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Zulfiqar A Lokhandwala
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Ami Patel
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Lin Chang
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Charalabos Pothoulakis
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Dimitrios Iliopoulos
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - David M Padua
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, Los Angeles, California; Division of Gastroenterology, Hepatology, and Parenteral Nutrition, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California.
| |
Collapse
|
34
|
Thomson A, Smart K, Somerville MS, Lauder SN, Appanna G, Horwood J, Sunder Raj L, Srivastava B, Durai D, Scurr MJ, Keita ÅV, Gallimore AM, Godkin A. The Ussing chamber system for measuring intestinal permeability in health and disease. BMC Gastroenterol 2019; 19:98. [PMID: 31221083 PMCID: PMC6585111 DOI: 10.1186/s12876-019-1002-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 05/28/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The relationship between intestinal epithelial integrity and the development of intestinal disease is of increasing interest. A reduction in mucosal integrity has been associated with ulcerative colitis, Crohn's disease and potentially could have links with colorectal cancer development. The Ussing chamber system can be utilised as a valuable tool for measuring gut integrity. Here we describe step-by-step methodology required to measure intestinal permeability of both mouse and human colonic tissue samples ex vivo, using the latest equipment and software. This system can be modified to accommodate other tissues. METHODS An Ussing chamber was constructed and adapted to support both mouse and human tissue to measure intestinal permeability, using paracellular flux and electrical measurements. Two mouse models of intestinal inflammation (dextran sodium sulphate treatment and T regulatory cell depletion using C57BL/6-FoxP3DTR mice) were used to validate the system along with human colonic biopsy samples. RESULTS Distinct regional differences in permeability were consistently identified within mouse and healthy human colon. In particular, mice showed increased permeability in the mid colonic region. In humans the left colon is more permeable than the right. Furthermore, inflammatory conditions induced chemically or due to autoimmunity reduced intestinal integrity, validating the use of the system. CONCLUSIONS The Ussing chamber has been used for many years to measure barrier function. However, a clear and informative methods paper describing the setup of modern equipment and step-by-step procedure to measure mouse and human intestinal permeability isn't available. The Ussing chamber system methodology we describe provides such detail to guide investigation of gut integrity.
Collapse
Affiliation(s)
- Amanda Thomson
- Institute of Infection and Immunity, School of Medicine, Cardiff, UK
- Present address: Institute of Cellular Medicine, Newcastle University, Newcastle Upon Tyne, UK
| | - Kathryn Smart
- Institute of Infection and Immunity, School of Medicine, Cardiff, UK
| | | | - Sarah N. Lauder
- Institute of Infection and Immunity, School of Medicine, Cardiff, UK
| | - Gautham Appanna
- Institute of Infection and Immunity, School of Medicine, Cardiff, UK
- Department of Gastroenterology and Hepatology, University Hospital Wales, Cardiff, UK
| | - James Horwood
- Department of Surgery, University Hospital Wales, Cardiff, UK
| | - Lawrence Sunder Raj
- Department of Gastroenterology and Hepatology, University Hospital Wales, Cardiff, UK
| | - Brijesh Srivastava
- Department of Gastroenterology and Hepatology, University Hospital Wales, Cardiff, UK
| | - Dharmaraj Durai
- Department of Gastroenterology and Hepatology, University Hospital Wales, Cardiff, UK
| | - Martin J. Scurr
- Institute of Infection and Immunity, School of Medicine, Cardiff, UK
| | - Åsa V. Keita
- Division of Surgery, Orthopedics & Oncology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Awen M. Gallimore
- Institute of Infection and Immunity, School of Medicine, Cardiff, UK
| | - Andrew Godkin
- Institute of Infection and Immunity, School of Medicine, Cardiff, UK
- Department of Gastroenterology and Hepatology, University Hospital Wales, Cardiff, UK
| |
Collapse
|
35
|
Berndt P, Winkler L, Cording J, Breitkreuz-Korff O, Rex A, Dithmer S, Rausch V, Blasig R, Richter M, Sporbert A, Wolburg H, Blasig IE, Haseloff RF. Tight junction proteins at the blood-brain barrier: far more than claudin-5. Cell Mol Life Sci 2019; 76:1987-2002. [PMID: 30734065 PMCID: PMC11105330 DOI: 10.1007/s00018-019-03030-7] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/15/2019] [Accepted: 01/28/2019] [Indexed: 10/27/2022]
Abstract
At the blood-brain barrier (BBB), claudin (Cldn)-5 is thought to be the dominant tight junction (TJ) protein, with minor contributions from Cldn3 and -12, and occludin. However, the BBB appears ultrastructurally normal in Cldn5 knock-out mice, suggesting that further Cldns and/or TJ-associated marvel proteins (TAMPs) are involved. Microdissected human and murine brain capillaries, quickly frozen to recapitulate the in vivo situation, showed high transcript expression of Cldn5, -11, -12, and -25, and occludin, but also abundant levels of Cldn1 and -27 in man. Protein levels were quantified by a novel epitope dilution assay and confirmed the respective mRNA data. In contrast to the in vivo situation, Cldn5 dominates BBB expression in vitro, since all other TJ proteins are at comparably low levels or are not expressed. Cldn11 was highly abundant in vivo and contributed to paracellular tightness by homophilic oligomerization, but almost disappeared in vitro. Cldn25, also found at high levels, neither tightened the paracellular barrier nor interconnected opposing cells, but contributed to proper TJ strand morphology. Pathological conditions (in vivo ischemia and in vitro hypoxia) down-regulated Cldn1, -3, and -12, and occludin in cerebral capillaries, which was paralleled by up-regulation of Cldn5 after middle cerebral artery occlusion in rats. Cldn1 expression increased after Cldn5 knock-down. In conclusion, this complete Cldn/TAMP profile demonstrates the presence of up to a dozen TJ proteins in brain capillaries. Mouse and human share a similar and complex TJ profile in vivo, but this complexity is widely lost under in vitro conditions.
Collapse
Affiliation(s)
- Philipp Berndt
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Lars Winkler
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125, Berlin, Germany.
| | - Jimmi Cording
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Olga Breitkreuz-Korff
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - André Rex
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Sophie Dithmer
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Valentina Rausch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Rosel Blasig
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Matthias Richter
- Max-Delbrück-Centrum für Molekulare Medizin, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Anje Sporbert
- Max-Delbrück-Centrum für Molekulare Medizin, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Hartwig Wolburg
- Institut für Pathologie und Neuropathologie, Universität Tübingen, Liebermeisterstraße 8, 72076, Tübingen, Germany
| | - Ingolf E Blasig
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Reiner F Haseloff
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125, Berlin, Germany.
| |
Collapse
|
36
|
Tervonen A, Ihalainen TO, Nymark S, Hyttinen J. Structural dynamics of tight junctions modulate the properties of the epithelial barrier. PLoS One 2019; 14:e0214876. [PMID: 30964903 PMCID: PMC6456171 DOI: 10.1371/journal.pone.0214876] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/21/2019] [Indexed: 01/09/2023] Open
Abstract
Tight junctions are dynamic structures that are crucial in establishing the diffusion and electrical barrier of epithelial monolayers. Dysfunctions in the tight junctions can impede this barrier function and lead to many pathological conditions. Unfortunately, detailed understanding of the non-specific permeation pathway through the tight junctions, the so-called leak pathway, is lacking. We created computational models of the leak pathway to describe the two main barrier measures, molecular permeability and transepithelial electric resistance while using common structural dynamics. Our results showed that the proposed alternatives for the leak pathway, the bicellular strand opening dynamics and the tricellular pores, contribute together with distinct degrees, depending on the epithelium. The models can also capture changes in the tight junction barrier caused by changes in tight junction protein composition. In addition, we observed that the molecular permeability was markedly more sensitive to changes in the tight junction structure and strand dynamics compared with transepithelial electric resistance. The results highlight that our model creates a good methodological framework to integrate knowledge on the tight junction structure as well as to provide insights and tools to advance tight junction research.
Collapse
Affiliation(s)
- Aapo Tervonen
- Faculty of Medicine and Health Technology and BioMediTech Institute, Tampere University, Tampere, Finland
- * E-mail:
| | - Teemu O. Ihalainen
- Faculty of Medicine and Health Technology and BioMediTech Institute, Tampere University, Tampere, Finland
| | - Soile Nymark
- Faculty of Medicine and Health Technology and BioMediTech Institute, Tampere University, Tampere, Finland
| | - Jari Hyttinen
- Faculty of Medicine and Health Technology and BioMediTech Institute, Tampere University, Tampere, Finland
| |
Collapse
|
37
|
Pyo JS, Kim NY, Cho WJ. Prognostic Role of Claudin-1 Immunohistochemistry in Malignant Solid Tumors: A Meta-Analysis. J Pathol Transl Med 2019; 53:173-179. [PMID: 30832458 PMCID: PMC6527940 DOI: 10.4132/jptm.2019.02.03] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 02/03/2019] [Indexed: 01/01/2023] Open
Abstract
Background Although the correlation between low claudin-1 expression and worse prognosis has been reported, details on the prognostic implications of claudin-1 expression in various malignant tumors remain unclear. The present study aimed to elucidate the prognostic roles of claudin- 1 immunohistochemistry (IHC) in various malignant tumors through a meta-analysis. Methods The study included 2,792 patients from 22 eligible studies for assessment of the correlation between claudin-1 expression and survival rate in various malignant tumors. A subgroup analysis based on the specific tumor and evaluation criteria of claudin-1 IHC was conducted. Results Low claudin-1 expression was significantly correlated with worse overall survival (OS) (hazard ratio [HR], 1.851; 95% confidence interval [CI], 1.506 to 2.274) and disease-free survival (DFS) (HR, 2.028; 95% CI, 1.313 to 3.134) compared to high claudin-1 expression. Breast, colorectal, esophageal, gallbladder, head and neck, and lung cancers, but not cervical, liver or stomach cancers, were significantly correlated with worse OS. Breast, colorectal, esophageal, and thyroid cancers with low claudin-1 expression were associated with poorer DFS. In the lower cut-off subgroup (< 25.0%) with respect to claudin-1 IHC, low claudin-1 expression was significantly correlated with worse OS and DFS. Conclusions Taken together, low claudin-1 IHC expression is significantly correlated with worse survival in various malignant tumors. More detailed criteria for claudin-1 IHC expression in various malignant tumors are needed for application in daily practice.
Collapse
Affiliation(s)
- Jung-Soo Pyo
- Departments of Pathology, Eulji University Hospital, Eulji University School of Medicine, Daejeon, Korea
| | - Nae Yu Kim
- Departments of Internal Medicine, Eulji University Hospital, Eulji University School of Medicine, Daejeon, Korea
| | - Won Jin Cho
- Department of Urology, Chosun University Hospital, Chosun University School of Medicine, Gwangju, Korea
| |
Collapse
|
38
|
Deng Z, Chen M, Xie H, Jian D, Xu S, Peng Q, Sha K, Liu Y, Zhang Y, Shi W, Li J. Claudin reduction may relate to an impaired skin barrier in rosacea. J Dermatol 2019; 46:314-321. [PMID: 30714633 DOI: 10.1111/1346-8138.14792] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/03/2019] [Indexed: 12/28/2022]
Affiliation(s)
- Zhili Deng
- Department of Dermatology Xiangya Hospital Central South University Changsha China
- Center for Molecular Medicine Xiangya Hospital Central South University Changsha China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province Central South University Changsha China
| | - Mengting Chen
- Department of Dermatology Xiangya Hospital Central South University Changsha China
- Center for Molecular Medicine Xiangya Hospital Central South University Changsha China
| | - Hongfu Xie
- Department of Dermatology Xiangya Hospital Central South University Changsha China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province Central South University Changsha China
| | - Dan Jian
- Department of Dermatology Xiangya Hospital Central South University Changsha China
| | - San Xu
- Department of Dermatology Xiangya Hospital Central South University Changsha China
- Center for Molecular Medicine Xiangya Hospital Central South University Changsha China
| | - Qinqin Peng
- Department of Dermatology Xiangya Hospital Central South University Changsha China
- Center for Molecular Medicine Xiangya Hospital Central South University Changsha China
| | - Ke Sha
- Department of Dermatology Xiangya Hospital Central South University Changsha China
- Center for Molecular Medicine Xiangya Hospital Central South University Changsha China
| | - Yingzi Liu
- Department of Dermatology Xiangya Hospital Central South University Changsha China
- Center for Molecular Medicine Xiangya Hospital Central South University Changsha China
| | - Yiya Zhang
- Department of Dermatology Xiangya Hospital Central South University Changsha China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province Central South University Changsha China
| | - Wei Shi
- Department of Dermatology Xiangya Hospital Central South University Changsha China
| | - Ji Li
- Department of Dermatology Xiangya Hospital Central South University Changsha China
- Center for Molecular Medicine Xiangya Hospital Central South University Changsha China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province Central South University Changsha China
| |
Collapse
|
39
|
Abstract
Redox signalling in the gastrointestinal mucosa is held in an intricate balance. Potent microbicidal mechanisms can be used by infiltrating immune cells, such as neutrophils, to protect compromised mucosae from microbial infection through the generation of reactive oxygen species. Unchecked, collateral damage to the surrounding tissue from neutrophil-derived reactive oxygen species can be detrimental; thus, maintenance and restitution of a breached intestinal mucosal barrier are paramount to host survival. Redox reactions and redox signalling have been studied for decades with a primary focus on contributions to disease processes. Within the past decade, an upsurge of exciting findings have implicated subtoxic levels of oxidative stress in processes such as maintenance of mucosal homeostasis, the control of protective inflammation and even regulation of tissue wound healing. Resident gut microbial communities have been shown to trigger redox signalling within the mucosa, which expresses similar but distinct enzymes to phagocytes. At the fulcrum of this delicate balance is the colonic mucosal epithelium, and emerging evidence suggests that precise control of redox signalling by these barrier-forming cells may dictate the outcome of an inflammatory event. This Review will address both the spectrum and intensity of redox activity pertaining to host-immune and host-microbiota crosstalk during homeostasis and disease processes in the gastrointestinal tract.
Collapse
|
40
|
Okui N, Kamata Y, Sagawa Y, Kuhara A, Hayashi K, Uwagawa T, Homma S, Yanaga K. Claudin 7 as a possible novel molecular target for the treatment of pancreatic cancer. Pancreatology 2019; 19:88-96. [PMID: 30416041 DOI: 10.1016/j.pan.2018.10.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 08/22/2018] [Accepted: 10/22/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND/OBJECTIVES Pancreatic cancer consists of various subpopulations of cells, some of which have aggressive proliferative properties. The molecules responsible for the aggressive proliferation of pancreatic cancer may become molecular targets for the therapies against pancreatic cancer. METHODS From a human pancreatic cancer cell line, MIA PaCa-2, MIA PaCa-2-A cells with an epithelial morphology and MIA PaCa-2-R cells with a non-epithelial morphology were clonogenically isolated by the limiting dilution method. Gene expression of these subpopulations was analyzed by DNA microarray. Gene knockdown was performed using siRNA. RESULTS Although the MIA PaCa-2-A and MIA PaCa-2-R cells displayed the same DNA short tandem repeat (STR) pattern identical to that of the parental MIA PaCa-2 cells, the MIA PaCa-2-A cells were more proliferative than the MIA PaCa-2-R cells both in culture and in tumor xenografts generated in immunodeficient mice. Furthermore, the MIA PaCa-2-A cells were more resistant to gemcitabine than the MIA PaCa-2-R cells. DNA microarray analysis revealed a high expression of claudin (CLDN) 7 in the MIA PaCa-2-A cells, as opposed to a low expression in the MIA PaCa-2-R cells. The knockdown of CLDN7 in the MIA PaCa-2-A cells induced a marked inhibition of proliferation. The MIA PaCa-2-A cells in which CLDN7 was knocked down exhibited a decreased expression of phosphorylated extracellular signal-regulated kinase (p-Erk)1/2 and G1 cell cycle arrest. CONCLUSIONS CLDN7 may be expressed in the rapidly proliferating and dominant cell population in human pancreatic cancer tissues and may be a novel molecular target for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Norimitsu Okui
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Yuko Kamata
- Division of Oncology, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Yukiko Sagawa
- Division of Oncology, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Akiko Kuhara
- Division of Oncology, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | - Kazumi Hayashi
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Tadashi Uwagawa
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Sadamu Homma
- Division of Oncology, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan.
| | - Katsuhiko Yanaga
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
41
|
Alam A, Neish A. Role of gut microbiota in intestinal wound healing and barrier function. Tissue Barriers 2018; 6:1539595. [PMID: 30404570 PMCID: PMC6389125 DOI: 10.1080/21688370.2018.1539595] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/30/2018] [Accepted: 10/11/2018] [Indexed: 12/19/2022] Open
Abstract
The mammalian intestine harbors a highly complex and abundant ensemble of bacteria that flourish in a nutrient-rich environment while profoundly influencing many aspects of host biology. The intestine coevolved with its resident microbes in a manner where the mucosa developed a barrier function to segregate the resident microbes from the rest of the body, and yet paradoxically, allowing integration of microbial signals for the host benefit. In this review, we provided a comprehensive overview of why the gut microbiota is key to the efficient development and maintenance of the intestinal barrier. We also highlighted how a destabilized equilibrium between gut microbiota and the host may eventuate in a wide range of intestinal diseases characterized by the disrupted intestinal barrier. Finally, the review delineated how microenvironmental changes in the injured mucosa result in an enrichment of a pro-regenerating consortium of bacteria, which augments mucosal wound repair and restoration of barrier functions.
Collapse
Affiliation(s)
- Ashfaqul Alam
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, USA
| | - Andrew Neish
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, USA
| |
Collapse
|
42
|
Curtis VF, Cartwright IM, Lee JS, Wang RX, Kao DJ, Lanis JM, Burney KM, Welch N, Hall CHT, Goldberg MS, Campbell EL, Colgan SP. Neutrophils as sources of dinucleotide polyphosphates and metabolism by epithelial ENPP1 to influence barrier function via adenosine signaling. Mol Biol Cell 2018; 29:2687-2699. [PMID: 30188771 PMCID: PMC6249842 DOI: 10.1091/mbc.e18-06-0377] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/20/2018] [Accepted: 08/27/2018] [Indexed: 12/15/2022] Open
Abstract
Extracellular adenosine signaling is established as a protective component in mucosal inflammatory responses. The sources of extracellular adenosine include enzymatic processing from nucleotides, such as ATP and AMP, that can be liberated from a variety of cell types, including infiltrating leukocytes. Here we demonstrate that activated human neutrophils are a source of diadenosine triphosphate (Ap3A), providing an additional source of nucleotides during inflammation. Profiling murine enteroids and intestinal epithelial cell lines revealed that intestinal epithelia prominently express apical and lateral ectonucleotide pyrophosphatase/phosphodiesterase-1 (ENPP1), a member of the ENPP family of enzymes that metabolize diadenosine phosphates, especially Ap3A. Extensions of these studies demonstrated that intestinal epithelia metabolize Ap3A to ADP and AMP, which are further metabolized to adenosine and made available to activate surface adenosine receptors. Using loss and gain of ENPP1 approaches, we revealed that ENPP1 coordinates epithelial barrier formation and promotes epithelial wound healing responses. These studies demonstrate the cooperative metabolism between Ap3A and ENPP1 function to provide a significant source of adenosine, subserving its role in inflammatory resolution.
Collapse
Affiliation(s)
- Valerie F. Curtis
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Ian M. Cartwright
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - J. Scott Lee
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Ruth X. Wang
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Daniel J. Kao
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Jordi M. Lanis
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Krista M. Burney
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Nichole Welch
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Caroline H. T. Hall
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Matthew S. Goldberg
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Eric L. Campbell
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Centre for Experimental Medicine, Queen’s University Belfast, Belfast BT7 1NN, Northern Ireland, UK
| | - Sean P. Colgan
- Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Rocky Mountain Veterans Affairs Hospital, Denver, CO 80220
| |
Collapse
|
43
|
Gross A, Pack LAP, Schacht GM, Kant S, Ungewiss H, Meir M, Schlegel N, Preisinger C, Boor P, Guldiken N, Krusche CA, Sellge G, Trautwein C, Waschke J, Heuser A, Leube RE, Strnad P. Desmoglein 2, but not desmocollin 2, protects intestinal epithelia from injury. Mucosal Immunol 2018; 11:1630-1639. [PMID: 30115995 DOI: 10.1038/s41385-018-0062-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 06/14/2018] [Accepted: 06/26/2018] [Indexed: 02/04/2023]
Abstract
Desmosomes are the least understood intercellular junctions in the intestinal epithelia and provide cell-cell adhesion via the cadherins desmoglein (Dsg)2 and desmocollin (Dsc)2. We studied these cadherins in Crohn's disease (CD) patients and in newly generated conditional villin-Cre DSG2 and DSC2 knockout mice (DSG2ΔIEC; DSC2ΔIEC). CD patients exhibited altered desmosomes and reduced Dsg2/Dsc2 levels. The intestines of both transgenic animal lines were histopathologically inconspicuous. However, DSG2ΔIEC, but not DSC2ΔIEC mice displayed an increased intestinal permeability, a wider desmosomal space as well as alterations in desmosomal and tight junction components. After dextran sodium sulfate (DSS) treatment and Citrobacter rodentium exposure, DSG2ΔIEC mice developed a more-pronounced colitis, an enhanced intestinal epithelial barrier disruption, leading to a stronger inflammation and activation of epithelial pSTAT3 signaling. No susceptibility to DSS-induced intestinal injury was noted in DSC2ΔIEC animals. Dsg2 interacted with the cytoprotective chaperone Hsp70. Accordingly, DSG2ΔIEC mice had lower Hsp70 levels in the plasma membrane compartment, whereas DSC2ΔIEC mice displayed a compensatory recruitment of galectin 3, a junction-tightening protein. Our results demonstrate that Dsg2, but not Dsc2 is required for the integrity of the intestinal epithelial barrier in vivo.
Collapse
Affiliation(s)
- Annika Gross
- Department of Medicine III and IZKF, University Hospital Aachen, Aachen, Germany
| | - Lotta A P Pack
- Department of Medicine III and IZKF, University Hospital Aachen, Aachen, Germany
| | - Gabriel M Schacht
- Department of Medicine III and IZKF, University Hospital Aachen, Aachen, Germany
| | - Sebastian Kant
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Aachen, Germany
| | - Hanna Ungewiss
- Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Michael Meir
- Department of Surgery I, University of Würzburg, Würzburg, Germany
| | - Nicolas Schlegel
- Department of Surgery I, University of Würzburg, Würzburg, Germany
| | | | - Peter Boor
- Institute of Pathology and Department of Nephrology, University Hospital Aachen, Aachen, Germany
| | - Nurdan Guldiken
- Department of Medicine III and IZKF, University Hospital Aachen, Aachen, Germany
| | - Claudia A Krusche
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Aachen, Germany
| | - Gernot Sellge
- Department of Medicine III and IZKF, University Hospital Aachen, Aachen, Germany
| | - Christian Trautwein
- Department of Medicine III and IZKF, University Hospital Aachen, Aachen, Germany
| | - Jens Waschke
- Department of Surgery I, University of Würzburg, Würzburg, Germany
| | - Arnd Heuser
- Max-Delbrück-Center of Molecular Medicine, Berlin, Germany
| | - Rudolf E Leube
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Aachen, Germany
| | - Pavel Strnad
- Department of Medicine III and IZKF, University Hospital Aachen, Aachen, Germany.
| |
Collapse
|
44
|
Tusk SE, Delalez NJ, Berry RM. Subunit Exchange in Protein Complexes. J Mol Biol 2018; 430:4557-4579. [DOI: 10.1016/j.jmb.2018.06.039] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/21/2018] [Accepted: 06/21/2018] [Indexed: 01/09/2023]
|
45
|
Workinger JL, Doyle RP, Bortz J. Challenges in the Diagnosis of Magnesium Status. Nutrients 2018; 10:E1202. [PMID: 30200431 PMCID: PMC6163803 DOI: 10.3390/nu10091202] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 08/24/2018] [Accepted: 08/27/2018] [Indexed: 12/31/2022] Open
Abstract
Magnesium is a critical mineral in the human body and is involved in ~80% of known metabolic functions. It is currently estimated that 60% of adults do not achieve the average dietary intake (ADI) and 45% of Americans are magnesium deficient, a condition associated with disease states like hypertension, diabetes, and neurological disorders, to name a few. Magnesium deficiency can be attributed to common dietary practices, medications, and farming techniques, along with estimates that the mineral content of vegetables has declined by as much as 80⁻90% in the last 100 years. However, despite this mineral's importance, it is poorly understood from several standpoints, not the least of which is its unique mechanism of absorption and sensitive compartmental handling in the body, making the determination of magnesium status difficult. The reliance on several popular sample assays has contributed to a great deal of confusion in the literature. This review will discuss causes of magnesium deficiency, absorption, handling, and compartmentalization in the body, highlighting the challenges this creates in determining magnesium status in both clinical and research settings.
Collapse
Affiliation(s)
- Jayme L Workinger
- Human Nutrition and Pharma, Balchem Corporation, 52 Sunrise Park Road, New Hampton, NY 10958, USA.
| | - Robert P Doyle
- Department of Chemistry, Center for Science and Technology, Syracuse University, 111 College Place, Syracuse, NY 13244, USA.
| | - Jonathan Bortz
- Human Nutrition and Pharma, Balchem Corporation, 52 Sunrise Park Road, New Hampton, NY 10958, USA.
| |
Collapse
|
46
|
Cyr DG, Gregory M. A special issue on the effects of toxicants on cellular junctions in development and reproduction. Reprod Toxicol 2018; 83:80-81. [PMID: 30172850 DOI: 10.1016/j.reprotox.2018.08.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Daniel G Cyr
- Laboratory for Reproductive Toxicology, INRS-Institut Armand-Frappier, 531 boulevard des Prairies, Laval, QC, H7V 1B7 Canada.
| | - Mary Gregory
- Laboratory for Reproductive Toxicology, INRS-Institut Armand-Frappier, 531 boulevard des Prairies, Laval, QC, H7V 1B7 Canada
| |
Collapse
|
47
|
Abstract
Approximately one-sixth of the worlds' population is infected with helminths and this class of parasite takes a major toll on domestic livestock. The majority of species of parasitic helminth that infect mammals live in the gut (the only niche for tapeworms) where they contact the hosts' epithelial cells. Here, the helminth-intestinal epithelial interface is reviewed in terms of the impact on, and regulation of epithelial barrier function, both intrinsic (epithelial permeability) and extrinsic (mucin, bacterial peptides, commensal bacteria) elements of the barrier. The data available on direct effects of helminths on epithelial permeability are scant, fragmentary and pales in comparison with knowledge of mobilization of immune reactions and effector cells in response to helminth parasites and how these impact intestinal barrier function. The interaction of helminth-host and helminth-host-bacteria is an important determinant of gut form and function and precisely defining these interactions will radically alter our understanding of normal gut physiology and pathophysiological reactions, revealing new approaches to infection with parasitic helminths, bacterial pathogens and idiopathic auto-inflammatory disease.
Collapse
Affiliation(s)
- Derek M McKay
- a Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology , Snyder Institute of Chronic Disease, Cumming School of Medicine, University of Calgary , Calgary , Alberta , Canada
| | - Adam Shute
- a Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology , Snyder Institute of Chronic Disease, Cumming School of Medicine, University of Calgary , Calgary , Alberta , Canada
| | - Fernando Lopes
- a Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology , Snyder Institute of Chronic Disease, Cumming School of Medicine, University of Calgary , Calgary , Alberta , Canada
| |
Collapse
|
48
|
Lee JS, Wang RX, Alexeev EE, Lanis JM, Battista KD, Glover LE, Colgan SP. Hypoxanthine is a checkpoint stress metabolite in colonic epithelial energy modulation and barrier function. J Biol Chem 2018; 293:6039-6051. [PMID: 29487135 PMCID: PMC5912467 DOI: 10.1074/jbc.ra117.000269] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 02/07/2018] [Indexed: 12/15/2022] Open
Abstract
Intestinal epithelial cells form a selectively permeable barrier to protect colon tissues from luminal microbiota and antigens and to mediate nutrient, fluid, and waste flux in the intestinal tract. Dysregulation of the epithelial cell barrier coincides with profound shifts in metabolic energy, especially in the colon, which exists in an energetically depleting state of physiological hypoxia. However, studies that systematically examine energy flux and adenylate metabolism during intestinal epithelial barrier development and restoration after disruption are lacking. Here, to delineate barrier-related energy flux, we developed an HPLC-based profiling method to track changes in energy flux and adenylate metabolites during barrier development and restoration. Cultured epithelia exhibited pooling of phosphocreatine and maintained ATP during barrier development. EDTA-induced epithelial barrier disruption revealed that hypoxanthine levels correlated with barrier resistance. Further studies uncovered that hypoxanthine supplementation improves barrier function and wound healing and that hypoxanthine appears to do so by increasing intracellular ATP, which improved cytoskeletal G- to F-actin polymerization. Hypoxanthine supplementation increased the adenylate energy charge in the murine colon, indicating potential to regulate adenylate energy charge-mediated metabolism in intestinal epithelial cells. Moreover, experiments in a murine colitis model disclosed that hypoxanthine loss during active inflammation correlates with markers of disease severity. In summary, our results indicate that hypoxanthine modulates energy metabolism in intestinal epithelial cells and is critical for intestinal barrier function.
Collapse
Affiliation(s)
- J Scott Lee
- From the Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado 80045 and
| | - Ruth X Wang
- From the Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado 80045 and
| | - Erica E Alexeev
- From the Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado 80045 and
| | - Jordi M Lanis
- From the Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado 80045 and
| | - Kayla D Battista
- From the Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado 80045 and
| | - Louise E Glover
- the School of Medicine, University College Dublin, Dublin, Ireland
| | - Sean P Colgan
- From the Department of Medicine and the Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado 80045 and
| |
Collapse
|
49
|
Amoozadeh Y, Anwer S, Dan Q, Venugopal S, Shi Y, Branchard E, Liedtke E, Ailenberg M, Rotstein OD, Kapus A, Szászi K. Cell confluence regulates claudin-2 expression: possible role for ZO-1 and Rac. Am J Physiol Cell Physiol 2018; 314:C366-C378. [DOI: 10.1152/ajpcell.00234.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Claudin-2 (Cldn-2) is a channel-forming tight junction (TJ) protein in the proximal tubules that mediates paracellular Na+ transport and has also emerged as a regulator of proliferation and migration. Expression of Cldn-2 is altered by numerous stimuli, but the underlying mechanisms remain incompletely understood. Here we show that Cldn-2 protein and mRNA expression were low in subconfluent tubular cells and increased during junction maturation. Cldn-1 or occludin did not exhibit similar confluence-dependence. Conversely, disruption of TJs by Ca2+ removal or silencing of zonula occludens-1 (ZO-1) or ZO-2 induced a large drop in Cldn-2 abundance. Immunofluorescent staining revealed a more uneven Cldn-2 staining in nascent, Cldn-1-positive TJs. Subconfluence and ZO-1 silencing augmented Cldn-2 degradation and reduced Cldn-2 promoter activity, suggesting that insertion into the TJs slows Cldn-2 turnover. Indeed, blocking endocytosis or lysosomal degradation increased Cldn-2 abundance. Cell confluence increased expression of the junctional adapters ZO-1 and -2, and the small GTPase Rac, and elevated Rac activity and p21-activated kinase (Pak) phosphorylation, suggesting that they might mediate confluence-dependent Cldn-2 regulation. Indeed, Rac silencing or Pak inhibition strongly reduced Cldn-2 protein abundance, which was likely the combined effect on turnover, as these interventions reduced Cldn-2 promoter activity and augmented Cldn-2 degradation. Taken together, our data suggest that TJ integrity and maturity, ZO-1 expression/TJ localization, and Rac/Pak control Cldn-2 degradation and synthesis. A feedback mechanism connecting Cldn-2 expression with junction remodeling, e.g., during wound healing, epithelial-mesenchymal transition, or tumor metastasis formation, may have important downstream effects on permeability, proliferation, and migration.
Collapse
Affiliation(s)
- Yasaman Amoozadeh
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital and Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Shaista Anwer
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital and Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Qinghong Dan
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital and Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Shruthi Venugopal
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital and Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Yixuan Shi
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital and Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Emily Branchard
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital and Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Elisabeth Liedtke
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital and Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Menachem Ailenberg
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital and Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Ori D. Rotstein
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital and Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - András Kapus
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital and Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Katalin Szászi
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital and Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
50
|
Pearce SC, Al-Jawadi A, Kishida K, Yu S, Hu M, Fritzky LF, Edelblum KL, Gao N, Ferraris RP. Marked differences in tight junction composition and macromolecular permeability among different intestinal cell types. BMC Biol 2018; 16:19. [PMID: 29391007 PMCID: PMC5793346 DOI: 10.1186/s12915-018-0481-z] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/03/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mammalian small intestinal tight junctions (TJ) link epithelial cells to one another and function as a permselective barrier, strictly modulating the passage of ions and macromolecules through the pore and leak pathways, respectively, thereby preventing the absorption of harmful compounds and microbes while allowing regulated transport of nutrients and electrolytes. Small intestinal epithelial permeability is ascribed primarily to the properties of TJs between adjoining enterocytes (ENTs), because there is almost no information on TJ composition and the paracellular permeability of nonenterocyte cell types that constitute a small but significant fraction of the intestinal epithelia. RESULTS Here we directed murine intestinal crypts to form specialized organoids highly enriched in intestinal stem cells (ISCs), absorptive ENTs, secretory goblet cells, or Paneth cells. The morphological and morphometric characteristics of these cells in organoids were similar to those in vivo. The expression of certain TJ proteins varied with cell type: occludin and tricellulin levels were high in both ISCs and Paneth cells, while claudin-1, -2, and -7 expression was greatest in Paneth cells, ISCs, and ENTs, respectively. In contrast, the distribution of claudin-15, zonula occludens 1 (ZO-1), and E-cadherin was relatively homogeneous. E-cadherin and claudin-7 marked mainly the basolateral membrane, while claudin-2, ZO-1, and occludin resided in the apical membrane. Remarkably, organoids enriched in ENTs or goblet cells were over threefold more permeable to 4 and 10 kDa dextran compared to those containing stem and Paneth cells. The TJ-regulator larazotide prevented the approximately tenfold increases in dextran flux induced by the TJ-disrupter AT1002 into organoids of different cell types, indicating that this ZO toxin nonselectively increases permeability. Forced dedifferentiation of mature ENTs results in the reacquisition of ISC-like characteristics in TJ composition and dextran permeability, suggesting that the post-differentiation properties of TJs are not hardwired. CONCLUSIONS Differentiation of adult intestinal stem cells into mature secretory and absorptive cell types causes marked, but potentially reversible, changes in TJ composition, resulting in enhanced macromolecular permeability of the TJ leak pathway between ENTs and between goblet cells. This work advances our understanding of how cell differentiation affects the paracellular pathway of epithelia.
Collapse
Affiliation(s)
- Sarah C Pearce
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
- Present address: Performance Nutrition Team, Combat Feeding Directorate, US Army, 15 General Greene Ave, Natick, MA, 01760-5018, USA
| | - Arwa Al-Jawadi
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Kunihiro Kishida
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
- Present address: Department of Science and Technology on Food Safety, Kindai University, Wakayama, 649-6493, Japan
| | - Shiyan Yu
- Department of Biological Sciences, Rutgers University, Life Science Center, 225 University Avenue, Newark, NJ, 07102, USA
| | - Madeleine Hu
- Department of Pathology & Laboratory Medicine, Center for Inflammation and Immunity, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Luke F Fritzky
- Advanced Microscopic Imaging Core Facility, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Karen L Edelblum
- Department of Pathology & Laboratory Medicine, Center for Inflammation and Immunity, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Life Science Center, 225 University Avenue, Newark, NJ, 07102, USA
| | - Ronaldo P Ferraris
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA.
| |
Collapse
|