1
|
Pouyiourou I, Fromm A, Piontek J, Rosenthal R, Furuse M, Günzel D. Ion permeability profiles of renal paracellular channel-forming claudins. Acta Physiol (Oxf) 2025; 241:e14264. [PMID: 39821681 PMCID: PMC11740656 DOI: 10.1111/apha.14264] [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: 07/31/2024] [Revised: 12/09/2024] [Accepted: 01/01/2025] [Indexed: 01/19/2025]
Abstract
AIM Members of the claudin protein family are the major constituents of tight junction strands and determine the permeability properties of the paracellular pathway. In the kidney, each nephron segment expresses a distinct subset of claudins that form either barriers against paracellular solute transport or charge- and size-selective paracellular channels. It was the aim of the present study to determine and compare the permeation properties of these renal paracellular ion channel-forming claudins. METHODS MDCK II cells, in which the five major claudins had been knocked out (claudin quintupleKO), were stably transfected with individual mouse Cldn2, -4, -8, -10a, -10b, or -15, or with dog Cldn16 or -19, or with a combination of mouse Cldn4 and Cldn8, or dog Cldn16 and Cldn19. Permeation properties were investigated in the Ussing chamber and claudin interactions by FRET assays. RESULTS Claudin-4 and -19 formed barriers against solute permeation. However, at low pH values and in the absence of HCO3 -, claudin-4 conveyed a weak chloride and nitrate permeability. Claudin-8 needed claudin-4 for assembly into TJ strands and abolished this anion preference. Claudin-2, -10a, -10b, -15, -16+19 formed highly permeable channels with distinctive permeation profiles for different monovalent and divalent anions or cations, but barriers against the permeation of ions of opposite charge and of the paracellular tracer fluorescein. CONCLUSION Paracellular ion permeabilities along the nephron are strictly determined by claudin expression patterns. Paracellular channel-forming claudins are specific for certain ions and thus lower transepithelial resistance, yet form barriers against the transport of other solutes.
Collapse
Affiliation(s)
- Ioanna Pouyiourou
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, RheumatologyCharité–Universitätsmedizin BerlinBerlinGermany
| | - Anja Fromm
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, RheumatologyCharité–Universitätsmedizin BerlinBerlinGermany
| | - Jörg Piontek
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, RheumatologyCharité–Universitätsmedizin BerlinBerlinGermany
| | - Rita Rosenthal
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, RheumatologyCharité–Universitätsmedizin BerlinBerlinGermany
| | - Mikio Furuse
- Division of Cell StructureNational Institute for Physiological SciencesOkazakiJapan
- Physiological Sciences ProgramGraduate Institute for Advanced Studies, SOKENDAIOkazakiJapan
- Nagoya University Graduate School of MedicineNagoyaJapan
| | - Dorothee Günzel
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, RheumatologyCharité–Universitätsmedizin BerlinBerlinGermany
| |
Collapse
|
2
|
Kesaraju S, Li Y, Xing J, Tracy M, Wannemo K, Holder A, Zhao P, Khan MA, Kainov J, Rana N, Sidahmed M, Hyoju S, Smith L, Matthews J, Tay S, Khalili-Araghi F, Rana M, Oakes SA, Shen L, Weber CR. Inflammation-Induced Claudin-2 Upregulation Limits Pancreatitis Progression by Enhancing Tight Junction-Controlled Pancreatic Ductal Transport. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.01.555960. [PMID: 39605652 PMCID: PMC11601259 DOI: 10.1101/2023.09.01.555960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Pancreatitis is an inflammatory disease of the pancreas that can arise due to various factors, including environmental risks such as diet, alcohol, and smoking, as well as genetic predispositions. In some cases, pancreatitis may progress and become chronic, leading to irreversible damage and impaired pancreatic function. Genome-wide association studies (GWAS) have identified polymorphisms at the X-linked CLDN2 locus as risk factors for both sporadic and alcohol-related chronic pancreatitis. CLDN2 encodes claudin-2 (CLDN2), a paracellular cation-selective channel localized at tight junctions and expressed in the pancreas and other secretory organs. However, whether and how CLDN2 may modify pancreatitis susceptibility remains poorly understood. We aimed to clarify the potential role of CLDN2 in the onset and progression of pancreatitis. We employed multiple methodologies to examine the role of CLDN2 in human pancreatic tissue, caerulein-induced experimental pancreatitis mouse model, and pancreatic ductal epithelial organoids. In both human chronic pancreatitis tissues and caerulein-induced experimental pancreatitis, CLDN2 protein was significantly upregulated in pancreatic ductal epithelial cells. Our studies using pancreatic ductal epithelial organoids and mice demonstrated the inflammatory cytokine IFNγ upregulates claudin-2 expression at both RNA and protein levels. Following caerulein treatment, Ifng KO mice had diminished upregulation of CLDN2 relative to WT mice, indicating that caerulein-induced claudin-2 expression is partially driven by IFNγ. Functionally, Cldn2 knockout mice developed more severe caerulein-induced experimental pancreatitis, indicating CLDN2 plays a protective role in pancreatitis development. Pancreatic ductal epithelial organoid-based studies demonstrated that CLDN2 is critical for sodium-dependent water transport and necessary for cAMP-driven, CFTR-dependent fluid secretion. These findings suggest that functional crosstalk between CLDN2 and CFTR is essential for fluid transport in pancreatic ductal epithelium, which may protect against pancreatitis by adjusting pancreatic ductal secretion to prevent worsening autodigestion and inflammation. In conclusion, our studies suggest CLDN2 upregulation during pancreatitis may play a protective role in limiting disease development, and decreased CLDN2 function may increase pancreatitis severity. These results point to the possibility of modulating pancreatic ductal CLDN2 function as an approach for therapeutic intervention of pancreatitis.
Collapse
|
3
|
Kim SU, Choi JA, Han MH, Choi JY, Park JH, Kim MS, Kwon YH. Tight junction protein changes in irritable bowel syndrome: the relation of age and disease severity. Korean J Intern Med 2024; 39:906-916. [PMID: 39551069 PMCID: PMC11569931 DOI: 10.3904/kjim.2024.097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/02/2024] [Accepted: 06/16/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND/AIMS The etiology of irritable bowel syndrome (IBS) is associated with intestinal mucosal barrier damage. However, changes in the tight junction (TJ) proteins in IBS have not been fully elucidated. This study aimed to evaluate TJ protein changes in IBS patients and the relationship between aging and disease severity. METHODS Thirty-six patients with IBS fulfilling the Rome IV criteria and twenty-four controls were included. To evaluate the change of TJ in the colonic mucosa, quantitative reverse transcription polymerase chain reaction, western blot, and immunohistochemistry (IHC) were performed, respectively. RESULTS The entire IBS group (n = 36) exhibited decreased levels of claudin-1 and -2 mRNA compared to the control group (n = 24), with statistical significance (p < 0.05). Additionally, in western blot analyses, both claudin-1 and ZO-1 levels were significantly reduced in the IBS group compared to the control group (n = 24) (p < 0.05). IHC analysis further revealed that ZO-1 expression was significantly lower in the IBS group than in the control group (p < 0.001). This trend of reduced ZO-1 expression was also observed in the moderate-to-severe IBS subgroup (p < 0.001). Significantly, ZO-1 expression was notably lower in both the young- (p = 0.036) and old-aged (p = 0.039) IBS groups compared to their respective age-matched control groups. Subtype analysis indicated a more pronounced decrease in ZO-1 expression with advancing age. CONCLUSION ZO-1 expression was especially decreased in the aged IBS group. These results suggest that ZO-1 might be the prominent TJ protein causing IBS in the aging population.
Collapse
Affiliation(s)
- Sang Un Kim
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Korea
| | - Ji A Choi
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Korea
| | - Man-Hoon Han
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Jin Young Choi
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Korea
| | - Ji Hye Park
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Korea
| | - Moon Sik Kim
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Yong Hwan Kwon
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Korea
| |
Collapse
|
4
|
Hirase R, Fujita T, Miyai T, Kawasaki H, Koseki H. Pharmacological Impacts of Mucopolysacccharide Polyphosphates in the Epidermis Involves Inhibition of Amphiregulin-Mediated Signals in Keratinocytes. Exp Dermatol 2024; 33:e70000. [PMID: 39422315 DOI: 10.1111/exd.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/26/2024] [Accepted: 09/29/2024] [Indexed: 10/19/2024]
Abstract
The epidermis, the most superficial layer of the human skin, serves a critical barrier function, protecting the body from external pathogens and allergens. Dysregulation of epidermal differentiation contributes to barrier dysfunction and has been implicated in the pathology of various dermatological diseases, including atopic dermatitis (AD). Mucopolysaccharide polysulphate (MPS) is a moisturising agent used to treat xerosis in patients with AD. However, its mechanism of action on keratinocytes, the main constituents of the epidermis, remains unclear. In this study, we investigated the effect of MPS on keratinocytes by subjecting adult human epidermal and three-dimensional cultured keratinocytes to MPS treatment, followed by transcriptome analysis. The analysis revealed that MPS treatment enhances keratinocyte differentiation and suppresses proliferation. We focused on amphiregulin (AREG), a membrane protein that belongs to the epidermal growth factor (EGF) family and possesses a heparin-binding domain, as a significant target among the genes altered by MPS. MPS exerted an inhibitory effect directly on AREG, rather than on EGF receptors or other members of the EGF family. Furthermore, AREG leads to a reduction in epidermal barrier function, whereas MPS contributes to barrier enhancement via AREG inhibition. Collectively, these findings suggest that MPS modulates barrier function through AREG inhibition, offering insights into potential therapeutic strategies for skin barrier restoration.
Collapse
Affiliation(s)
- Ryo Hirase
- Graduate School of Medicine, Chiba University, Chiba, Japan
- Kyoto R&D Center, Maruho Co., Ltd., Kyoto, Japan
| | | | - Tomohiro Miyai
- RIKEN Center for Integrative Medical Sciences (IMS-RCAI), Kanagawa, Japan
| | - Hiroshi Kawasaki
- RIKEN Center for Integrative Medical Sciences (IMS-RCAI), Kanagawa, Japan
| | - Haruhiko Koseki
- Graduate School of Medicine, Chiba University, Chiba, Japan
- RIKEN Center for Integrative Medical Sciences (IMS-RCAI), Kanagawa, Japan
| |
Collapse
|
5
|
Kimura R, Hashimoto S, Eguchi H, Morikawa Y, Suenami K, Yoshino Y, Matsunaga T, Endo S, Ikari A. Enhancement of chemoresistance by claudin-1-mediated formation of amino acid barriers in human lung adenocarcinoma A549 cells. Arch Biochem Biophys 2024; 759:110106. [PMID: 39067558 DOI: 10.1016/j.abb.2024.110106] [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: 04/15/2024] [Revised: 06/19/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Claudin-1 (CLDN1) is highly expressed in human lung adenocarcinoma-derived A549 cells and is involved in the augmentation of chemoresistance. However, the mechanism of chemoresistance is not fully understood. In the tumor microenvironment, cancer cells are exposed to stress conditions such as hypoxia and malnutrition. Here, we investigated the effect of CLDN1 expression on amino acid (AA) flux and chemoresistance using A549 cells. The expression of L-type AA transporters, LAT1 and LAT3, was decreased by CLDN1 silencing in A549 spheroids. A reduction in extracellular AA concentration increased the expression of these AA transporters in two-dimensional (2D) cultured cells. The paracellular AA flux except for Ser, Thr, Tyr, Ala, and Gly was enhanced by CLDN1 silencing. These results suggest that CLDN1 forms a paracellular barrier to some AAs, leading to the elevation of LAT1/3 expression in spheroids. The production of reactive oxygen species in the mitochondria and cytosol was decreased by CLDN1 silencing in spheroids, resulting in downregulation of the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and its target antioxidant genes. CLDN1 silencing enhanced the cytotoxicity of anticancer drugs including doxorubicin and cisplatin, which was blocked by sulforaphane, an inducer of Nrf2 signaling. Similarly, the anticancer-induced toxicity was enhanced by Nrf2 silencing. In 2D cultured cells, the anticancer-induced toxicity was attenuated by AA deficiency and sulforaphane. We suggest that CLDN1 forms an AA barrier in spheroids, leading to the augmentation of Nrf2-dependent chemoresistance in A549 cells.
Collapse
Affiliation(s)
- Riho Kimura
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Shotaro Hashimoto
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Hiroaki Eguchi
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Yoshifumi Morikawa
- Forensic Science Laboratory, Gifu Prefectural Police Headquarters, Gifu, 500-8501, Japan
| | - Koichi Suenami
- Forensic Science Laboratory, Gifu Prefectural Police Headquarters, Gifu, 500-8501, Japan
| | - Yuta Yoshino
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Toshiyuki Matsunaga
- Laboratory of Bioinformatics, Gifu Pharmaceutical University, Gifu, 502-8585, Japan
| | - Satoshi Endo
- Drug Design Laboratory, Gifu University, Gifu, 501-1194, Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan.
| |
Collapse
|
6
|
Marsch P, Rajagopal N, Nangia S. Biophysics of claudin proteins in tight junction architecture: Three decades of progress. Biophys J 2024; 123:2363-2378. [PMID: 38859584 PMCID: PMC11365114 DOI: 10.1016/j.bpj.2024.06.010] [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: 01/17/2024] [Revised: 05/19/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024] Open
Abstract
Tight junctions are cell-cell adhesion complexes that act as gatekeepers of the paracellular space. Formed by several transmembrane proteins, the claudin family performs the primary gate-keeping function. The claudin proteins form charge and size-selective diffusion barriers to maintain homeostasis across endothelial and epithelial tissue. Of the 27 known claudins in mammals, some are known to seal the paracellular space, while others provide selective permeability. The differences in permeability arise due to the varying expression levels of claudins in each tissue. The tight junctions are observed as strands in freeze-fracture electron monographs; however, at the molecular level, tight junction strands form when multiple claudin proteins assemble laterally (cis assembly) within a cell and head-on (trans assembly) with claudins of the adjacent cell in a zipper-like architecture, closing the gap between the neighboring cells. The disruption of tight junctions caused by changing claudin expression levels or mutations can lead to diseases. Therefore, knowledge of the molecular architecture of the tight junctions and how that is tied to tissue-specific function is critical for fighting diseases. Here, we review the current understanding of the tight junctions accrued over the last three decades from experimental and computational biophysics perspectives.
Collapse
Affiliation(s)
- Patrick Marsch
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York
| | - Nandhini Rajagopal
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York
| | - Shikha Nangia
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York.
| |
Collapse
|
7
|
Dithmer S, Blasig IE, Fraser PA, Qin Z, Haseloff RF. The Basic Requirement of Tight Junction Proteins in Blood-Brain Barrier Function and Their Role in Pathologies. Int J Mol Sci 2024; 25:5601. [PMID: 38891789 PMCID: PMC11172262 DOI: 10.3390/ijms25115601] [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: 02/07/2024] [Revised: 03/10/2024] [Accepted: 03/28/2024] [Indexed: 06/21/2024] Open
Abstract
This review addresses the role of tight junction proteins at the blood-brain barrier (BBB). Their expression is described, and their role in physiological and pathological processes at the BBB is discussed. Based on this, new approaches are depicted for paracellular drug delivery and diagnostics in the treatment of cerebral diseases. Recent data provide convincing evidence that, in addition to its impairment in the course of diseases, the BBB could be involved in the aetiology of CNS disorders. Further progress will be expected based on new insights in tight junction protein structure and in their involvement in signalling pathways.
Collapse
Affiliation(s)
- Sophie Dithmer
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125 Berlin, Germany (I.E.B.)
| | - Ingolf E. Blasig
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125 Berlin, Germany (I.E.B.)
| | | | - Zhihai Qin
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100049, China
| | - Reiner F. Haseloff
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125 Berlin, Germany (I.E.B.)
| |
Collapse
|
8
|
Akinsuyi OS, Xhumari J, Ojeda A, Roesch LFW. Gut permeability among Astronauts during Space missions. LIFE SCIENCES IN SPACE RESEARCH 2024; 41:171-180. [PMID: 38670644 DOI: 10.1016/j.lssr.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 02/02/2024] [Accepted: 03/13/2024] [Indexed: 04/28/2024]
Abstract
The space environment poses substantial challenges to human physiology, including potential disruptions in gastrointestinal health. Gut permeability has only recently become widely acknowledged for its potential to cause adverse effects on a systemic level, rendering it a critical factor to investigate in the context of spaceflight. Here, we propose that astronauts experience the onset of leaky gut during space missions supported by transcriptomic and metagenomic analysis of human and murine samples. A genetic map contributing to intestinal permeability was constructed from a systematic review of current literature. This was referenced against our re-analysis of three independent transcriptomic datasets which revealed significant changes in gene expression patterns associated with the gut barrier. Specifically, in astronauts during flight, we observed a substantial reduction in the expression genes that are crucial for intestinal barrier function, goblet cell development, gut microbiota modulation, and immune responses. Among rodent spaceflight studies, differential expression of cytokines, chemokines, and genes which regulate mucin production and post-translational modifications suggest a similar dysfunction of intestinal permeability. Metagenomic analysis of feces from two murine studies revealed a notable reduction probiotic, short chain fatty acid-producing bacteria and an increase in the Gram-negative pathogens, including Citrobacter rodentium, Enterobacter cloacea, Klebsiella aerogenes, and Proteus hauseri which promote LPS circulation, a recipe for barrier disruption and systemic inflammatory activation. These findings emphasize the critical need to understand the underlying mechanisms and develop interventions to maintain gastrointestinal health in space.
Collapse
Affiliation(s)
- Oluwamayowa S Akinsuyi
- Institute of Food and Agricultural Sciences, Department of Microbiology and Cell Science, University of Florida, Gainesville 32611, FL, USA
| | - Jessica Xhumari
- Institute of Food and Agricultural Sciences, Department of Microbiology and Cell Science, University of Florida, Gainesville 32611, FL, USA
| | - Amanda Ojeda
- Institute of Food and Agricultural Sciences, Department of Microbiology and Cell Science, University of Florida, Gainesville 32611, FL, USA
| | - Luiz F W Roesch
- Institute of Food and Agricultural Sciences, Department of Microbiology and Cell Science, University of Florida, Gainesville 32611, FL, USA.
| |
Collapse
|
9
|
Huang T, Zhang ZY, Qiu ZL, Li L, Liu XX, Wang L, Wang ZY, Li ZP, Xiao GS, Wang W. Effect of Cymbopogon martini (Roxb.) Will.Watson essential oil on antioxidant activity, immune and intestinal barrier-related function, and gut microbiota in pigeons infected by Candida albicans. Front Pharmacol 2024; 15:1380277. [PMID: 38628645 PMCID: PMC11018936 DOI: 10.3389/fphar.2024.1380277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
Essential oils are potential alternatives to antibiotics for preventing Candida albicans (C. albicans) infection which is responsible for economic losses in the pigeon industry. Cymbopogon martini essential oil (EO) can inhibit pathogens, particularly fungal pathogens but its potential beneficial effects on C. albicans-infected pigeons remain unclear. Therefore, we investigated the impact of C. martini EO on antioxidant activity, immune response, intestinal barrier function, and intestinal microbiota in C. albicans-infected pigeons. The pigeons were divided into four groups as follows: (1) NC group: C. albicans uninfected/C. martini EO untreated group; (2) PC group: C. albicans infected/C. martini EO untreated group; (3) LPA group: C. albicans infected/1% C. martini EO treated group; and (4) HPA group: C. albicans infected/2% C. martini EO treated group. The pigeons were infected with C. albicans from day of age 35 to 41 and treated with C. martini EO from day of age 42 to 44, with samples collected on day of age 45 for analysis. The results demonstrated that C. martini EO prevented the reduction in the antioxidant enzymes SOD and GSH-Px causes by C. albicans challenge in pigeons. Furthermore, C. martini EO could decrease the relative expression of IL-1β, TGF-β, and IL-8 in the ileum, as well as IL-1β and IL-8 in the crop, while increasing the relative expression of Claudin-1 in the ileum and the crop and Occludin in the ileum in infected pigeons. Although the gut microbiota composition was not significantly affected by C. martini EO, 2% C. martini EO increased the abundance of Alistipes and Pedobacter. In conclusion, the application of 2% C. martini EO not only enhanced the level of antioxidant activity and the expression of genes related to intestinal barrier function but also inhibited inflammatory genes in C. albicans-infected pigeons and increased the abundance of gut bacteria that are resistant to C. albicans.
Collapse
Affiliation(s)
- Ting Huang
- Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Zheng-Yue Zhang
- Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Zhi-Lin Qiu
- Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Lin Li
- Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Xian-Xi Liu
- Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Lei Wang
- Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Zi-Ying Wang
- Meizhou Jinlv Modern Agriculture Development Co., Ltd., Meizhou, China
| | - Zhi-Peng Li
- Guangdong Baoning Agriculture and Animal Husbandry Technology Co., Ltd., Meizhou, China
| | - Geng-Sheng Xiao
- Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Wei Wang
- Zhongkai University of Agriculture and Engineering, Guangzhou, China
| |
Collapse
|
10
|
Cornelius V, Droessler L, Amasheh S. Quercetin Improves Barrier Properties in Porcine Small Intestine but Not in Peyer's Patches. Int J Mol Sci 2024; 25:1530. [PMID: 38338808 PMCID: PMC10855467 DOI: 10.3390/ijms25031530] [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: 12/21/2023] [Revised: 01/18/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Peyer's patches (PPs) are part of the gut-associated lymphatic tissue (GALT) and represent the first line of the intestinal immunological defense. They consist of follicles with lymphocytes and an overlying subepithelial dome with dendritic cells and macrophages, and they are covered by the follicle-associated epithelium (FAE). A sealed paracellular pathway in the FAE is crucial for the controlled uptake of luminal antigens. Quercetin is the most abundant plant flavonoid and has a barrier-strengthening effect on tight junctions (TJs), a protein complex that regulates the paracellular pathway. In this study, we aimed to analyze the effect of quercetin on porcine PPs and the surrounding villus epithelium (VE). We incubated both tissue types for 4 h in Ussing chambers, recorded the transepithelial electrical resistance (TEER), and measured the unidirectional tracer flux of [3H]-mannitol. Subsequently, we analyzed the expression, protein amount, and localization of three TJ proteins, claudin 1, claudin 2, and claudin 4. In the PPs, we could not detect an effect of quercetin after 4 h, neither on TEER nor on the [3H]-mannitol flux. In the VE, quercetin led to a higher TEER value, while the [3H]-mannitol flux was unchanged. The pore-forming claudin 2 was decreased while the barrier-forming claudin 4 was increased and the expression was upregulated. Claudin 1 was unchanged and all claudins could be located in the paracellular membrane by immunofluorescence microscopy. Our study shows the barrier-strengthening effect of quercetin in porcine VE by claudin 4 upregulation and a claudin 2 decrease. Moreover, it underlines the different barrier properties of PPs compared to the VE.
Collapse
Affiliation(s)
| | | | - Salah Amasheh
- Institute of Veterinary Physiology, School of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany
| |
Collapse
|
11
|
Zou B, Long F, Xue F, Chen C, Zhang X, Qu M, Xu L. Protective Effects of Niacin on Rumen Epithelial Cell Barrier Integrity in Heat-Stressed Beef Cattle. Animals (Basel) 2024; 14:313. [PMID: 38275773 PMCID: PMC10812637 DOI: 10.3390/ani14020313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/28/2023] [Accepted: 01/05/2024] [Indexed: 01/27/2024] Open
Abstract
The present study investigates the theoretical basis for maintaining normal physiological functions in heat-stressed beef cattle by exploring the effects of niacin supplementation on the permeability of the rumen epithelial cell barrier. Herein, 12 Jinjiang bulls with an average weight of approximately 400 ± 20.0 kg were randomly divided into three groups, thermoneutral (TN), heat-stressed (HS), and heat-stressed niacin-supplemented (HN) groups, with 4 bulls in each group. The experiment spanned 70 days, and the plasma concentrations of D-lactic acid, diamine oxidase (DAO), lipopolysaccharides (LPSs), and inflammatory cytokines were analyzed. Additionally, we assessed the gene expression of tight junction proteins to understand the effect of niacin supplementation on heat-stressed beef cattle. Our results revealed that heat stress significantly increased the D-lactic acid and LPS levels in beef cattle plasma on days 30 and 45 of the experiment (p < 0.05). Moreover, it led to a significant rise in DAO levels on day 30 (p < 0.05). Niacin supplementation significantly reduced the LPS levels on day 30 (p < 0.05). Heat stress significantly elevated the plasma concentrations of inflammatory cytokines interleukin-1β (IL-1β), IL-2, IL-6, and tumor necrosis factor-α (TNF-α) (p < 0.05), while reducing the IL-4 concentration (p < 0.05). However, niacin supplementation effectively mitigated the concentrations of these inflammatory factors by reducing IL-1β, IL-2, IL-6, and TNF-α concentrations and increasing IL-4 concentrations. The mRNA expressions of tight junction proteins zonula occluden-1 (ZO-1), claudin-1, claudin-4, and claudin-7 were significantly downregulated (p < 0.05) in the HS group compared to those in the TN group, and those of ZO-1 and occludin were significantly upregulated (p < 0.05) in the HN group compared to those in the HS group. Notably, no significant differences were observed in ruminal papillae length and width among the studied groups (p > 0.05). Our findings indicate that heat stress adversely impacted the tight junction structure of the rumen epithelium, leading to a significant reduction in the expression of tight junction protein mRNA. Consequently, heat stress impaired the rumen mucosal barrier function, resulting in increased intestinal permeability. The mechanism underlying this effect may be associated with the decreased expression of tight junction protein genes in the rumen epithelial cells. However, niacin supplementation mitigated the detrimental effects of heat stress on intestinal permeability in beef cattle and increased the expression of tight junction protein genes in the rumen epithelium, thereby effectively protecting the rumen barrier in heat-stressed beef cattle. These results highlight the potential of nicotinic acid as a protective agent against the negative impacts of heat stress on intestinal integrity in beef cattle.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lanjiao Xu
- Jiangxi Province Key Laboratory of Animal Nutrition, Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang 330045, China; (B.Z.); (F.L.); (F.X.); (C.C.); (X.Z.); (M.Q.)
| |
Collapse
|
12
|
Raya-Sandino A, Lozada-Soto KM, Rajagopal N, Garcia-Hernandez V, Luissint AC, Brazil JC, Cui G, Koval M, Parkos CA, Nangia S, Nusrat A. Claudin-23 reshapes epithelial tight junction architecture to regulate barrier function. Nat Commun 2023; 14:6214. [PMID: 37798277 PMCID: PMC10556055 DOI: 10.1038/s41467-023-41999-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 09/26/2023] [Indexed: 10/07/2023] Open
Abstract
Claudin family tight junction proteins form charge- and size-selective paracellular channels that regulate epithelial barrier function. In the gastrointestinal tract, barrier heterogeneity is attributed to differential claudin expression. Here, we show that claudin-23 (CLDN23) is enriched in luminal intestinal epithelial cells where it strengthens the epithelial barrier. Complementary approaches reveal that CLDN23 regulates paracellular ion and macromolecule permeability by associating with CLDN3 and CLDN4 and regulating their distribution in tight junctions. Computational modeling suggests that CLDN23 forms heteromeric and heterotypic complexes with CLDN3 and CLDN4 that have unique pore architecture and overall net charge. These computational simulation analyses further suggest that pore properties are interaction-dependent, since differently organized complexes with the same claudin stoichiometry form pores with unique architecture. Our findings provide insight into tight junction organization and propose a model whereby different claudins combine to form multiple distinct complexes that modify epithelial barrier function by altering tight junction structure.
Collapse
Affiliation(s)
- Arturo Raya-Sandino
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Nandhini Rajagopal
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, USA
| | | | - Anny-Claude Luissint
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jennifer C Brazil
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Guiying Cui
- Department of Pediatrics, Emory + Children's Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael Koval
- Departments of Medicine and Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Charles A Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Shikha Nangia
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, USA.
| | - Asma Nusrat
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
13
|
Abstract
Sequential expression of claudins, a family of tight junction proteins, along the nephron mirrors the sequential expression of ion channels and transporters. Only by the interplay of transcellular and paracellular transport can the kidney efficiently maintain electrolyte and water homeostasis in an organism. Although channel and transporter defects have long been known to perturb homeostasis, the contribution of individual tight junction proteins has been less clear. Over the past two decades, the regulation and dysregulation of claudins have been intensively studied in the gastrointestinal tract. Claudin expression patterns have, for instance, been found to be affected in infection and inflammation, or in cancer. In the kidney, a deeper understanding of the causes as well as the effects of claudin expression alterations is only just emerging. Little is known about hormonal control of the paracellular pathway along the nephron, effects of cytokines on renal claudin expression or relevance of changes in paracellular permeability to the outcome in any of the major kidney diseases. By summarizing current findings on the role of specific claudins in maintaining electrolyte and water homeostasis, this Review aims to stimulate investigations on claudins as prognostic markers or as druggable targets in kidney disease.
Collapse
Affiliation(s)
- Luca Meoli
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dorothee Günzel
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
14
|
Houillier P, Lievre L, Hureaux M, Prot-Bertoye C. Mechanisms of paracellular transport of magnesium in intestinal and renal epithelia. Ann N Y Acad Sci 2023; 1521:14-31. [PMID: 36622354 DOI: 10.1111/nyas.14953] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Magnesium is the fourth most abundant cation in the body. It plays a critical role in many biological processes, including the process of energy release. Paracellular transport of magnesium is mandatory for magnesium homeostasis. In addition to intestinal absorption that occurs in part across the paracellular pathway, magnesium is reabsorbed by the kidney tubule. The bulk of magnesium is reabsorbed through the paracellular pathway in the proximal tubule and the thick ascending limb of the loop of Henle. The finding that rare genetic diseases due to pathogenic variants in genes encoding specific claudins (CLDNs), proteins located at the tight junction that determine the selectivity and the permeability of the paracellular pathway, led to an awareness of their importance in magnesium homeostasis. Familial hypomagnesemia with hypercalciuria and nephrocalcinosis is caused by a loss of function of CLDN16 or CLDN19. Pathogenic CLDN10 variants cause HELIX syndrome, which is associated with a severe renal loss of sodium chloride and hypermagnesemia. The present review summarizes the current knowledge of the mechanisms and factors involved in paracellular magnesium permeability. The review also highlights some of the unresolved questions that need to be addressed.
Collapse
Affiliation(s)
- Pascal Houillier
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Paris, France
- CNRS ERL 8228 - Laboratoire de Physiologie Rénale et Tubulopathies, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Physiologie, Paris, France
- Centre de Référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA), Paris, France
- Centre de Référence des Maladies Rares du Calcium et du Phosphate, Paris, France
| | - Loïc Lievre
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Paris, France
- CNRS ERL 8228 - Laboratoire de Physiologie Rénale et Tubulopathies, Paris, France
| | - Marguerite Hureaux
- Centre de Référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA), Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France
- Paris Centre de Recherche Cardio-vasculaire, INSERM, Université Paris Cité, Paris, France
| | - Caroline Prot-Bertoye
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Paris, France
- CNRS ERL 8228 - Laboratoire de Physiologie Rénale et Tubulopathies, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Physiologie, Paris, France
- Centre de Référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA), Paris, France
- Centre de Référence des Maladies Rares du Calcium et du Phosphate, Paris, France
| |
Collapse
|
15
|
Wei W, Li W, Yang L, Weeramantry S, Ma L, Fu P, Zhao Y. Tight junctions and acute kidney injury. J Cell Physiol 2023; 238:727-741. [PMID: 36815285 DOI: 10.1002/jcp.30976] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 01/27/2023] [Accepted: 02/04/2023] [Indexed: 02/24/2023]
Abstract
Acute kidney injury (AKI) is characterized by a rapid reduction in kidney function caused by various etiologies. Tubular epithelial cell dysregulation plays a pivotal role in the pathogenesis of AKI. Tight junction (TJ) is the major molecular structure that connects adjacent epithelial cells and is critical in maintaining barrier function and determining the permeability of epithelia. TJ proteins are dysregulated in various types of AKI, and some reno-protective drugs can reverse TJ changes caused by insult. An in-depth understanding of TJ regulation and its causality with AKI will provide more insight to the disease pathogenesis and will shed light on the potential role of TJs to serve as novel therapeutic targets in AKI.
Collapse
Affiliation(s)
- Wei Wei
- Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weiying Li
- Department of Internal Medicine, Florida Hospital/AdventHealth, Orlando, Florida, USA
| | - Letian Yang
- Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Savidya Weeramantry
- Department of Internal Medicine, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Liang Ma
- Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ping Fu
- Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuliang Zhao
- Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
16
|
Acute Stress Regulates Sex-Related Molecular Responses in the Human Jejunal Mucosa: Implications for Irritable Bowel Syndrome. Cells 2023; 12:cells12030423. [PMID: 36766765 PMCID: PMC9913488 DOI: 10.3390/cells12030423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/17/2023] [Accepted: 01/23/2023] [Indexed: 02/03/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a prevalent gastrointestinal disorder linked to intestinal barrier dysfunction and life stress. We have previously reported that female sex per se determines an increased susceptibility to intestinal barrier dysfunction after cold pain stress (CPS). We aimed to identify sex-related molecular differences in response to CPS in healthy subjects to understand the origin of sex bias predominance in IBS. In 13 healthy males and 21 females, two consecutive jejunal biopsies were obtained using Watson's capsule, at baseline, and ninety minutes after CPS. Total mucosal RNA and protein were isolated from jejunal biopsies. Expression of genes related to epithelial barrier (CLDN1, CLDN2, OCLN, ZO-1, and ZO-3), mast cell (MC) activation (TPSAB1, SERPINA1), and the glucocorticoid receptor (NR3C1) were analyzed using RT-qPCR. NR3C1, ZO-1 and OCLN protein expression were evaluated through immunohistochemistry and western blot, and mucosal inflammation through MC, lymphocyte, and eosinophil numbering. Autonomic, hormonal, and psychological responses to CPS were monitored. We found an increase in jejunal MCs, a reduced CLDN1 and OCLN expression, and an increased CLDN2 and SERPINA1 expression 90 min after CPS. We also found a significant decrease in ZO-1, OCLN, and NR3C1 gene expression, and a decrease in OCLN protein expression only in females, when compared to males. CPS induced a significant increase in blood pressure, plasma cortisol and ACTH, and subjective stress perception in all participants. Specific and independent sex-related molecular responses in epithelial barrier regulation are unraveled by acute stress in the jejunum of healthy subjects and may partially explain female predominance in IBS.
Collapse
|
17
|
Günzel D. Is there a molecular basis for solvent drag in the renal proximal tubule? Pflugers Arch 2023; 475:277-281. [PMID: 36418493 PMCID: PMC9849172 DOI: 10.1007/s00424-022-02773-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022]
Abstract
The concept of solvent drag, i.e., water and solutes sharing the same pore and their transport being frictionally coupled, was first proposed in the early 1950s. During the following decades, it was applied to transport processes across cell membranes as well as transport along the paracellular pathway. Water-driven solute transport was proposed as the major mechanism for electrolyte and nutrient absorption in the small intestine and for Cl- and HCO3- reabsorption in the renal proximal tubule. With the discovery of aquaporins as transcellular route for water transport and the claudin protein family as the major determinant of paracellular transport properties, new mechanistic insights in transepithelial water and solute transport are emerging and call for a reassessment of the solvent drag concept. Current knowledge does not provide a molecular basis for relevant solvent drag-driven, paracellular nutrient, and inorganic anion (re-)absorption. For inorganic cation transport, in contrast, solvent drag along claudin-2-formed paracellular channels appears feasible.
Collapse
Affiliation(s)
- Dorothee Günzel
- Clinical Physiology/Nutritional Medicine, Department of Gastroenterology, Rheumatology and Infectious Diseases, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203, Berlin, Germany.
| |
Collapse
|
18
|
Canse C, Yildirim E, Yaba A. Overview of junctional complexes during mammalian early embryonic development. Front Endocrinol (Lausanne) 2023; 14:1150017. [PMID: 37152932 PMCID: PMC10158982 DOI: 10.3389/fendo.2023.1150017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/28/2023] [Indexed: 05/09/2023] Open
Abstract
Cell-cell junctions form strong intercellular connections and mediate communication between blastomeres during preimplantation embryonic development and thus are crucial for cell integrity, polarity, cell fate specification and morphogenesis. Together with cell adhesion molecules and cytoskeletal elements, intercellular junctions orchestrate mechanotransduction, morphokinetics and signaling networks during the development of early embryos. This review focuses on the structure, organization, function and expressional pattern of the cell-cell junction complexes during early embryonic development. Understanding the importance of dynamic junction formation and maturation processes will shed light on the molecular mechanism behind developmental abnormalities of early embryos during the preimplantation period.
Collapse
Affiliation(s)
- Ceren Canse
- Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Ecem Yildirim
- Department of Histology and Embryology, Yeditepe University Faculty of Medicine, Istanbul, Türkiye
| | - Aylin Yaba
- Department of Histology and Embryology, Yeditepe University Faculty of Medicine, Istanbul, Türkiye
- *Correspondence: Aylin Yaba,
| |
Collapse
|
19
|
Hulme L, Hochstetler A, Schwerk C, Schroten H, Ishikawa H, Tung CY, Perrin B, Blazer-Yost B. Characterization of TRPV4-mediated signaling pathways in an optimized human choroid plexus epithelial cell line. Am J Physiol Cell Physiol 2022; 323:C1823-C1842. [PMID: 35938676 PMCID: PMC9744646 DOI: 10.1152/ajpcell.00193.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 12/14/2022]
Abstract
The objectives of these studies were twofold: 1) to characterize the human choroid plexus papilloma (HIBCPP) cell line as a model of the blood-cerebrospinal fluid barrier (BCSFB) via morphology, tightness, and polarization of transporters in choroid plexus epithelia (CPe), and 2) to utilize Ussing-style electrophysiology to elucidate signaling pathways associated with the activation of the transient receptor potential vanilloid 4 (TRPV4) channel involved in cerebrospinal fluid (CSF) secretion. RT-PCR was implemented to determine gene expression of cell fate markers, junctional complex proteins, and transporters of interest. Scanning electron microscopy and confocal three-dimensional renderings of cultures grown on permeable supports were utilized to delineate the morphology of the brush border, junctional complexes, and polarization of key transporters. Electrophysiology was used to understand and explore TRPV4-mediated signaling in the HIBCPP cell line, considering both short-circuit current (Isc) and conductance responses. HIBCPP cells grown under optimized culture conditions exhibited minimal multilayering, developed an intermediate resistance monolayer, retained differentiation properties, and expressed, and correctly localized, junctional proteins and native transporters. We found that activation of TRPV4 resulted in a robust, multiphasic change in electrogenic ion flux and increase in conductance accompanied by substantial fluid secretion. This response appears to be modulated by a number of different effectors, implicating phospholipase C (PLC), protein kinase C (PKC), and phosphoinositide 3-kinase (PI3K) in TRPV4-mediated ion flux. The HIBCPP cell line is a representative model of the human BCSFB, which can be utilized for studies of transporter function, intracellular signaling, and regulation of CSF production.
Collapse
Affiliation(s)
- Louise Hulme
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| | - Alexandra Hochstetler
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| | - Christian Schwerk
- Department of Pediatrics, Pediatric Infectious Diseases, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Horst Schroten
- Department of Pediatrics, Pediatric Infectious Diseases, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hiroshi Ishikawa
- Department of Neurosurgery, Laboratory of Clinical Regenerative Medicine, University of Tsukuba, Ibaraki, Japan
| | - Chun-Yu Tung
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| | - Benjamin Perrin
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| | - Bonnie Blazer-Yost
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| |
Collapse
|
20
|
Xu W, Zhang Z, Lu Y, Li M, Li J, Tao W. Traditional Chinese medicine Tongxie Yaofang treating irritable bowel syndrome with diarrhea and type 2 diabetes mellitus in rats with liver-depression and spleen-deficiency: A preliminary study. Front Nutr 2022; 9:968930. [PMID: 36438735 PMCID: PMC9686328 DOI: 10.3389/fnut.2022.968930] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 10/10/2022] [Indexed: 10/05/2023] Open
Abstract
Tongxie Yaofang (TXYF), a Traditional Chinese Medicine (TCM) with four components as follows: Rhizoma Atractylodis Macrocephalae (baizhu), Radix Paeoniae Alba (baishao), Pericarpium Citri Reticulatae (chenpi) and Radix Saposhnikovia Divaricata (fangfeng), benefits irritable bowel syndrome (IBS). Nonetheless, proofs of this formula ameliorating D-IBS and T2DM are required. This research aimed at investigating the efficacy of TXYF in treating inflammation in rats with D-IBS and T2DM using animal models. In this study, gavage with high-fat diet, fasciculation, and senna was given to develop rat models with target diseases. To determine intestinal inflammations, major inflammatory factors, and intestinal permeability proteins, H&E staining, ELISA, and immunohistochemistry methods were employed, respectively. This study also utilized Western blot to discover potential inflammatory targets. Results of this research illustrates that TXYF treatment reduced the level of TNF-α, IL-1β, and IL-6, and raised the IL-10 concentration in liver-depressed spleende ficient rats with D-IBS and T2DM, indicating controlled inflammatory reactions. Staining analysis also showed improved disease states of animal models. Furthermore, efficient rebounds of claudin-1, an intestinal permeability-associated protein, were detected. Moreover, TXYF may treat D-IBS and T2DM in rats via the rage pathway.
Collapse
Affiliation(s)
- Weidong Xu
- Department of Traditional Chinese Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Zhiyi Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Ye Lu
- Department of Traditional Chinese Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Mengxi Li
- Department of Traditional Chinese Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jiayao Li
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Wenhua Tao
- Department of Traditional Chinese Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
21
|
Fujikawa M, Sugimoto H, Tamura R, Fujikawa K, Yamagishi A, Ueda Y. Effects of mucopolysaccharide polysulphate on tight junction barrier in human epidermal keratinocytes. Exp Dermatol 2022; 31:1676-1684. [PMID: 35770317 PMCID: PMC9796026 DOI: 10.1111/exd.14637] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 06/20/2022] [Accepted: 06/27/2022] [Indexed: 12/30/2022]
Abstract
Tight junctions (TJs) play important roles in epidermal barrier function and their dysfunction is involved in the pathogenesis of various skin diseases, including atopic dermatitis (AD). Mucopolysaccharide polysulphate (MPS) is the active ingredient of a moisturizing agent used to treat xerosis in patients with AD; however, its mechanism of action on TJ barrier function remains unclear. To elucidate the effects of MPS on TJs, adult human epidermal keratinocyte (HEKa) cells were exposed to MPS, subjected to Western blotting and quantitative PCR analyses for the investigation of TJ-related factors. MPS treatment significantly increased the mRNA and protein expression of claudin-1 (CLDN1) and zonula occludens-1, and significantly increased transepithelial electrical resistance (TEER), which indicates TJ integrity. Conversely, the sulphated and non-sulphated glycosaminoglycans, chondroitin sulphate and hyaluronic acid, respectively, had little effect on TEER or the expression of mRNAs or TJ-related proteins. Interestingly, MPS treatment also inactivated the extracellular signal-regulated kinase signalling pathway, which is known to negatively regulate CLDN1 expression. Furthermore, MPS notably improved the reduction in CLDN1 expression and TEER caused by histamine, which is upregulated in the skin of patients with AD and is known to disrupt the TJ barrier function. Taken together, these findings demonstrate that treatment with the moisturizing agent, MPS, can repair TJ dysfunction and could therefore represent a new therapeutic option for treating patients with AD.
Collapse
Affiliation(s)
| | | | - Rie Tamura
- Kyoto R&D Center, Maruho Co., Ltd.KyotoJapan
| | | | | | - Yuhki Ueda
- Kyoto R&D Center, Maruho Co., Ltd.KyotoJapan
| |
Collapse
|
22
|
Mucosal expression of Ca and P transporters and claudins in the small intestine of broilers is altered by dietary Ca:P in a limestone particle size dependent manner. PLoS One 2022; 17:e0273852. [PMID: 36048795 PMCID: PMC9436080 DOI: 10.1371/journal.pone.0273852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 08/16/2022] [Indexed: 12/01/2022] Open
Abstract
High calcium (Ca) intake and fine limestone reduces precaecal phosphorus (P) absorption independently of P solubility in broilers. This study aimed to determine whether dietary total Ca: total P ratio (Ca:P) and limestone particle size (LPS) affect gene expression of P transporters in the small intestine. A total of 384 one-day-old Ross 308 male broiler chickens received diets low (0.50), medium (1.00) or high (1.75) in Ca:P containing either fine (160 μm) or coarse (1062 μm) limestone, in a 3×2 factorial arrangement. Expression of Ca- and P-related genes were determined using real-time quantitative PCR (RT-qPCR) in duodenum and jejunum. Increasing dietary Ca:P decreased duodenal calcium-sensing receptor (CaSR), calbindin-D28k (CaBP-D28k), plasma membrane Ca-ATPase 1 (PMCA1) and sodium-coupled P cotransporter type IIb (NaPi-IIb), but not transient receptor potential canonical 1 (TRPC1) mRNA. This effect was greater with fine limestone when Ca:P increased from low to medium, but greater with coarse limestone when increased from medium to high. A similar inhibitory effect was observed for jejunal CaBP-D28k expression where increasing dietary Ca:P and fine limestone decreased CaSR mRNA, while dietary Ca:P decreased TRPC1 mRNA only for coarse limestone. It also decreased jejunal NaPi-IIb mRNA irrespective of LPS. Dietary treatments did not affect jejunal PMCA1 mRNA expression or that of inorganic phosphate transporter 1 and 2 and xenotropic and polytropic retrovirus receptor 1 in both intestinal segments. Dietary Ca increase reduced mucosal claudin-2 mRNA in both segments, and jejunal zonula occludens-1 (ZO-1) mRNA only for coarse limestone. In conclusion, increasing dietary Ca:P reduced expression of duodenal P transporters (NaPi-IIb) in a LPS dependent manner, hence Ca induced reduction in intestinal P absorption is mediated by decreasing P transporters expression. Dietary Ca reduces Ca digestibility by downregulating mRNA expression of both Ca permeable claudin-2 and Ca transporters (CaBP-D28k, PMCA1).
Collapse
|
23
|
Gonschior H, Schmied C, Van der Veen RE, Eichhorst J, Himmerkus N, Piontek J, Günzel D, Bleich M, Furuse M, Haucke V, Lehmann M. Nanoscale segregation of channel and barrier claudins enables paracellular ion flux. Nat Commun 2022; 13:4985. [PMID: 36008380 PMCID: PMC9411157 DOI: 10.1038/s41467-022-32533-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 08/04/2022] [Indexed: 11/09/2022] Open
Abstract
The paracellular passage of ions and small molecules across epithelia is controlled by tight junctions, complex meshworks of claudin polymers that form tight seals between neighboring cells. How the nanoscale architecture of tight junction meshworks enables paracellular passage of specific ions or small molecules without compromising barrier function is unknown. Here we combine super-resolution stimulated emission depletion microscopy in live and fixed cells and tissues, multivariate classification of super-resolution images and fluorescence resonance energy transfer to reveal the nanoscale organization of tight junctions formed by mammalian claudins. We show that only a subset of claudins can assemble into characteristic homotypic meshworks, whereas tight junctions formed by multiple claudins display nanoscale organization principles of intermixing, integration, induction, segregation, and exclusion of strand assemblies. Interestingly, channel-forming claudins are spatially segregated from barrier-forming claudins via determinants mainly encoded in their extracellular domains also known to harbor mutations leading to human diseases. Electrophysiological analysis of claudins in epithelial cells suggests that nanoscale segregation of distinct channel-forming claudins enables barrier function combined with specific paracellular ion flux across tight junctions. Meshworks of claudin polymers control the paracellular transport and barrier properties of epithelial tight junctions. Here, the authors show different claudin nanoscale organization principles, finding that claudin segregation enables barrier formation and paracellular ion flux across tight junctions.
Collapse
Affiliation(s)
- Hannes Gonschior
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany
| | - Christopher Schmied
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany
| | | | - Jenny Eichhorst
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany
| | - Nina Himmerkus
- Institute of Physiology, Christian-Albrechts-Universität zu Kiel, 24118, Kiel, Germany
| | - Jörg Piontek
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité - Universitätsmedizin Berlin, 12203, Berlin, Germany
| | - Dorothee Günzel
- Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology, Rheumatology, Charité - Universitätsmedizin Berlin, 12203, Berlin, Germany
| | - Markus Bleich
- Institute of Physiology, Christian-Albrechts-Universität zu Kiel, 24118, Kiel, Germany
| | - Mikio Furuse
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi, 444-8787, Japan.,Department of Physiological Sciences, School of Life Science, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi, 444-8585, Japan
| | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany.,Faculty of Biology, Chemistry and Pharmacy, Freie Universität Berlin, 14195, Berlin, Germany
| | - Martin Lehmann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125, Berlin, Germany.
| |
Collapse
|
24
|
He J, Zhang Y, Hu Z, Zhang L, Shao G, Xie Z, Nie Y, Li W, Li Y, Chen L, Huang B, Chu F, Feng K, Lin W, Li H, Chen W, Zhang X, Xie Q. Recombinant Muscovy Duck Parvovirus Led to Ileac Damage in Muscovy Ducklings. Viruses 2022; 14:v14071471. [PMID: 35891451 PMCID: PMC9315717 DOI: 10.3390/v14071471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 06/29/2022] [Accepted: 06/29/2022] [Indexed: 02/04/2023] Open
Abstract
Waterfowl parvovirus (WPFs) has multiple effects on the intestinal tract, but the effects of recombinant Muscovy duck parvovirus (rMDPV) have not been elucidated. In this study, 48 one-day-old Muscovy ducklings were divided into an infected group and a control group. Plasma and ileal samples were collected from both groups at 2, 4, 6, and 8 days post-infection (dpi), both six ducklings at a time. Next, we analyzed the genomic sequence of the rMDPV strain. Results showed that the ileal villus structure was destroyed seriously at 4, 6, 8 dpi, and the expression of ZO-1, Occludin, and Claudin-1 decreased at 4, 6 dpi; 4, 6, 8 dpi; and 2, 6 dpi, respectively. Intestinal cytokines IFN-α, IL-1β and IL-6 increased at 6 dpi; 8 dpi; and 6, 8 dpi, respectively, whereas IL-2 decreased at 6, 8 dpi. The diversity of ileal flora increased significantly at 4 dpi and decreased at 8 dpi. The bacteria Ochrobactrum and Enterococcus increased and decreased at 4, 8 dpi; 2, 4 dpi, respectively. Plasma MDA increased at 2 dpi, SOD, CAT, and T-AOC decreased at 2, 4, 8 dpi; 4, 8 dpi; and 4, 6, 8 dpi, respectively. These results suggest that rMDPV infection led to early intestinal barrier dysfunction, inflammation, ileac microbiota disruption, and oxidative stress.
Collapse
Affiliation(s)
- Jiahui He
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.H.); (Y.Z.); (Z.H.); (G.S.); (Z.X.); (Y.N.); (W.L.); (Y.L.); (L.C.); (B.H.); (F.C.); (K.F.); (W.L.); (H.L.); (W.C.)
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, China
| | - Yukun Zhang
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.H.); (Y.Z.); (Z.H.); (G.S.); (Z.X.); (Y.N.); (W.L.); (Y.L.); (L.C.); (B.H.); (F.C.); (K.F.); (W.L.); (H.L.); (W.C.)
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, China
| | - Zezhong Hu
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.H.); (Y.Z.); (Z.H.); (G.S.); (Z.X.); (Y.N.); (W.L.); (Y.L.); (L.C.); (B.H.); (F.C.); (K.F.); (W.L.); (H.L.); (W.C.)
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, China
| | - Luxuan Zhang
- School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou 510006, China;
| | - Guanming Shao
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.H.); (Y.Z.); (Z.H.); (G.S.); (Z.X.); (Y.N.); (W.L.); (Y.L.); (L.C.); (B.H.); (F.C.); (K.F.); (W.L.); (H.L.); (W.C.)
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, China
| | - Zi Xie
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.H.); (Y.Z.); (Z.H.); (G.S.); (Z.X.); (Y.N.); (W.L.); (Y.L.); (L.C.); (B.H.); (F.C.); (K.F.); (W.L.); (H.L.); (W.C.)
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, China
| | - Yu Nie
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.H.); (Y.Z.); (Z.H.); (G.S.); (Z.X.); (Y.N.); (W.L.); (Y.L.); (L.C.); (B.H.); (F.C.); (K.F.); (W.L.); (H.L.); (W.C.)
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, China
| | - Wenxue Li
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.H.); (Y.Z.); (Z.H.); (G.S.); (Z.X.); (Y.N.); (W.L.); (Y.L.); (L.C.); (B.H.); (F.C.); (K.F.); (W.L.); (H.L.); (W.C.)
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, China
| | - Yajuan Li
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.H.); (Y.Z.); (Z.H.); (G.S.); (Z.X.); (Y.N.); (W.L.); (Y.L.); (L.C.); (B.H.); (F.C.); (K.F.); (W.L.); (H.L.); (W.C.)
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, China
| | - Liyi Chen
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.H.); (Y.Z.); (Z.H.); (G.S.); (Z.X.); (Y.N.); (W.L.); (Y.L.); (L.C.); (B.H.); (F.C.); (K.F.); (W.L.); (H.L.); (W.C.)
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, China
| | - Benli Huang
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.H.); (Y.Z.); (Z.H.); (G.S.); (Z.X.); (Y.N.); (W.L.); (Y.L.); (L.C.); (B.H.); (F.C.); (K.F.); (W.L.); (H.L.); (W.C.)
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, China
| | - Fengsheng Chu
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.H.); (Y.Z.); (Z.H.); (G.S.); (Z.X.); (Y.N.); (W.L.); (Y.L.); (L.C.); (B.H.); (F.C.); (K.F.); (W.L.); (H.L.); (W.C.)
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, China
| | - Keyu Feng
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.H.); (Y.Z.); (Z.H.); (G.S.); (Z.X.); (Y.N.); (W.L.); (Y.L.); (L.C.); (B.H.); (F.C.); (K.F.); (W.L.); (H.L.); (W.C.)
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, China
- Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou 510642, China
| | - Wencheng Lin
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.H.); (Y.Z.); (Z.H.); (G.S.); (Z.X.); (Y.N.); (W.L.); (Y.L.); (L.C.); (B.H.); (F.C.); (K.F.); (W.L.); (H.L.); (W.C.)
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, China
- Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou 510642, China
| | - Hongxin Li
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.H.); (Y.Z.); (Z.H.); (G.S.); (Z.X.); (Y.N.); (W.L.); (Y.L.); (L.C.); (B.H.); (F.C.); (K.F.); (W.L.); (H.L.); (W.C.)
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, China
- Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou 510642, China
| | - Weiguo Chen
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.H.); (Y.Z.); (Z.H.); (G.S.); (Z.X.); (Y.N.); (W.L.); (Y.L.); (L.C.); (B.H.); (F.C.); (K.F.); (W.L.); (H.L.); (W.C.)
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, China
- Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou 510642, China
| | - Xinheng Zhang
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.H.); (Y.Z.); (Z.H.); (G.S.); (Z.X.); (Y.N.); (W.L.); (Y.L.); (L.C.); (B.H.); (F.C.); (K.F.); (W.L.); (H.L.); (W.C.)
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, China
- Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou 510642, China
- Correspondence: (X.Z.); (Q.X.)
| | - Qingmei Xie
- Heyuan Branch, Guangdong Provincial Laboratory of Lingnan Modern Agricultural Science and Technology, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (J.H.); (Y.Z.); (Z.H.); (G.S.); (Z.X.); (Y.N.); (W.L.); (Y.L.); (L.C.); (B.H.); (F.C.); (K.F.); (W.L.); (H.L.); (W.C.)
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, Guangzhou 510642, China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou 510642, China
- Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou 510642, China
- Correspondence: (X.Z.); (Q.X.)
| |
Collapse
|
25
|
Jafari NV, Rohn JL. The urothelium: a multi-faceted barrier against a harsh environment. Mucosal Immunol 2022; 15:1127-1142. [PMID: 36180582 PMCID: PMC9705259 DOI: 10.1038/s41385-022-00565-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/18/2022] [Accepted: 08/28/2022] [Indexed: 02/04/2023]
Abstract
All mucosal surfaces must deal with the challenge of exposure to the outside world. The urothelium is a highly specialized layer of stratified epithelial cells lining the inner surface of the urinary bladder, a gruelling environment involving significant stretch forces, osmotic and hydrostatic pressures, toxic substances, and microbial invasion. The urinary bladder plays an important barrier role and allows the accommodation and expulsion of large volumes of urine without permitting urine components to diffuse across. The urothelium is made up of three cell types, basal, intermediate, and umbrella cells, whose specialized functions aid in the bladder's mission. In this review, we summarize the recent insights into urothelial structure, function, development, regeneration, and in particular the role of umbrella cells in barrier formation and maintenance. We briefly review diseases which involve the bladder and discuss current human urothelial in vitro models as a complement to traditional animal studies.
Collapse
Affiliation(s)
- Nazila V Jafari
- Department of Renal Medicine, Division of Medicine, University College London, Royal Free Hospital Campus, London, UK
| | - Jennifer L Rohn
- Department of Renal Medicine, Division of Medicine, University College London, Royal Free Hospital Campus, London, UK.
| |
Collapse
|
26
|
Jo CH, Kim S, Kim GH. Claudins in kidney health and disease. Kidney Res Clin Pract 2022; 41:275-287. [PMID: 35354245 PMCID: PMC9184838 DOI: 10.23876/j.krcp.21.279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/29/2021] [Indexed: 11/04/2022] Open
Abstract
Claudins are strategically located to exert their physiologic actions along with the nephron segments from the glomerulus. Claudin-1 is normally located in the Bowman’s capsule, but its overexpression can reach the podocytes and lead to albuminuria. In the proximal tubule (PT), claudin-2 forms paracellular channels selective for water, Na+, K+, and Ca2+. Claudin-2 gene mutations are associated with hypercalciuria and kidney stones. Claudin-10 has two splice variants, -10a and -10b; Claudin-10a acts as an anion-selective channel in the PT, and claudin-10b functions as a cation-selective pore in the thick ascending limb (TAL). Claudin-16 and claudin-19 mediate paracellular transport of Na+, Ca2+, and Mg2+ in the TAL, where the expression of claudin-3/16/19 and claudin-10b are mutually exclusive. The claudin-16 or -19 mutation causes familial hypomagnesemia with hypercalciuria and nephrocalcinosis. Claudin-14 polymorphisms have been linked to increased risk of hypercalciuria. Claudin-10b mutations produce HELIX syndrome, which encompasses hypohidrosis, electrolyte imbalance, lacrimal gland dysfunction, ichthyosis, and xerostomia. Hypercalciuria and magnesuria in metabolic acidosis are related to downregulation of PT and TAL claudins. In the TAL, stimulation of calcium-sensing receptors upregulates claudin-14 and negatively acts on the claudin-16/19 complex. Claudin-3 acts as a general barrier to ions in the collecting duct. If this barrier is disturbed, urine acidification might be impaired. Claudin-7 forms a nonselective paracellular channel facilitating Cl– and Na+ reabsorption in the collecting ducts. Claudin-4 and -8 serve as anion channels and mediate paracellular Cl– transport; their upregulation may contribute to pseudohypoaldosteronism II and salt-sensitive hypertension.
Collapse
Affiliation(s)
- Chor ho Jo
- Hanyang Biomedical Research Institute, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Sua Kim
- Hanyang Biomedical Research Institute, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Gheun-Ho Kim
- Hanyang Biomedical Research Institute, Hanyang University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
- Correspondence: Gheun-Ho Kim Department of Internal Medicine, Hanyang University College of Medicine, 222-1 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea. E-mail:
| |
Collapse
|
27
|
Ishikawa S, Nikaido M, Otani T, Ogata K, Iida H, Inai Y, Tamaoki S, Inai T. Inhibition of Retinoid X Receptor Improved the Morphology, Localization of Desmosomal Proteins, and Paracellular Permeability in Three-Dimensional Cultures of Mouse Keratinocytes. Microscopy (Oxf) 2022; 71:152-160. [PMID: 35289919 PMCID: PMC9169536 DOI: 10.1093/jmicro/dfac007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/04/2022] [Accepted: 02/16/2022] [Indexed: 11/14/2022] Open
Abstract
Retinoic acid (RA) plays an important role in epithelial homeostasis and influences the morphology, proliferation, differentiation and permeability of epithelial cells. Mouse keratinocytes, K38, reconstituted non-keratinized stratified epithelium in three-dimensional (3D) cultures with serum, which contains retinol (a source of RA), but the morphology was different from in vivo epithelium. The formed epithelium was thick, with loosened cell–cell contacts. Here, we investigated whether the inhibition of RA receptor (RAR)/retinoid X receptor (RXR)-mediated signaling by an RXR antagonist, HX 531, improved K38 3D cultures in terms of morphology and intercellular junctions. The epithelium formed by 0.5 μM HX531 was thin, and the intercellular space was narrowed because of the restoration of the layer-specific distribution of desmoglein (DSG)-1, DSG3 and plakoglobin (PG). Moreover, the levels of desmosomal proteins and tight junction proteins, including DSG1, DSG2, DSG3, PG, claudin (CLDN)-1 and CLDN4 increased, but the adherens junction protein, E-cadherin, did not show any change. Furthermore, CLDN1 was recruited to occludin-positive cell–cell contacts in the superficial cells and transepithelial electrical resistance was increased. Therefore, K38 3D cultures treated with 0.5 μM HX531 provides a useful in vitro model to study intercellular junctions in the non-keratinized epithelium.
Collapse
Affiliation(s)
- Shoko Ishikawa
- Department of Oral Growth and Development, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
| | - Misaki Nikaido
- Department of Odontology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
| | - Takahito Otani
- Department of Morphological Biology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
| | - Kayoko Ogata
- Department of Morphological Biology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, 814-0193, Japan
| | - Hiroshi Iida
- Laboratory of Zoology, Graduate School of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Yuko Inai
- Division of General Dentistry, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Sachio Tamaoki
- Department of Oral Growth and Development, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
| | - Tetsuichiro Inai
- Department of Morphological Biology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, 814-0193, Japan
| |
Collapse
|
28
|
Chen X, Wang Y, Chen D, Yu B, Huang Z. Dietary ferulic acid supplementation improves intestinal antioxidant capacity and intestinal barrier function in weaned piglets. Anim Biotechnol 2021; 33:356-361. [PMID: 34802366 DOI: 10.1080/10495398.2021.2003807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
This study was conducted to explore the effects of dietary ferulic acid (FA) supplementation on intestinal antioxidant capacity and intestinal barrier function in weaned piglets. Eighteen 21-day-old castrated male DLY (Duroc × Landrace × Yorkshire) weaned piglets were randomly divided into control, 0.05% FA, and 0.45% FA groups, respectively. The experiment lasted for 5 weeks. The results showed that dietary 0.05 and 0.45% FA supplementation significantly increased catalase activity (p < 0.001), the protein levels of nuclear factor E2-related factor 2 (Nrf2) and NAD(P)H quinone dehydrogenase 1 (p < 0.05), and the mRNA levels of superoxide dismutase 1, glutathione reductase and Nrf2 (p < 0.05) in jejunum when compared with the control group. Dietary 0.05% FA supplementation also increased the mRNA level of glutathione S-transferase (p < 0.05) in jejunum. Meanwhile, Dietary 0.05 and 0.45% FA supplementation significantly increased the protein expression of zonula occludens 1 (ZO-1) (p < 0.05), and dietary supplementation of 0.05% FA increased the mRNA levels of ZO-1, zonula occludens 2, mucin 1, mucin 2, occluding, and claudin-1 (p < 0.05) in jejunum. Together, our data suggest that dietary 0.05% FA supplementation improves the intestinal antioxidant capacity and intestinal barrier function of weaned piglets.
Collapse
Affiliation(s)
- Xiaoling Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Youxia Wang
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Daiwen Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Bing Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhiqing Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
29
|
High-Fat Diet Induces Disruption of the Tight Junction-Mediated Paracellular Barrier in the Proximal Small Intestine Before the Onset of Type 2 Diabetes and Endotoxemia. Dig Dis Sci 2021; 66:3359-3374. [PMID: 33104937 DOI: 10.1007/s10620-020-06664-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 10/06/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND/AIM A link between an impaired intestinal barrier, endotoxemia, and the pathogenesis of metabolic diseases, such as type 2 diabetes mellitus (T2DM), has been proposed. In previous work, we have demonstrated that the tight junction (TJ)-mediated intestinal barrier in ileum/colon was marginally changed in prediabetic mice; therefore, it does not seem to mainly contribute to the T2DM onset. In this study, the TJ-mediated epithelial barrier in the duodenum and jejunum was evaluated in mice during the development of type 2 prediabetes. METHODS/RESULTS HF diet induced prediabetes after 60 days associated with a significant rise in intestinal permeability to the small-sized marker Lucifer yellow in these mice, with no histological signs of mucosal inflammation or rupture of the proximal intestine epithelium. As revealed by immunofluorescence, TJ proteins, such as claudins-1, -2, -3, and ZO-1, showed a significant decrease in junctional content in duodenum and jejunum epithelia, already after 15 days of treatment, suggesting a rearrangement of the TJ structure. However, no significant change in total cell content of these proteins was observed in intestinal epithelium homogenates, as assessed by immunoblotting. Despite the changes in intestinal permeability and TJ structure, the prediabetic mice showed similar LPS, zonulin, and TNF-α levels in plasma or adipose tissue, and in intestinal segments as compared to the controls. CONCLUSION Disruption of the TJ-mediated paracellular barrier in the duodenum and jejunum is an early event in prediabetes development, which occurs in the absence of detectable endotoxemia/inflammation and may contribute to the HF diet-induced increase in intestinal permeability.
Collapse
|
30
|
Monaco A, Ovryn B, Axis J, Amsler K. The Epithelial Cell Leak Pathway. Int J Mol Sci 2021; 22:ijms22147677. [PMID: 34299297 PMCID: PMC8305272 DOI: 10.3390/ijms22147677] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 01/08/2023] Open
Abstract
The epithelial cell tight junction structure is the site of the transepithelial movement of solutes and water between epithelial cells (paracellular permeability). Paracellular permeability can be divided into two distinct pathways, the Pore Pathway mediating the movement of small ions and solutes and the Leak Pathway mediating the movement of large solutes. Claudin proteins form the basic paracellular permeability barrier and mediate the movement of small ions and solutes via the Pore Pathway. The Leak Pathway remains less understood. Several proteins have been implicated in mediating the Leak Pathway, including occludin, ZO proteins, tricellulin, and actin filaments, but the proteins comprising the Leak Pathway remain unresolved. Many aspects of the Leak Pathway, such as its molecular mechanism, its properties, and its regulation, remain controversial. In this review, we provide a historical background to the evolution of the Leak Pathway concept from the initial examinations of paracellular permeability. We then discuss current information about the properties of the Leak Pathway and present current theories for the Leak Pathway. Finally, we discuss some recent research suggesting a possible molecular basis for the Leak Pathway.
Collapse
Affiliation(s)
- Ashley Monaco
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Boulevard, Old Westbury, NY 11568, USA; (A.M.); (J.A.)
| | - Ben Ovryn
- Department of Physics, New York Institute of Technology, Northern Boulevard, Old Westbury, NY 11568, USA;
| | - Josephine Axis
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Boulevard, Old Westbury, NY 11568, USA; (A.M.); (J.A.)
| | - Kurt Amsler
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Northern Boulevard, Old Westbury, NY 11568, USA; (A.M.); (J.A.)
- Correspondence: ; Tel.: +1-516-686-3716
| |
Collapse
|
31
|
Domínguez G, Cardiel E, Sánchez E, Hernández PR. Assessment of the effects of exposure to extremely low-frequency magnetic fields on MDCK epithelial cell lines under a controlled environment. JOURNAL OF RADIATION RESEARCH 2021; 62:259-268. [PMID: 33592097 PMCID: PMC7948907 DOI: 10.1093/jrr/rrab001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 12/08/2020] [Indexed: 06/12/2023]
Abstract
To assess the effects of exposure to extremely low-frequency magnetic fields (ELF-MFs) on MDCK cell lines, experiments were performed in a chamber under controlled conditions (temperature, humidity and CO2). Therefore, the measured physicochemical and electrical changes in the cells are due solely to the magnetic field exposure and not to external factors. A developed sinusoidal magnetic field generator produced the ELF-MFs with a uniform magnetic field and adjustable intensity and frequency. Three experimental indicators were used: (i) transepithelial electrical impedance (TEEI); (ii) cell migration and proliferation; and (iii) expression of the proteins of the tight junctions, and changes in the area and shape of the cell nuclei. No significant effects on TEEI values were observed when 10 and 50 G 60 Hz magnetic fields were applied to confluent cell monolayers. There were no significant differences in migration and proliferation of the cell monolayer exposed to 60 Hz magnetic fields10 and 50 G , but a contact inhibition factor was observed. The expression of the CLDN-1 protein decreased by 90% compared with the control, while ZO-1 protein expression increased by 120%. No significant effects were observed in the area and shape of the cell nuclei. Experimentation in a controlled environment, under physiological conditions, ensures that the observed effects were strictly due to exposure to magnetic fields. Different exposure conditions are necessary to determine the impact on TEEI and cell migration-proliferation indicators.
Collapse
Affiliation(s)
- Gonzalo Domínguez
- Department of Electrical Engineering, Bioelectronics section, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, Mexico
| | - Eladio Cardiel
- Department of Electrical Engineering, Bioelectronics section, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, Mexico
| | - Elsa Sánchez
- Department of Physiology, Biophysics, and Neuroscience, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, Mexico
| | | |
Collapse
|
32
|
Chan YJ, Liao PL, Tsai CH, Cheng YW, Lin FL, Ho JD, Chen CY, Li CH. Titanium dioxide nanoparticles impair the inner blood-retinal barrier and retinal electrophysiology through rapid ADAM17 activation and claudin-5 degradation. Part Fibre Toxicol 2021; 18:4. [PMID: 33422125 PMCID: PMC7796566 DOI: 10.1186/s12989-020-00395-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 12/21/2020] [Indexed: 11/30/2022] Open
Abstract
Background Depending on their distinct properties, titanium dioxide nanoparticles (TiO2-NPs) are manufactured extensively and widely present in our daily necessities, with growing environmental release and public concerns. In sunscreen formulations, supplementation of TiO2-NPs may reach up to 25% (w/w). Ocular contact with TiO2-NPs may occur accidentally in certain cases, allowing undesirable risks to human vision. This study aimed to understand the barrier integrity of retinal endothelial cells in response to TiO2-NP exposure. bEnd.3 cells and human retinal endothelial cells (HRECs) were exposed to TiO2-NP, followed by examination of their tight junction components and functions. Results TiO2-NP treatment apparently induced a broken structure of the junctional plaques, conferring decreased transendothelial electrical resistance, a permeable paracellular cleft, and improved cell migration in vitro. This might involve rapid activation of metalloproteinase, a disintegrin and metalloproteinase 17 (ADAM17), and ADAM17-mediated claudin-5 degradation. For the in vivo study, C57BL/6 mice were administered a single dose of TiO2-NP intravitreally and then subjected to a complete ophthalmology examination. Fluorescein leakage and reduced blood flow at the optical disc indicated a damaged inner blood-retinal barrier induced by TiO2-NPs. Inappreciable change in the thickness of retinal sublayers and alleviated electroretinography amplitude were observed in the TiO2-NP-treated eyes. Conclusions Overall, our data demonstrate that TiO2-NP can damage endothelial cell function, thereby affecting retinal electrophysiology. Supplementary Information The online version contains supplementary material available at 10.1186/s12989-020-00395-7.
Collapse
Affiliation(s)
- Yen-Ju Chan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 110, Taiwan
| | - Po-Lin Liao
- School of Pharmacy, Taipei Medical University, Taipei, Taiwan.,Institute of Food Safety and Health Risk Assessment, School of Pharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Chi-Hao Tsai
- School of Pharmacy, Taipei Medical University, Taipei, Taiwan.,Institute of Food Safety and Health Risk Assessment, School of Pharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Wen Cheng
- School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Fan-Li Lin
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Jau-Der Ho
- Department of Ophthalmology, Taipei Medical University, Taipei, Taiwan
| | - Ching-Yi Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 110, Taiwan.,School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Ching-Hao Li
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan. .,Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 110, Taiwan.
| |
Collapse
|
33
|
Wang Y, Wang D, Wang J, Li K, Heng C, Jiang L, Cai C, Zhan X. Effects of different stocking densities on tracheal barrier function and its metabolic changes in finishing broilers. Poult Sci 2020; 99:6307-6316. [PMID: 33248546 PMCID: PMC7704944 DOI: 10.1016/j.psj.2020.09.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/11/2020] [Accepted: 09/03/2020] [Indexed: 12/22/2022] Open
Abstract
In the present study, we evaluated the effects of various stocking densities on the tracheal barrier and plasma metabolic profiles of finishing broilers. We randomly assigned 1,440 Lingnan Yellow feathered broilers (age 22 d) to 5 different stocking density groups (8 m-2, 10 m-2, 12 m-2, 14 m-2, and 16 m-2). Each of these consisted of 3 replicates. The interleukin (IL)-1β and IL-10 concentrations were substantially higher in the 16 m-2 treatment group than they were in the 8 m-2 and 10 m-2 treatment groups (P < 0.05). Nevertheless, IL-4 did not significantly differ among the 5 treatments (P > 0.05). The tracheal mucosae of the birds in the 16 m-2 group (high stocking density, HSD) were considerably thicker than those for the birds in the 10 m-2 group (control, CSD). Relative to CSD, the claudin1 expression level was lower, and the muc2 and caspase3 expression levels were higher for HSD. Compared with CSD, 10 metabolites were significantly upregulated (P < 0.05), and 7 were significantly downregulated (P < 0.05) in HSD. Most of these putative diagnostic biomarkers were implicated in matter biosynthesis and energy metabolism. A metabolic pathway analysis revealed that the most relevant and critical biomarkers were pentose and glucuronate interconversions and the pentose phosphate pathway. Activation of the aforementioned pathways may partially counteract the adverse effects of the stress induced by high stocking density. This work helped improve our understanding of the harmful effects of high stocking density on the tracheal barrier and identified 2 metabolic pathways that might be associated with high stocking density-induced metabolic disorders in broilers.
Collapse
Affiliation(s)
- Yuanyuan Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Dianchun Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Jiangshui Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Kaixuan Li
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Chianning Heng
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Lei Jiang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Chenhao Cai
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Xiuan Zhan
- College of Animal Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
34
|
Ramírez-Flores CJ, Cruz-Mirón R, Lagunas-Cortés N, Mondragón-Castelán M, Mondragon-Gonzalez R, González-Pozos S, Mondragón-Flores R. Toxoplasma gondii excreted/secreted proteases disrupt intercellular junction proteins in epithelial cell monolayers to facilitate tachyzoites paracellular migration. Cell Microbiol 2020; 23:e13283. [PMID: 33108050 DOI: 10.1111/cmi.13283] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/14/2020] [Accepted: 10/23/2020] [Indexed: 12/19/2022]
Abstract
Toxoplasma gondii shows high dissemination and migration properties across biological barriers infecting immunologically privileged organs. Toxoplasma uses different routes for dissemination; however, the mechanisms are not fully understood. Herein, we studied the effects of proteases present in excretion/secretion products (ESPs) of Toxoplasma on MDCK cell monolayers. Ultrastructural analysis showed that ESPs of Toxoplasma disrupt the intercellular junctions (IJ) of adjacent cells. The tight junction (TJ) proteins ZO-1, occludin, and claudin-1 suffered a progressive decrease in protein levels upon ESPs treatment. In addition, ESPs induced mislocalization of such TJ proteins, along with the adherent junction protein E-cadherin, and this was prevented by pre-treating the ESPs with protease inhibitors. Reorganisation of cytoskeleton proteins was also observed. Endocytosis inhibitors, Dyngo®-4a and Dynasore, impeded the modifications, suggesting that TJ proteins internalisation is triggered by the ESPs proteases hence contributing to the loss of IJ. The observed disruption in TJ proteins went in line with a decrease in the transepithelial electrical resistance of the monolayers, which was significantly blocked by pre-treating ESPs with metalloprotease and serine protease inhibitors. Moreover, exposure of cell monolayers to ESPs facilitated paracellular migration of tachyzoites. Our results demonstrate that Toxoplasma ESPs contain proteases that can disrupt the IJ of epithelial monolayers and this could facilitate the paracellular route for Toxoplasma tissue dissemination and migration.
Collapse
Affiliation(s)
- Carlos J Ramírez-Flores
- Departamento de Bioquímica, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México, Mexico
| | - Rosalba Cruz-Mirón
- Departamento de Bioquímica, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México, Mexico
| | - Noé Lagunas-Cortés
- Departamento de Bioquímica, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México, Mexico
| | - Mónica Mondragón-Castelán
- Departamento de Bioquímica, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México, Mexico
| | - Ricardo Mondragon-Gonzalez
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México, Mexico
| | | | - Ricardo Mondragón-Flores
- Departamento de Bioquímica, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México, Mexico
| |
Collapse
|
35
|
Dalghi MG, Montalbetti N, Carattino MD, Apodaca G. The Urothelium: Life in a Liquid Environment. Physiol Rev 2020; 100:1621-1705. [PMID: 32191559 PMCID: PMC7717127 DOI: 10.1152/physrev.00041.2019] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/02/2020] [Accepted: 03/14/2020] [Indexed: 02/08/2023] Open
Abstract
The urothelium, which lines the renal pelvis, ureters, urinary bladder, and proximal urethra, forms a high-resistance but adaptable barrier that surveils its mechanochemical environment and communicates changes to underlying tissues including afferent nerve fibers and the smooth muscle. The goal of this review is to summarize new insights into urothelial biology and function that have occurred in the past decade. After familiarizing the reader with key aspects of urothelial histology, we describe new insights into urothelial development and regeneration. This is followed by an extended discussion of urothelial barrier function, including information about the roles of the glycocalyx, ion and water transport, tight junctions, and the cellular and tissue shape changes and other adaptations that accompany expansion and contraction of the lower urinary tract. We also explore evidence that the urothelium can alter the water and solute composition of urine during normal physiology and in response to overdistension. We complete the review by providing an overview of our current knowledge about the urothelial environment, discussing the sensor and transducer functions of the urothelium, exploring the role of circadian rhythms in urothelial gene expression, and describing novel research tools that are likely to further advance our understanding of urothelial biology.
Collapse
Affiliation(s)
- Marianela G Dalghi
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Nicolas Montalbetti
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Marcelo D Carattino
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Gerard Apodaca
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
36
|
Moinard A, Payen C, Ouguerram K, André A, Hernandez J, Drut A, Biourge VC, Suchodolski JS, Flanagan J, Nguyen P, Leray V. Effects of High-Fat Diet at Two Energetic Levels on Fecal Microbiota, Colonic Barrier, and Metabolic Parameters in Dogs. Front Vet Sci 2020; 7:566282. [PMID: 33102570 PMCID: PMC7545960 DOI: 10.3389/fvets.2020.566282] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/21/2020] [Indexed: 12/22/2022] Open
Abstract
Increased consumption of energy-rich foods is a key factor in overweight, obesity, and associated metabolic disorders. This would be, at least in part, related to microbiota disturbance. In rodent models of obesity, microbiota disruption has been associated with alteration of the intestinal barrier, endotoxemia, inflammation grade, and insulin sensitivity. The aim of the present study was to assess the effects of a high-fat diet (HFD), fed at two energetic levels, on microbiota, intestinal barrier, and inflammatory and metabolic parameters in dogs. A HFD (33% fat as fed, 4,830 kcal/kg) was given to 24 healthy Beagle dogs at 100% (HF-100; n = 8) and at 150% (HF-150; n = 16) of their maintenance energy requirements for 8 weeks. Analysis of similarity revealed a significant difference in gut microbiota β-diversity following the diet compared to week 0 in both groups while α-diversity was lower only in the HF-150 group. Firmicutes/Bacteroidetes ratio was higher in the HF-150 group compared to the HF-100 group at weeks 2 and 8. A reduction in insulin sensitivity was observed over time in the HF150 group. Neither endotoxemia nor inflammation was observed in either group, did not find supporting data for the hypothesis that the microbiota is involved in the decline of insulin sensitivity through metabolic endotoxemia and low-grade inflammation. Colonic permeability was increased at week 4 in both groups and returned to initial levels at week 8, and was associated with modifications to the expression of genes involved in colonic barrier function. The increase in intestinal permeability may have been caused by the altered intestinal microbiota and increased expression of genes encoding tight junction proteins might indicate a compensatory mechanism to restore normal permeability. Although simultaneous changes to the microbiota, barrier permeability, inflammatory, and metabolic status have not been observed, such a causal link cannot be excluded in dogs overfed on a HFD. Further studies are necessary to better understand the link between HFD, intestinal microbiota and the host.
Collapse
Affiliation(s)
- Alex Moinard
- Nutrition, PhysioPathology and Pharmacology Unit (NP3), Oniris, College of Veterinary Medicine, Food Sciences and Engineering, CRNH, Nantes, France
| | - Cyrielle Payen
- Nutrition, PhysioPathology and Pharmacology Unit (NP3), Oniris, College of Veterinary Medicine, Food Sciences and Engineering, CRNH, Nantes, France
| | - Khadija Ouguerram
- UMR 1280 Physiopathology of Nutritional Adaptations (PhAN), INRAE, CRNH, West Human Nutrition Research Center, CHU, Nantes, France
| | - Agnès André
- Nutrition, PhysioPathology and Pharmacology Unit (NP3), Oniris, College of Veterinary Medicine, Food Sciences and Engineering, CRNH, Nantes, France
| | - Juan Hernandez
- USC 1383 Cellular and Molecular Immunoendocrinology (IECM), INRAE, Oniris, College of Veterinary Medicine, Food Sciences and Engineering, Nantes, France
| | - Amandine Drut
- Nutrition, PhysioPathology and Pharmacology Unit (NP3), Oniris, College of Veterinary Medicine, Food Sciences and Engineering, CRNH, Nantes, France
| | | | - Jan S Suchodolski
- Gastrointestinal Laboratory, Texas A&M University, College Station, TX, United States
| | | | - Patrick Nguyen
- Nutrition, PhysioPathology and Pharmacology Unit (NP3), Oniris, College of Veterinary Medicine, Food Sciences and Engineering, CRNH, Nantes, France
| | - Véronique Leray
- Nutrition, PhysioPathology and Pharmacology Unit (NP3), Oniris, College of Veterinary Medicine, Food Sciences and Engineering, CRNH, Nantes, France
| |
Collapse
|
37
|
Tu Y, Wu W, Guo Y, Lu F, Xu D, Li X, Zhao Y, He L. Upregulation of hsa-miR-31-3p induced by ultraviolet affects keratinocytes permeability barrier by targeting CLDN1. Biochem Biophys Res Commun 2020; 532:626-632. [PMID: 32907715 DOI: 10.1016/j.bbrc.2020.06.113] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 06/23/2020] [Indexed: 01/19/2023]
Abstract
Chronic actinic dermatitis (CAD) is a photoallergic skin disease with complicated pathogenesis. However, skin barrier dysfunction may be involved according to clinical manifestation. To investigate the mechanism of CAD barrier dysfunction, noninvasive detection of skin barrier and small RNA sequencing were carried out. Quantitative real-time PCR (qRT-PCR) was used to evaluate the expression levels of hsa-miR-31-3p and CLDN1. The correlation between hsa-miR-31-3p and CAD severity was explored. Further, dual-luciferase reporter assay was performed to identify the relationship between hsa-miR-31-3p and CLDN1. In addition, expression of hsa-miR-31-3p was detected after ultraviolet (UV) irradiation. Influences of hsa-miR-31-3p on primary human keratinocytes barrier were assessed by FITC-Dextran permeability assay. Moreover, western blot was used to detect the expression of claudin-1, filaggrin, loricrin and involucrin. Our results showed that transepidermal water loss (TEWL) significantly increased in CAD, while stratum corneum hydration (SCH) significantly decreased. The expression of hsa-miR-31-3p was up-regulated in CAD while CLDN1 was down-regulated. Hsa-miR-31-3p was correlated with TEWL, UV-MED (minimal erythema dose) and clinical severity scores of CAD (CSS-CAD). Dual-luciferase reporter assay confirmed that hsa-miR-31-3p targeted the 3'UTR region of CLDN1. Moreover, hsa-miR-31-3p was induced by UVB (0-30 mJ/cm2) and UVA (0-4 J/cm2). Furthermore, overexpression of hsa-miR-31-3p increased FITC-Dextran flux of primary human keratinocytes and reduced the expression of claudin-1, filaggrin, loricrin and involucrin. In conclusion, we demonstrated that hsa-miR-31-3p induced by UV was correlated with CAD severity, which played an important role in regulating keratinocytes permeability barrier through targeting CLDN1.
Collapse
Affiliation(s)
- Yunhua Tu
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China; Department of Dermatology, The Second People's Hospital of Guiyang, Guizhou, 550000, China
| | - Wenjuan Wu
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Yanni Guo
- Department of Dermatology, The Second Affiliated Hospital of Fujian Medical University, Fujian, 675000, China
| | - Fengyan Lu
- Department of Dermatology, The First People's Hospital of Qujing, Qujing, 655000, China
| | - Dan Xu
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Xing Li
- Department of Dermatology, People's Hospital of Chuxiong Yi Autonomous Prefecture, Chuxiong, 675000, China
| | - Yueting Zhao
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Li He
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| |
Collapse
|
38
|
Liu M, Xie W, Wan X, Deng T. Clostridium butyricum protects intestinal barrier function via upregulation of tight junction proteins and activation of the Akt/mTOR signaling pathway in a mouse model of dextran sodium sulfate-induced colitis. Exp Ther Med 2020; 20:10. [PMID: 32934675 PMCID: PMC7471846 DOI: 10.3892/etm.2020.9138] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 07/01/2020] [Indexed: 12/14/2022] Open
Abstract
Clostridium butyricum (CB), a probiotic, is a gram-positive obligate anaerobic bacillus with acid and heat resistant properties. Previous studies have reported that CB has beneficial effects in intestinal diseases and regulates intestinal function. The aim of the present study was to investigate the protective effects and mechanisms of CB on the intestinal barrier function. Mice were randomly divided into three experimental groups (n=15 mice/group), including control, dextran sodium sulfate (DSS) and DSS + CB. In the DSS and DSS + CB groups colitis was induced with 3% DSS dissolved in drinking water for 7 days. DSS + CB group mice were co-treated daily with 200 µl (2x108 CFU) CB solution via gavage. The intestinal mucosal barrier function in mice was assessed by measuring FITC-labeled 4-kDa dextran (molecular weight, 4,000 Da) flux and by analyzing the expression of tight junction (TJ)-related proteins using western blot analysis. In addition, the secretion levels of tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6, IL-10 and IL-13, and the concentration of malondialdehyde, glutathione and superoxide dismutase were detected using ELISAs to determine inflammation and oxidative stress, respectively. The activation status of the Akt/mTOR signaling pathway was also investigated using western blot analysis. The results demonstrated that, in mice with DSS-induced colitis treatment, co-treatment with CB attenuated colitis symptoms and intestinal permeability, increased the expression levels of TJ-related proteins, decreased TNF-α, IL-1β and IL-13 secretion levels but increased those of IL-10, and reduced oxidative stress. Additionally, CB elevated the phosphorylation of Akt, mTOR and p70 ribosomal protein S6 kinase. Collectively, the present results indicated that CB protected intestinal barrier function and decreased intestinal mucosal permeability via upregulating the expression levels of TJ-related proteins in a mouse model of DSS-induced colitis. Moreover, the results suggested that the effects of CB could be mediated by the Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Miao Liu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Key Laboratory of Hubei Province for Digestive System Disease, Wuhan, Hubei 430060, P.R. China.,Central Laboratory of Renmin Hospital, Wuhan, Hubei 430060, P.R. China
| | - Wenjie Xie
- Central Laboratory of Renmin Hospital, Wuhan, Hubei 430060, P.R. China.,Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xinyue Wan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Key Laboratory of Hubei Province for Digestive System Disease, Wuhan, Hubei 430060, P.R. China.,Central Laboratory of Renmin Hospital, Wuhan, Hubei 430060, P.R. China
| | - Tao Deng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Key Laboratory of Hubei Province for Digestive System Disease, Wuhan, Hubei 430060, P.R. China.,Central Laboratory of Renmin Hospital, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
39
|
Uc PY, Miranda J, Raya-Sandino A, Alarcón L, Roldán ML, Ocadiz-Delgado R, Cortés-Malagón EM, Chávez-Munguía B, Ramírez G, Asomoza R, Shoshani L, Gariglio P, González-Mariscal L. E7 oncoprotein from human papillomavirus 16 alters claudins expression and the sealing of epithelial tight junctions. Int J Oncol 2020; 57:905-924. [PMID: 32945372 PMCID: PMC7473757 DOI: 10.3892/ijo.2020.5105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 04/16/2020] [Indexed: 11/24/2022] Open
Abstract
Tight junctions (TJs) are cell-cell adhesion structures frequently altered by oncogenic transformation. In the present study the role of human papillomavirus (HPV) 16 E7 oncoprotein on the sealing of TJs was investigated and also the expression level of claudins in mouse cervix and in epithelial Madin-Darby Canine Kidney (MDCK) cells. It was found that there was reduced expression of claudins -1 and -10 in the cervix of 7-month-old transgenic K14E7 mice treated with 17β-estradiol (E2), with invasive cancer. In addition, there was also a transient increase in claudin-1 expression in the cervix of 2-month-old K14E7 mice, and claudin-10 accumulated at the border of cells in the upper layer of the cervix in FvB mice treated with E2, and in K14E7 mice treated with or without E2. These changes were accompanied by an augmented paracellular permeability of the cervix in 2- and 7-monthold FvB mice treated with E2, which became more pronounced in K14E7 mice treated with or without E2. In MDCK cells the stable expression of E7 increased the space between adjacent cells and altered the architecture of the monolayers, induced the development of an acute peak of transepithelial electrical resistance accompanied by a reduced expression of claudins -1, -2 and -10, and an increase in claudin-4. Moreover, E7 enhances the ability of MDCK cells to migrate through a 3D matrix and induces cell stiffening and stress fiber formation. These observations revealed that cell transformation induced by HPV16 E7 oncoprotein was accompanied by changes in the pattern of expression of claudins and the degree of sealing of epithelial TJs.
Collapse
Affiliation(s)
- Perla Yaceli Uc
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - Jael Miranda
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - Arturo Raya-Sandino
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - Lourdes Alarcón
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - María Luisa Roldán
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - Rodolfo Ocadiz-Delgado
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - Enoc Mariano Cortés-Malagón
- Research Unit on Genetics and Cancer, Research Division, Hospital Juárez de México, Mexico City 07760, Mexico
| | - Bibiana Chávez-Munguía
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - Georgina Ramírez
- Department of Electrical Engineering, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - René Asomoza
- Department of Electrical Engineering, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - Liora Shoshani
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - Patricio Gariglio
- Department of Genetics and Molecular Biology, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| | - Lorenza González-Mariscal
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies, Mexico City 07360, Mexico
| |
Collapse
|
40
|
Areco VA, Kohan R, Talamoni G, Tolosa de Talamoni NG, Peralta López ME. Intestinal Ca 2+ absorption revisited: A molecular and clinical approach. World J Gastroenterol 2020; 26:3344-3364. [PMID: 32655262 PMCID: PMC7327788 DOI: 10.3748/wjg.v26.i24.3344] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/11/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023] Open
Abstract
Ca2+ has an important role in the maintenance of the skeleton and is involved in the main physiological processes. Its homeostasis is controlled by the intestine, kidney, bone and parathyroid glands. The intestinal Ca2+ absorption occurs mainly via the paracellular and the transcellular pathways. The proteins involved in both ways are regulated by calcitriol and other hormones as well as dietary factors. Fibroblast growth factor 23 (FGF-23) is a strong antagonist of vitamin D action. Part of the intestinal Ca2+ movement seems to be vitamin D independent. Intestinal Ca2+ absorption changes according to different physiological conditions. It is promoted under high Ca2+ demands such as growth, pregnancy, lactation, dietary Ca2+ deficiency and high physical activity. In contrast, the intestinal Ca2+ transport decreases with aging. Oxidative stress inhibits the intestinal Ca2+ absorption whereas the antioxidants counteract the effects of prooxidants leading to the normalization of this physiological process. Several pathologies such as celiac disease, inflammatory bowel diseases, Turner syndrome and others occur with inhibition of intestinal Ca2+ absorption, some hypercalciurias show Ca2+ hyperabsorption, most of these alterations are related to the vitamin D endocrine system. Further research work should be accomplished in order not only to know more molecular details but also to detect possible therapeutic targets to ameliorate or avoid the consequences of altered intestinal Ca2+ absorption.
Collapse
Affiliation(s)
- Vanessa A Areco
- Laboratorio “Dr. Fernando Cañas”, Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba 5000, Argentina
| | - Romina Kohan
- Laboratorio “Dr. Fernando Cañas”, Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba 5000, Argentina
| | - Germán Talamoni
- Laboratorio “Dr. Fernando Cañas”, Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba 5000, Argentina
| | - Nori G Tolosa de Talamoni
- Laboratorio “Dr. Fernando Cañas”, Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba 5000, Argentina
| | - María E Peralta López
- Laboratorio “Dr. Fernando Cañas”, Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba 5000, Argentina
| |
Collapse
|
41
|
Singhal R, Shah YM. Oxygen battle in the gut: Hypoxia and hypoxia-inducible factors in metabolic and inflammatory responses in the intestine. J Biol Chem 2020; 295:10493-10505. [PMID: 32503843 DOI: 10.1074/jbc.rev120.011188] [Citation(s) in RCA: 201] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/04/2020] [Indexed: 12/13/2022] Open
Abstract
The gastrointestinal tract is a highly proliferative and regenerative tissue. The intestine also harbors a large and diverse microbial population collectively called the gut microbiome (microbiota). The microbiome-intestine cross-talk includes a dynamic exchange of gaseous signaling mediators generated by bacterial and intestinal metabolisms. Moreover, the microbiome initiates and maintains the hypoxic environment of the intestine that is critical for nutrient absorption, intestinal barrier function, and innate and adaptive immune responses in the mucosal cells of the intestine. The response to hypoxia is mediated by hypoxia-inducible factors (HIFs). In hypoxic conditions, the HIF activation regulates the expression of a cohort of genes that promote adaptation to hypoxia. Physiologically, HIF-dependent genes contribute to the aforementioned maintenance of epithelial barrier function, nutrient absorption, and immune regulation. However, chronic HIF activation exacerbates disease conditions, leading to intestinal injury, inflammation, and colorectal cancer. In this review, we aim to outline the major roles of physiological and pathological hypoxic conditions in the maintenance of intestinal homeostasis and in the onset and progression of disease with a major focus on understanding the complex pathophysiology of the intestine.
Collapse
Affiliation(s)
- Rashi Singhal
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA .,Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
42
|
Urban F, Hajek K, Naber T, Anczykowski B, Schäfer M, Wegener J. P ETER-assay: Combined Impedimetric Detection of Permeability (P E) and Resistance (TER) of Barrier-Forming Cell Layers. Sci Rep 2020; 10:7373. [PMID: 32355192 PMCID: PMC7192940 DOI: 10.1038/s41598-020-63624-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 12/05/2019] [Indexed: 11/09/2022] Open
Abstract
Epithelial and endothelial barrier function is typically studied in vitro by growing the cells of interest on permeable supports that are sandwiched between two fluid compartments. This setup mimics the physiological situation with the cell layer as the diffusion barrier at the interface between two chemically distinct fluids. Routinely, the barrier function is quantitatively described by two key parameters: (i) the transepithelial or transendothelial electrical resistance (TER) as a measure of the permeability for small inorganic ions and (ii) the permeability coefficient (PE) as a descriptor of the permeability for molecular tracers. So far the two parameters have been determined in separate experiments. This study introduces a device that allows for simultaneous detection of PE and TER of the very same cell monolayer in one single experiment (PETER-assay). The novel approach is entirely based on AC impedance measurements in two different modes, so that TER and PE become available in real time. The new approach is demonstrated for three epithelial cell lines derived from the kidney (MDCK-I, MDCK-II, NRK) with very different barrier properties under stationary conditions and when challenged by barrier-breaking fungal toxin cytochalasin D. PETER provides an excellent time-resolution and completely automated data collection.
Collapse
Affiliation(s)
- Florian Urban
- Universitaet Regensburg, Institut fuer Analytische Chemie, Chemo- & Biosensorik, Universitaetsstr. 31, 93053, Regensburg (G), Germany
| | - Kathrin Hajek
- Universitaet Regensburg, Institut fuer Analytische Chemie, Chemo- & Biosensorik, Universitaetsstr. 31, 93053, Regensburg (G), Germany
| | - Tobias Naber
- Universitaet Regensburg, Institut fuer Analytische Chemie, Chemo- & Biosensorik, Universitaetsstr. 31, 93053, Regensburg (G), Germany
| | | | - Marcus Schäfer
- nanoAnalytics GmbH, Heisenbergstr. 11, 48149, Münster (G), Germany
| | - Joachim Wegener
- Universitaet Regensburg, Institut fuer Analytische Chemie, Chemo- & Biosensorik, Universitaetsstr. 31, 93053, Regensburg (G), Germany. .,Fraunhofer Research Institution for Microsystems and Solid State Technologies EMFT, 80686, Muenchen (G), Germany.
| |
Collapse
|
43
|
Louer EM, Günzel D, Rosenthal R, Carmone C, Yi G, Stunnenberg HG, den Hollander AI, Deen PM. Differential day-night expression of tight junction components in murine retinal pigment epithelium. Exp Eye Res 2020; 193:107985. [DOI: 10.1016/j.exer.2020.107985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/30/2020] [Accepted: 02/20/2020] [Indexed: 02/06/2023]
|
44
|
Butkevych E, Lobo de Sá FD, Nattramilarasu PK, Bücker R. Contribution of Epithelial Apoptosis and Subepithelial Immune Responses in Campylobacter jejuni- Induced Barrier Disruption. Front Microbiol 2020; 11:344. [PMID: 32210941 PMCID: PMC7067706 DOI: 10.3389/fmicb.2020.00344] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/17/2020] [Indexed: 12/17/2022] Open
Abstract
Campylobacter jejuni is a widespread zoonotic pathogen and the leading bacterial cause of foodborne gastroenteritis in humans. Previous infection studies showed disruption of intercellular contacts, induction of epithelial apoptosis, and immune activation, all three contributing to intestinal barrier dysfunction leading to diarrhea. The present study aims to determine the impact of subepithelial immune cells on intestinal barrier dysfunction during Campylobacter jejuni infection and the underlying pathological mechanisms. Infection was performed in a co-culture of confluent monolayers of the human colon cell line HT-29/B6-GR/MR and THP-1 immune cells. Twenty-two hours after infection, transepithelial electrical resistance (TER) was decreased by 58 ± 6% compared to controls. The infection resulted in an increase in permeability for fluorescein (332 Da; 4.5-fold) and for FITC-dextran (4 kDa; 3.5-fold), respectively. In contrast, incubation of the co-culture with the pan-caspase inhibitor Q-VD-OPh during the infection resulted in a complete recovery of the decrease in TER and a normalization of flux values. Fluorescence microscopy showed apoptotic fragmentation in infected cell monolayers resulting in a 5-fold increase of the apoptotic ratio, accompanied by an increased caspase-3 cleavage and caspase-3/7 activity, which both were not present after Q-VD-OPh treatment. Western blot analysis revealed increased claudin-1 and claudin-2 protein expression. Inhibition of apoptosis induction did not normalize these tight junction changes. TNFα concentration was increased during the infection in the co-culture. In conclusion, Campylobacter jejuni infection and the consequent subepithelial immune activation cause intestinal barrier dysfunction mainly through caspase-3-dependent epithelial apoptosis. Concomitant tight junction changes were caspase-independent. Anti-apoptotic and immune-modulatory substances appear to be promising agents for treatment of campylobacteriosis.
Collapse
Affiliation(s)
- Eduard Butkevych
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Fábia Daniela Lobo de Sá
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Praveen Kumar Nattramilarasu
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Roland Bücker
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
45
|
Miyazono S, Otani T, Ogata K, Kitagawa N, Iida H, Inai Y, Matsuura T, Inai T. The reduced susceptibility of mouse keratinocytes to retinoic acid may be involved in the keratinization of oral and esophageal mucosal epithelium. Histochem Cell Biol 2020; 153:225-237. [PMID: 32006103 DOI: 10.1007/s00418-020-01845-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2020] [Indexed: 10/24/2022]
Abstract
Keratinocytes take up serum-derived retinol (vitamin A) and metabolize it to all-trans-retinoic acid (atRA), which binds to the nuclear retinoic acid receptor (RAR). We previously reported that serum-affected keratinocyte differentiation and function; namely, it inhibited keratinization, decreased loricrin (LOR) and claudin (CLDN) 1 expression, increased keratin (K) 4 and CLDN4 levels, and reduced paracellular permeability in three-dimensional (3D) cultures of mouse keratinocytes (COCA). Contrarily, RAR inhibition reversed these changes. Here, we aimed to examine whether atRA exerted the same effects as serum, and whether it was involved in the differential oral mucosa keratinization among animal species. Porcine oral mucosal keratinocytes, which form non-keratinized epithelium in vivo, established keratinized epithelium in 3D cultures. Both mouse and porcine sera induced non-keratinized epithelium at 0.1% in COCA 3D cultures. Although atRA caused the same changes as serum, its effective concentration differed. atRA inhibited keratinization at 0.1 nM and 1 nM in porcine or human keratinocytes and COCA, respectively. Furthermore, atRA upregulated CLDN7 in the cytoplasm but not in cell-cell contacts. These atRA-induced changes were reverted by RAR inhibition. The results indicate that serum-induced changes are probably due to the effect of serum-derived atRA, and that mouse keratinocytes require higher atRA concentrations to suppress keratinization than porcine and human keratinocytes. We propose that the lower susceptibility of mouse keratinocytes to atRA, rather than a lower retinol concentration, is a possible reason for the keratinization of mouse oral mucosal epithelium.
Collapse
Affiliation(s)
- Shoji Miyazono
- Department of Oral Rehabilitation, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka, 814-0193, Japan
| | - Takahito Otani
- Department of Morphological Biology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka, 814-0193, Japan
| | - Kayoko Ogata
- Department of Morphological Biology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka, 814-0193, Japan
| | - Norio Kitagawa
- Department of Morphological Biology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka, 814-0193, Japan
| | - Hiroshi Iida
- Laboratory of Zoology, Graduate School of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Yuko Inai
- Division of General Dentistry, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takashi Matsuura
- Department of Oral Rehabilitation, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka, 814-0193, Japan
| | - Tetsuichiro Inai
- Department of Morphological Biology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka, 814-0193, Japan.
| |
Collapse
|
46
|
Bhat AA, Syed N, Therachiyil L, Nisar S, Hashem S, Macha MA, Yadav SK, Krishnankutty R, Muralitharan S, Al-Naemi H, Bagga P, Reddy R, Dhawan P, Akobeng A, Uddin S, Frenneaux MP, El-Rifai W, Haris M. Claudin-1, A Double-Edged Sword in Cancer. Int J Mol Sci 2020; 21:ijms21020569. [PMID: 31952355 PMCID: PMC7013445 DOI: 10.3390/ijms21020569] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/12/2020] [Accepted: 01/13/2020] [Indexed: 12/11/2022] Open
Abstract
Claudins, a group of membrane proteins involved in the formation of tight junctions, are mainly found in endothelial or epithelial cells. These proteins have attracted much attention in recent years and have been implicated and studied in a multitude of diseases. Claudins not only regulate paracellular transepithelial/transendothelial transport but are also critical for cell growth and differentiation. Not only tissue-specific but the differential expression in malignant tumors is also the focus of claudin-related research. In addition to up- or down-regulation, claudin proteins also undergo delocalization, which plays a vital role in tumor invasion and aggressiveness. Claudin (CLDN)-1 is the most-studied claudin in cancers and to date, its role as either a tumor promoter or suppressor (or both) is not established. In some cancers, lower expression of CLDN-1 is shown to be associated with cancer progression and invasion, while in others, loss of CLDN-1 improves the patient survival. Another topic of discussion regarding the significance of CLDN-1 is its localization (nuclear or cytoplasmic vs perijunctional) in diseased states. This article reviews the evidence regarding CLDN-1 in cancers either as a tumor promoter or suppressor from the literature and we also review the literature regarding the pattern of CLDN-1 distribution in different cancers, focusing on whether this localization is associated with tumor aggressiveness. Furthermore, we utilized expression data from The Cancer Genome Atlas (TCGA) to investigate the association between CLDN-1 expression and overall survival (OS) in different cancer types. We also used TCGA data to compare CLDN-1 expression in normal and tumor tissues. Additionally, a pathway interaction analysis was performed to investigate the interaction of CLDN-1 with other proteins and as a future therapeutic target.
Collapse
Affiliation(s)
- Ajaz A. Bhat
- Division of Translational Medicine, Research Branch, Sidra Medicine, Doha 26999, Qatar; (A.A.B.); (N.S.); (S.N.); (S.H.); (S.K.Y.)
| | - Najeeb Syed
- Division of Translational Medicine, Research Branch, Sidra Medicine, Doha 26999, Qatar; (A.A.B.); (N.S.); (S.N.); (S.H.); (S.K.Y.)
| | - Lubna Therachiyil
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (L.T.); (R.K.); (S.U.)
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | - Sabah Nisar
- Division of Translational Medicine, Research Branch, Sidra Medicine, Doha 26999, Qatar; (A.A.B.); (N.S.); (S.N.); (S.H.); (S.K.Y.)
| | - Sheema Hashem
- Division of Translational Medicine, Research Branch, Sidra Medicine, Doha 26999, Qatar; (A.A.B.); (N.S.); (S.N.); (S.H.); (S.K.Y.)
| | - Muzafar A. Macha
- Department of Biotechnology, Central University of Kashmir, Ganderbal, Jammu and Kashmir 191201, India;
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Santosh K. Yadav
- Division of Translational Medicine, Research Branch, Sidra Medicine, Doha 26999, Qatar; (A.A.B.); (N.S.); (S.N.); (S.H.); (S.K.Y.)
| | - Roopesh Krishnankutty
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (L.T.); (R.K.); (S.U.)
| | | | - Hamda Al-Naemi
- Laboratory Animal Research Center, Qatar University, Doha 2713, Qatar; (S.M.); (H.A.-N.)
| | - Puneet Bagga
- Center for Magnetic Resonance and Optical Imaging, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (P.B.); (R.R.)
| | - Ravinder Reddy
- Center for Magnetic Resonance and Optical Imaging, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; (P.B.); (R.R.)
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Anthony Akobeng
- Department of Pediatric Gastroenterology, Sidra Medicine, Doha 26999, Qatar;
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (L.T.); (R.K.); (S.U.)
| | | | - Wael El-Rifai
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Mohammad Haris
- Division of Translational Medicine, Research Branch, Sidra Medicine, Doha 26999, Qatar; (A.A.B.); (N.S.); (S.N.); (S.H.); (S.K.Y.)
- Laboratory Animal Research Center, Qatar University, Doha 2713, Qatar; (S.M.); (H.A.-N.)
- Correspondence: ; Tel.: +974-4003-7407
| |
Collapse
|
47
|
Retinoic acid signalling adjusts tight junction permeability in response to air-liquid interface conditions. Cell Signal 2020; 65:109421. [DOI: 10.1016/j.cellsig.2019.109421] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/14/2019] [Accepted: 09/15/2019] [Indexed: 12/12/2022]
|
48
|
Effects of Claudin-1 on the Action of Clostridium perfringens Enterotoxin in Caco-2 Cells. Toxins (Basel) 2019; 11:toxins11100582. [PMID: 31601044 PMCID: PMC6832201 DOI: 10.3390/toxins11100582] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 12/11/2022] Open
Abstract
Clostridium perfringens enterotoxin (CPE) contributes to diarrhea and an often-lethal enterotoxemia. CPE action starts when it binds to claudin receptors, forming a small complex (90 kDa). Six small complexes then oligomerize to create prepores, followed by insertion of beta-hairpins from CPE to form beta-barrel pores named CH-1 or CH-2. Of the ~27 members of the human claudin protein family, only some bind CPE. However, both receptor claudins and the nonreceptor claudin-1 (CLDN-1) are associated with the small and CH-1/CH-2 CPE complexes. Therefore, this study evaluated whether claudin-1 affects CPE action by generating a CLDN-1 null mutant in Caco-2 cells using CRISPR-Cas9. Compared to wild-type Caco-2 cells, paracellular permeability of the CLDN-1 mutant was significantly enhanced, suggesting that claudin-1 may reduce CPE absorption during enterotoxemia. The CLDN-1 mutant was also markedly more sensitive than wild-type Caco-2 cells to apically-applied CPE. The mechanism behind this increased sensitivity involved higher CPE binding by the CLDN-1 mutant vs. wild-type Caco-2 cells, which led to more CH-1/CH-2 complex formation. However, the CH-1/CH-2 complexes formed by the CLDN-1 mutant were less stable or trypsin resistant than those of wild-type cells. These results indicate that, although a nonreceptor, CLDN-1 positively and negatively influences CPE action.
Collapse
|
49
|
Sivanantham A, Pattarayan D, Rajasekar N, Kannan A, Loganathan L, Bethunaickan R, Mahapatra SK, Palanichamy R, Muthusamy K, Rajasekaran S. Tannic acid prevents macrophage-induced pro-fibrotic response in lung epithelial cells via suppressing TLR4-mediated macrophage polarization. Inflamm Res 2019; 68:1011-1024. [DOI: 10.1007/s00011-019-01282-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/27/2019] [Accepted: 08/29/2019] [Indexed: 02/08/2023] Open
|
50
|
Wang Y, Sun L, Chen S, Guo S, Yue T, Hou Q, Feng M, Xu H, Liu Y, Wang P, Pan Y. The administration of Escherichia coli Nissle 1917 ameliorates irinotecan-induced intestinal barrier dysfunction and gut microbial dysbiosis in mice. Life Sci 2019; 231:116529. [PMID: 31173781 DOI: 10.1016/j.lfs.2019.06.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/28/2019] [Accepted: 06/02/2019] [Indexed: 02/08/2023]
Abstract
AIMS The present study investigated the effect of Escherichia coli Nissle 1917 (EcN) on irinotecan-induced intestinal barrier dysfunction and gut microbial dysbiosis in a mouse model and in the human colonic cells lines Caco-2. MATERIALS AND METHODS Male BALB/c mice received irinotecan intraperitoneal injection with or without EcN administration intragastrically. Body weight, diarrhea severity, intestinal permeability and histopathological analysis of ileum epithelia of mice from different groups were assessed. The expression and localization of tight junction proteins were examined using western blot and immunofluorescence. Gut microbiota structure and diversity were measured with 16 S rRNA sequencing. Caco-2 monolayers were incubated with EcN culture supernatant (EcNsup) or SN-38 and the monolayer barrier function was assessed by transepithelial electrical resistance (TER) and FITC-dextran 4000 Da (FD-4) flux. KEY FINDINGS Pretreatment with EcN significantly attenuated irinotecan-induced weight loss and diarrhea in mice. In addition, EcN inhibited the increased intestinal permeability and decreased Claudin-1 expression in irinotecan-treated mice. Furthermore, irinotecan treatment decreased the diversity of gut microbiota and increased the relative abundance of Proteobacteria compared to control group. EcN administration ameliorated the gut microbiota dysbiosis. In Caco-2 monolayers, EcNsup ameliorated the decreased TER and increased FD-4 flux elicited by SN-38. Moreover, EcNsup attenuated SN-38-induced altered localization and distribution of Claudin-1 in Caco-2 monolayers. SIGNIFICANCE Our results indicated that the administration of EcN protected against irinotecan-induced intestinal injury by regulating intestinal barrier function and gut microbiota.
Collapse
Affiliation(s)
- Yurong Wang
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Lie Sun
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Shanwen Chen
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Shihao Guo
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Taohua Yue
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Qisheng Hou
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Mei Feng
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Hao Xu
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Yucun Liu
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Pengyuan Wang
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Yisheng Pan
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China.
| |
Collapse
|