1
|
Rehmani T, Dias AP, Applin BD, Salih M, Tuana BS. SLMAP3 is essential for neurulation through mechanisms involving cytoskeletal elements, ABP, and PCP. Life Sci Alliance 2024; 7:e202302545. [PMID: 39366759 PMCID: PMC11452652 DOI: 10.26508/lsa.202302545] [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: 12/19/2023] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024] Open
Abstract
SLMAP3 is a tail-anchored membrane protein that targets subcellular organelles and is believed to regulate Hippo signaling. The global loss of SLMAP3 causes late embryonic lethality in mice, with some embryos exhibiting neural tube defects such as craniorachischisis. We show here that SLMAP3 -/- embryos display reduced length and increased width of neural plates, signifying arrested convergent extension. The expression of planar cell polarity (PCP) components Dvl2/3 and the activity of the downstream targets ROCK2, cofilin, and JNK1/2 were dysregulated in SLMAP3 -/- E12.5 brains. Furthermore, the cytoskeletal proteins (γ-tubulin, actin, and nestin) and apical components (PKCζ and ZO-1) were mislocalized in neural tubes of SLMAP3 -/- embryos, with a subsequent decrease in colocalization of PCP proteins (Fzd6 and pDvl2). However, no changes in PCP or cytoskeleton proteins were found in cultured neuroepithelial cells depleted of SLMAP3, suggesting an essential requirement for SLMAP3 for these processes in vivo for neurulation. The loss of SLMAP3 had no impact on Hippo signaling in SLMAP3 -/- embryos, brains, and neural tubes. Proteomic analysis revealed SLMAP3 in an interactome with cytoskeletal components, including nestin, tropomyosin 4, intermediate filaments, plectin, the PCP protein SCRIB, and STRIPAK members in embryonic brains. These results reveal a crucial role of SLMAP3 in neural tube development by regulating the cytoskeleton organization and PCP pathway.
Collapse
Affiliation(s)
- Taha Rehmani
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Ana Paula Dias
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Billi Dawn Applin
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Maysoon Salih
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Balwant S Tuana
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| |
Collapse
|
2
|
Seitz M, Song Y, Lian XL, Ma Z, Jain E. Soft Polyethylene Glycol Hydrogels Support Human PSC Pluripotency and Morphogenesis. ACS Biomater Sci Eng 2024; 10:4525-4540. [PMID: 38973308 PMCID: PMC11234337 DOI: 10.1021/acsbiomaterials.4c00923] [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: 05/20/2024] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 07/09/2024]
Abstract
Lumenogenesis within the epiblast represents a critical step in early human development, priming the embryo for future specification and patterning events. However, little is known about the specific mechanisms that drive this process due to the inability to study the early embryo in vivo. While human pluripotent stem cell (hPSC)-based models recapitulate many aspects of the human epiblast, most approaches for generating these 3D structures rely on ill-defined, reconstituted basement membrane matrices. Here, we designed synthetic, nonadhesive polyethylene glycol (PEG) hydrogel matrices to better understand the role of matrix mechanical cues in iPSC morphogenesis, specifically elastic modulus. First, we identified a narrow range of hydrogel moduli that were conducive to the hPSC viability, pluripotency, and differentiation. We then used this platform to investigate the effects of the hydrogel modulus on lumenogenesis, finding that matrices of intermediate stiffness yielded the most epiblast-like aggregates. Conversely, stiffer matrices impeded lumen formation and apico-basal polarization, while the softest matrices yielded polarized but aberrant structures. Our approach offers a simple, modular platform for modeling the human epiblast and investigating the role of matrix cues in its morphogenesis.
Collapse
Affiliation(s)
- Michael
P. Seitz
- Department
of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Bioinspired
Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Yuanhui Song
- Department
of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Bioinspired
Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Xiaojun Lance Lian
- Department
of Biomedical Engineering, The Huck Institutes of the Life Sciences,
Department of Biology, Pennsylvania State
University, University
Park, Pennsylvania 16802, United States
| | - Zhen Ma
- Department
of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Bioinspired
Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Era Jain
- Department
of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Bioinspired
Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| |
Collapse
|
3
|
Tang YW, Jiang MY, Cao JW, Wan F. Triptolide decreases podocytes permeability by regulating TET2-mediated hydroxymethylation of ZO-1. Exp Biol Med (Maywood) 2024; 249:10051. [PMID: 38881848 PMCID: PMC11176508 DOI: 10.3389/ebm.2024.10051] [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: 11/13/2023] [Accepted: 05/07/2024] [Indexed: 06/18/2024] Open
Abstract
Podocyte injury or dysfunction can lead to proteinuria and glomerulosclerosis. Zonula occludens 1 (ZO-1) is a tight junction protein which connects slit diaphragm (SD) proteins to the actin cytoskeleton. Previous studies have shown that the expression of ZO-1 is decreased in chronic kidney disease (CKD). Thus, elucidation of the regulation mechanism of ZO-1 has considerable clinical importance. Triptolide (TP) has been reported to exert a strong antiproteinuric effect by inhibiting podocyte epithelial mesenchymal transition (EMT) and inflammatory response. However, the underlying mechanisms are still unclear. We found that TP upregulates ZO-1 expression and increases the fluorescence intensity of ZO-1 in a puromycin aminonucleoside (PAN)-induced podocyte injury model. Permeablity assay showed TP decreases podocyte permeability in PAN-treated podocyte. TP also upregulates the DNA demethylase TET2. Our results showed that treatment with the DNA methyltransferase inhibitors 5-azacytidine (5-AzaC) and RG108 significantly increased ZO-1 expression in PAN-treated podocytes. Methylated DNA immunoprecipitation (MeDIP) and hydroxymethylated DNA immunoprecipitation (hMeDIP) results showed that TP regulates the methylation status of the ZO-1 promoter. Knockdown of TET2 decreased ZO-1 expression and increased methylation of its promoter, resulting in the increase of podocyte permeability. Altogether, these results indicate that TP upregulates the expression of ZO-1 and decreases podocyte permeability through TET2-mediated 5 mC demethylation. These findings suggest that TP may alleviate podocyte permeability through TET2-mediated hydroxymethylation of ZO-1.
Collapse
Affiliation(s)
- Yue-Wen Tang
- Department of Nephrology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
| | - Meng-Ya Jiang
- Department of Nephrology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Jia-Wei Cao
- Department of Nephrology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Feng Wan
- Department of Nephrology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, China
| |
Collapse
|
4
|
Groh AC, Möller-Kerutt A, Gilhaus K, Höffken V, Nedvetsky P, Kleimann S, Behrens M, Ghosh S, Hansen U, Krahn MP, Ebnet K, Pavenstädt H, Ludwig A, Weide T. PALS1 is a key regulator of the lateral distribution of tight junction proteins in renal epithelial cells. J Cell Sci 2024; 137:jcs261303. [PMID: 38265145 DOI: 10.1242/jcs.261303] [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: 05/03/2023] [Accepted: 12/04/2023] [Indexed: 01/25/2024] Open
Abstract
The evolutionarily conserved apical Crumbs (CRB) complex, consisting of the core components CRB3a (an isoform of CRB3), PALS1 and PATJ, plays a key role in epithelial cell-cell contact formation and cell polarization. Recently, we observed that deletion of one Pals1 allele in mice results in functional haploinsufficiency characterized by renal cysts. Here, to address the role of PALS1 at the cellular level, we generated CRISPR/Cas9-mediated PALS1-knockout MDCKII cell lines. The loss of PALS1 resulted in increased paracellular permeability, indicating an epithelial barrier defect. This defect was associated with a redistribution of several tight junction-associated proteins from bicellular to tricellular contacts. PALS1-dependent localization of tight junction proteins at bicellular junctions required its interaction with PATJ. Importantly, reestablishment of the tight junction belt upon transient F-actin depolymerization or upon Ca2+ removal was strongly delayed in PALS1-deficient cells. Additionally, the cytoskeleton regulator RhoA was redistributed from junctions into the cytosol under PALS1 knockout. Together, our data uncover a critical role of PALS1 in the coupling of tight junction proteins to the F-actin cytoskeleton, which ensures their correct distribution along bicellular junctions and the formation of tight epithelial barrier.
Collapse
Affiliation(s)
- Ann-Christin Groh
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Department Molecular Nephrology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| | - Annika Möller-Kerutt
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Department Molecular Nephrology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| | - Kevin Gilhaus
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Department Molecular Nephrology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| | - Verena Höffken
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Department Molecular Nephrology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| | - Pavel Nedvetsky
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Medical Cell Biology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| | - Simon Kleimann
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Department Molecular Nephrology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| | - Malina Behrens
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Department Molecular Nephrology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| | - Sujasha Ghosh
- School of Biological Sciences and NTU Institute of Structural Biology (NISB), Nanyang Technological University, 60 Nanyang Drive, 637551 Singapore City, Singapore
| | - Uwe Hansen
- University Hospital of Münster, Institute of Musculoskeletal Medicine (IMM), Head Core Facility Electron Microscopy, Domagkstraße 3, 48149 Münster, Germany
| | - Michael P Krahn
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Medical Cell Biology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| | - Klaus Ebnet
- Institute-associated Research Group "Cell adhesion and cell polarity", Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Von-Esmarch-Straße 56, 48149 Münster, Germany
| | - Hermann Pavenstädt
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Department Molecular Nephrology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| | - Alexander Ludwig
- School of Biological Sciences and NTU Institute of Structural Biology (NISB), Nanyang Technological University, 60 Nanyang Drive, 637551 Singapore City, Singapore
| | - Thomas Weide
- University Hospital of Münster (UKM), Internal Medicine D (MedD), Department Molecular Nephrology, Albert-Schweitzer-Campus 1 Building A14, 48149 Münster, Germany
| |
Collapse
|
5
|
Smith FM, Kosman DJ. Loss of filamentous actin, tight junction protein expression, and paracellular barrier integrity in frataxin-deficient human brain microvascular endothelial cells-implications for blood-brain barrier physiology in Friedreich's ataxia. Front Mol Biosci 2024; 10:1299201. [PMID: 38274097 PMCID: PMC10808331 DOI: 10.3389/fmolb.2023.1299201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/22/2023] [Indexed: 01/27/2024] Open
Abstract
Introduction: Friedreich's Ataxia (FRDA) is the most prevalent inherited ataxia. FRDA results from loss of Frataxin (FXN), an essential mitochondrial iron trafficking protein. FRDA starts with an early burst of neurodegeneration of the dorsal root ganglion and cerebellar dentate nuclei, followed by progressive brain iron accumulation in the latter. End stage disease includes cardiac fibrosis that contributes to hypertrophic cardiomyopathy. The microvasculature plays an essential barrier role in both brain and heart homeostasis, thus an investigation of this tissue system in FRDA is essential to the delineation of the cellular dysfunction in this genetic disorder. Previous reports have identified cytoskeletal alterations in non-barrier forming FRDA cell models, but physiological consequences are limited. Methods: We investigated brain microvascular endothelial cell integrity in FRDA in a model of the blood-brain barrier (BBB). We have knocked down FXN in immortalized human brain microvascular endothelial cells (hBMVEC), which compose the microcapillaries of the BBB, by using shRNA. We confirmed known cellular pathophysiologies of FXN-knockdown including decreased energy metabolism, markers of oxidative stress, and increased cell size. Results: We investigated cytoskeletal architecture, identifying decreased filamentous actin and Occludin and Claudin-5 tight junction protein expression in shFXN hBMVECs. This was consistent with decreased transendothelial electrical resistance (TEER) and increased paracellular tracer flux during early barrier formation. shFXN hBMVEC start with only 67% barrier integrity of the controls, and flux a paracellular tracer at 800% of physiological levels. Discussion: We identified that insufficient FXN levels in the hBMVEC BBB model causes changes in cytoskeletal architecture and tight junction protein abundance, co-incident with increased barrier permeability. Changes in the integrity of the BBB may be related to patient brain iron accumulation, neuroinflammation, neurodegeneration, and stroke. Furthermore, our findings implicate other barrier cells, e.g., the cardiac microvasculature, loci of disease pathology in FRDA.
Collapse
Affiliation(s)
- Frances M. Smith
- Jacobs School of Medicine and Biomedical Sciences, Department of Biochemistry, The State University of New York at Buffalo, Buffalo, NY, United States
| | | |
Collapse
|
6
|
Park A, Choi S, Do J, Kim Y, Kim KS, Koh E, Park KS. ZO-1 regulates the migration of mesenchymal stem cells in cooperation with α-catenin in response to breast tumor cells. Cell Death Discov 2024; 10:19. [PMID: 38212369 PMCID: PMC10784548 DOI: 10.1038/s41420-023-01793-4] [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: 10/16/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/13/2024] Open
Abstract
Mesenchymal stem cells are recruited from the bone marrow into breast tumors, contributing to the creation of a tumor microenvironment that fosters tropism for breast tumors. However, the intrinsic mechanisms underlying the recruitment of bone marrow-derived mesenchymal stem cells (MSCs) into the breast tumor microenvironment are still under investigation. Our discoveries identified zonula occludens-1 (ZO-1) as a specific intrinsic molecule that plays a vital role in mediating the collective migration of MSCs towards breast tumor cells and transforming growth factor beta (TGF-β), which is a crucial factor secreted by breast tumor cells. Upon migration in response to MDA-MB-231 cells and TGF-β, MSCs showed increased formation of adherens junction-like structures (AJs) expressing N-cadherin and α-catenin at their cell-cell contacts. ZO-1 was found to be recruited into the AJs at the cell-cell contacts between MSCs. Additionally, ZO-1 collaborated with α-catenin to regulate AJ formation, dependently on the SH3 and GUK domains of the ZO-1 protein. ZO-1 knockdown led to the impaired migration of MSCs in response to the stimuli and subsequent downregulation of AJs formation at the cell-cell contacts during MSCs migration. Overall, our study highlights the novel role of ZO-1 in guiding MSC migration towards breast tumor cells, suggesting its potential as a new strategy for controlling and re-engineering the breast tumor microenvironment.
Collapse
Affiliation(s)
- Aran Park
- Graduate School of Biotechnology, Kyung Hee University, Yongin, 17104, Korea
| | - Sanghyuk Choi
- Graduate School of Biotechnology, Kyung Hee University, Yongin, 17104, Korea
| | - Jungbeom Do
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, 02447, Korea
| | - Youngjae Kim
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, 02447, Korea
| | - Kyung-Sup Kim
- Department of Biochemistry and Molecular Biology, Institute of Genetic Science, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Eunjin Koh
- Department of Biochemistry and Molecular Biology, Institute of Genetic Science, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Ki-Sook Park
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, 02447, Korea.
- East-West Medical Research Institute, Kyung Hee University, Seoul, 02447, Korea.
| |
Collapse
|
7
|
Rungrasameviriya P, Santilinon A, Atichartsintop P, Hadpech S, Thongboonkerd V. Tight junction and kidney stone disease. Tissue Barriers 2024; 12:2210051. [PMID: 37162265 PMCID: PMC10832927 DOI: 10.1080/21688370.2023.2210051] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/30/2023] [Indexed: 05/11/2023] Open
Abstract
Defects of tight junction (TJ) are involved in many diseases related to epithelial cell functions, including kidney stone disease (KSD), which is a common disease affecting humans for over a thousand years. This review provides brief overviews of KSD and TJ, and summarizes the knowledge on crystal-induced defects of TJ in renal tubular epithelial cells (RTECs) in KSD. Calcium oxalate (CaOx) crystals, particularly COM, disrupt TJ via p38 MAPK and ROS/Akt/p38 MAPK signaling pathways, filamentous actin (F-actin) reorganization and α-tubulin relocalization. Stabilizing p38 MAPK signaling, reactive oxygen species (ROS) production, F-actin and α-tubulin by using SB239063, N-acetyl-L-cysteine (NAC), phalloidin and docetaxel, respectively, successfully prevent the COM-induced TJ disruption and malfunction. Additionally, genetic disorders of renal TJ, including mutations and single nucleotide polymorphisms (SNPs) of CLDN2, CLDN10b, CLDN14, CLDN16 and CLDN19, also affect KSD. Finally, the role of TJ as a potential target for KSD therapeutics and prevention is also discussed.
Collapse
Affiliation(s)
- Papart Rungrasameviriya
- Nawamethee Project, Doctor of Medicine Program, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Aticha Santilinon
- Nawamethee Project, Doctor of Medicine Program, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Palita Atichartsintop
- Nawamethee Project, Doctor of Medicine Program, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sudarat Hadpech
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
8
|
Min S, Tao W, Miao Y, Li Y, Wu T, He X, Zhang Y, Liu B, Meng Z, Han K, Liu S, Li L, Chen J, Zhao S, Zhang J, Zhang X. Dual Delivery of Tetramethylpyrazine and miR-194-5p Using Soft Mesoporous Organosilica Nanoparticles for Acute Lung Injury Therapy. Int J Nanomedicine 2023; 18:6469-6486. [PMID: 38026537 PMCID: PMC10640848 DOI: 10.2147/ijn.s420802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
Background The respiratory system is intensely damaged by acute lung injury (ALI). The anti-inflammatory effects of tetramethylpyrazine (TMP) against ALI have been confirmed, but it exhibits a short half-life. miR-194-5p could directly target Rac1, but the internalization rate of miRNA cells was low. Purpose To explore the potential of the soft mesoporous organic silica nanoplatform (NPs) as carriers for delivery of TMP and miR-194-5p through the tail vein. Methods NPs@TMP and NPs@PEI@miR-194-5p were added to the HUVEC cell-lines, in vitro, to observe the cell uptake and cytotoxic effects. In vivo experiments were conducted by injecting fluorescently labeled NPs through the tail vein and tracking distribution. Therapeutic and toxic side-effects were analyzed systemically. Results In vitro study exhibited that NPs have no toxic effect on HUVECs within the experimental parameters and have excellent cellular uptake. The IVIS Spectrum Imaging System shows that NPs accumulate mainly in the lungs. NPs@TMP treatment can improved oxidative stress and inflammation levels in ALI mice and inhibited the TLR4/NLRP3/caspase 1 pathway. NPs@PEI@miR-194-5p can inhibit the Rac1/ZO-1/occludin pathway and improved endothelial cell permeability in ALI mice. The co-treatment of NPs@TMP and NPs@PEI@miR-194-5p can significantly improved the survival rates of the mice, reduced pulmonary capillary permeability and improved pathological injury in ALI mice. Innovation This study combined traditional Chinese medicine, bioinformatics, cellular molecular biology and nanobiomedicine to study the pathogenesis and treatment of ALI. The rate of cellular internalization was improved by changing the shape and hardness of nanoparticles. NPs@TMP and NPs@PEI@miR-194-5p combined application can significantly improve the survival condition and pathological injury of mice. Conclusion NPs loaded with TMP and miR-194-5p showed a greater therapeutic effect in ALI mice.
Collapse
Affiliation(s)
- Simin Min
- Bengbu Medical College Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, People’s Republic of China
- Suzhou Hospital of Anhui Medical University, Suzhou, Anhui, 234000, People’s Republic of China
| | - Weiting Tao
- Bengbu Medical College Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, People’s Republic of China
| | - Yuchen Miao
- Department of Chemistry, Bengbu Medical College, Bengbu, Anhui, 233030, People’s Republic of China
| | - Yan Li
- School of Medicine and Health Engineering, Changzhou University, Changzhou, Jiangsu, 213164, People’s Republic of China
| | - Tianyu Wu
- School of Public Health, Bengbu Medical College, Bengbu, Anhui, 233030, People’s Republic of China
| | - Xiaoyu He
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233030, People’s Republic of China
| | - Yijing Zhang
- School of Clinical Medicine, Bengbu Medical College, Bengbu, Anhui, 233030, People’s Republic of China
| | - Bangye Liu
- School of Clinical Medicine, Bengbu Medical College, Bengbu, Anhui, 233030, People’s Republic of China
| | - Zixin Meng
- School of Clinical Medicine, Bengbu Medical College, Bengbu, Anhui, 233030, People’s Republic of China
| | - Ke Han
- Bengbu Medical College Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, People’s Republic of China
| | - Saisai Liu
- Bengbu Medical College Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, People’s Republic of China
| | - Li Li
- Bengbu Medical College Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, People’s Republic of China
| | - Jie Chen
- Bengbu Medical College Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, People’s Republic of China
| | - Shidi Zhao
- Bengbu Medical College Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, People’s Republic of China
| | - Junjie Zhang
- Department of Chemistry, Bengbu Medical College, Bengbu, Anhui, 233030, People’s Republic of China
| | - Xiaonan Zhang
- Bengbu Medical College Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Bengbu Medical College, Bengbu, Anhui, 233030, People’s Republic of China
| |
Collapse
|
9
|
Song M, Zhang Z, Li Y, Xiang Y, Li C. Midgut microbiota affects the intestinal barrier by producing short-chain fatty acids in Apostichopus japonicus. Front Microbiol 2023; 14:1263731. [PMID: 37915855 PMCID: PMC10616862 DOI: 10.3389/fmicb.2023.1263731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/15/2023] [Indexed: 11/03/2023] Open
Abstract
Introduction The intestinal microbiota participates in host physiology and pathology through metabolites, in which short-chain fatty acids (SCFAs) are considered principal products and have extensive influence on intestine homeostasis. It has been reported that skin ulceration syndrome (SUS), the disease of Apostichopus japonicus caused by Vibrio splendidus, is associated with the alteration of the intestinal microbiota composition. Method To investigate whether the intestinal microbiota affects A. japonicus health via SCFAs, in this study, we focus on the SCFA profiling and intestinal barrier function in A. japonicus treated with V. splendidus. Results and discussion We found that V. splendidus could destroy the mid-intestine integrity and downregulate the expression of tight junction proteins ZO-1 and occludin in A. japonicus, which further dramatically decreased microorganism abundance and altered SCFAs contents. Specifically, acetic acid is associated with the largest number of microorganisms and has a significant correlation with occludin and ZO-1 among the seven SCFAs. Furthermore, our findings showed that acetic acid could maintain the intestinal barrier function by increasing the expression of tight junction proteins and rearranging the tight junction structure by regulating F-actin in mid-intestine epithelial cells. Thus, our results provide insights into the effects of the gut microbiome and SCFAs on intestine barrier homeostasis and provide essential knowledge for intervening in SUS by targeting metabolites or the gut microbiota.
Collapse
Affiliation(s)
- Mingshan Song
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China
| | - Zhen Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China
| | - Yanan Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China
| | - Yangxi Xiang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China
| | - Chenghua Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
10
|
Sutton K, Nash T, Sives S, Borowska D, Mitchell J, Vohra P, Stevens MP, Vervelde L. Disentangling the innate immune responses of intestinal epithelial cells and lamina propria cells to Salmonella Typhimurium infection in chickens. Front Microbiol 2023; 14:1258796. [PMID: 37854334 PMCID: PMC10579587 DOI: 10.3389/fmicb.2023.1258796] [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: 07/14/2023] [Accepted: 09/19/2023] [Indexed: 10/20/2023] Open
Abstract
Salmonella enterica serovar Typhimurium (STm) is a major foodborne pathogen and poultry are a key reservoir of human infections. To understand the host responses to early stages of Salmonella infection in poultry, we infected 2D and 3D enteroids, the latter of which contains leukocytes, neurons, and mesenchymal cells that are characteristic of the lamina propria. We infected these enteroids with wild-type (WT STm), a non-invasive mutant lacking the prgH gene (ΔprgH STm), or treated them with STm lipopolysaccharide (LPS) and analyzed the expression of innate immune related genes by qPCR at 4 and 8 h. The localization of the tight junction protein, ZO-1, expression was disrupted in WT STm infected enteroids but not ΔprgH STm or LPS treated enteroids, suggesting a loss of epithelial barrier integrity. The innate immune response to LPS was more pronounced in 2D enteroids compared to 3D enteroids and by 8 hpi, the response in 3D enteroids was almost negligible. However, when STm adhered to or invaded the enteroids, both 2D and 3D enteroids exhibited an upregulation of inflammatory responses. The presence of lamina propria cells in 3D enteroids resulted in the unique expression of genes associated with immune functions involved in regulating inflammation. Moreover, 2D and 3D enteroids showed temporal differences in response to bacterial invasion or adherence. At 8 hpi, innate responses in 3D but not 2D enteroids continued to increase after infection with WT STm, whereas the responses to the non-invasive strain decreased at 8 hpi in both 2D and 3D enteroids. In conclusion, STm infection of chicken enteroids recapitulated several observations from in vivo studies of Salmonella-infected chickens, including altered epithelial barrier integrity based on ZO-1 expression and inflammatory responses. Our findings provide evidence that Salmonella-infected enteroids serve as effective models for investigating host-pathogen interactions and exploring the molecular mechanisms of microbial virulence although the 3D model mimics the host more accurately due to the presence of a lamina propria.
Collapse
Affiliation(s)
- Kate Sutton
- Division of Immunology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Tessa Nash
- Division of Immunology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Samantha Sives
- Division of Immunology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Dominika Borowska
- Division of Immunology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Jordan Mitchell
- Division of Immunology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Prerna Vohra
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Mark P. Stevens
- Division of Bacteriology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Lonneke Vervelde
- Division of Immunology, The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
11
|
Ouyang H, Li W, Hansen SH. p120 RasGAP and ZO-2 are essential for Hippo signaling and tumor suppressor function mediated by p190A RhoGAP. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.541483. [PMID: 37292741 PMCID: PMC10245842 DOI: 10.1101/2023.05.22.541483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
ARHGAP35 , which encodes p190A RhoGAP (p190A), is a major cancer gene. p190A is a tumor suppressor that activates the Hippo pathway. p190A was originally cloned via direct binding to p120 RasGAP (RasGAP). Here, we determine that a novel interaction of p190A with the tight junction-associated protein ZO-2 is dependent on RasGAP. We establish that both RasGAP and ZO-2 are necessary for p190A to activate LATS kinases, elicit mesenchymal-to-epithelial transition, promote contact inhibition of cell proliferation and suppress tumorigenesis. Moreover, RasGAP and ZO-2 are required for transcriptional modulation by p190A. Finally, we demonstrate that low ARHGAP35 expression is associated with shorter survival in patients with high, but not low, transcript levels of TJP2 encoding ZO-2. Hence, we define a tumor suppressor interactome of p190A that includes ZO-2, an established constituent of the Hippo pathway, and RasGAP, which despite strong association with Ras signaling, is essential for p190A to activate LATS kinases.
Collapse
|
12
|
Qin S, Peng Y, She F, Zhang J, Li L, Chen F. Positive effects of selenized-oligochitosan on zearalenone-induced intestinal dysfunction in piglets. Front Vet Sci 2023; 10:1184969. [PMID: 37261113 PMCID: PMC10228365 DOI: 10.3389/fvets.2023.1184969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/28/2023] [Indexed: 06/02/2023] Open
Abstract
This paper assessed the positive effects of selenized-oligochitosan (SOC) on zearalenone(ZEN)-induced intestinal dysfunction in piglets. Sixty piglets were randomly divided into 4 groups. Group C was fed the basal diet as a control and Group Z was supplemented with 2 μg/g ZEN in the basal diet; Group ZS1 and ZS2 were supplemented with 0.3 or 0.5 μg/g SOC (calculated by selenium), in addition to 2 μg/g ZEN in the basal diet. After 42 days, ileal mucosal structure, digestive enzyme activities, tight junction protein mRNA expressions, plasma D-lactate and D-xylose contents, and plasma diamine oxidase activities were determined. Compare with Group C, ileal villus height, value of villus height/crypt depth, trypsin, lipase and α-amylase activities, occluding, claudin-1 and ZO-1 mRNA expressions, and plasma D-xylose levels were significantly decreased (p < 0.01) in piglets of group Z; while compare to Group C, ileal crypt depth, plasma D-lactate contents and diamine oxidase activities were significantly increased in piglets of group Z (p < 0.01 or p < 0.05). Compare with Group Z, ileal villus height, lipase and α-amylase activities, occluding, claudin-1 and ZO-1 mRNA expressions, and plasma D-xylose levels were significantly elevated in piglets of group ZS1 and ZS2 (p < 0.01); while compare to Group Z, plasma D-lactate and diamine oxidase contents were significantly reduced in piglets of group ZS1 and ZS2 (p < 0.01 or p < 0.05). Compare with Group Z, value of villus height/crypt depth and trypsin activity were significantly promoted in piglets of group ZS2 (p < 0.01); whereas ileal crypt depth was significantly reduced in piglets of group ZS2 (p <0.01).Thus, SOC can mitigate ZEN-induced intestinal dysfunction in piglets.
Collapse
Affiliation(s)
- Shunyi Qin
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
- Department of Agricultural Science and Technology, Hotan Vocational and Technical College, Hotan, People's Republic of China
| | - Yukai Peng
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Fuze She
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Jianbin Zhang
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Liuan Li
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Fu Chen
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
13
|
Skamrahl M, Schünemann J, Mukenhirn M, Pang H, Gottwald J, Jipp M, Ferle M, Rübeling A, Oswald T, Honigmann A, Janshoff A. Cellular segregation in cocultures is driven by differential adhesion and contractility on distinct timescales. Proc Natl Acad Sci U S A 2023; 120:e2213186120. [PMID: 37011207 PMCID: PMC10104523 DOI: 10.1073/pnas.2213186120] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 03/02/2023] [Indexed: 04/05/2023] Open
Abstract
Cellular sorting and pattern formation are crucial for many biological processes such as development, tissue regeneration, and cancer progression. Prominent physical driving forces for cellular sorting are differential adhesion and contractility. Here, we studied the segregation of epithelial cocultures containing highly contractile, ZO1/2-depleted MDCKII cells (dKD) and their wild-type (WT) counterparts using multiple quantitative, high-throughput methods to monitor their dynamical and mechanical properties. We observe a time-dependent segregation process governed mainly by differential contractility on short (<5 h) and differential adhesion on long (>5 h) timescales. The overly contractile dKD cells exert strong lateral forces on their WT neighbors, thereby apically depleting their surface area. Concomitantly, the tight junction-depleted, contractile cells exhibit weaker cell-cell adhesion and lower traction force. Drug-induced contractility reduction and partial calcium depletion delay the initial segregation but cease to change the final demixed state, rendering differential adhesion the dominant segregation force at longer timescales. This well-controlled model system shows how cell sorting is accomplished through a complex interplay between differential adhesion and contractility and can be explained largely by generic physical driving forces.
Collapse
Affiliation(s)
- Mark Skamrahl
- University of Göttingen, Institute of Physical Chemistry,37077Göttingen, Germany
| | - Justus Schünemann
- University of Göttingen, Institute of Physical Chemistry,37077Göttingen, Germany
| | - Markus Mukenhirn
- Max Planck Institute of Molecular Cell Biology and Genetics,01307Dresden, Germany
| | - Hongtao Pang
- University of Göttingen, Institute of Physical Chemistry,37077Göttingen, Germany
| | - Jannis Gottwald
- University of Göttingen, Institute of Physical Chemistry,37077Göttingen, Germany
| | - Marcel Jipp
- University of Göttingen, Institute of Physical Chemistry,37077Göttingen, Germany
| | - Maximilian Ferle
- University of Göttingen, Institute of Physical Chemistry,37077Göttingen, Germany
| | - Angela Rübeling
- University of Göttingen, Institute of Organic and Biomolecular Chemistry, Göttingen37077, Germany
| | - Tabea A. Oswald
- University of Göttingen, Institute of Organic and Biomolecular Chemistry, Göttingen37077, Germany
| | - Alf Honigmann
- Max Planck Institute of Molecular Cell Biology and Genetics,01307Dresden, Germany
| | - Andreas Janshoff
- University of Göttingen, Institute of Physical Chemistry,37077Göttingen, Germany
| |
Collapse
|
14
|
Li X, Liu YJ, Wang Y, Liu YF, Xu YJ. Epoxy Triglyceride Enhances Intestinal Permeability via Caspase-1/NLRP3/GSDMD and cGAS-STING Pathways in Dextran Sulfate Sodium-Induced Colitis Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:4371-4381. [PMID: 36857113 DOI: 10.1021/acs.jafc.2c08134] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Oxidized triglyceride monomers are the main cytotoxic products of deep-frying oil. However, its impact on the intestinal barrier, the first health guardian, remains unknown. In this study, HPLC-MS/MS analysis revealed that the epoxy group is the main oxidation product, indicating that it may be the main cytotoxic factor. Therefore, 1-9,10-epoxystearic ester, 2,3-dioleic acid (EGT) and glycerol trioleate (GT) were used to reveal the effect of the epoxy group on the intestinal barrier of dextran sulfate sodium-induced colitis. Characteristics analysis showed that EGT could aggravate intestinal damage. The relative mRNA expression analysis suggested that EGT could activate Caspase-1/NLRP3/GSDMD, thereby inducing pyroptosis. The proinflammatory cytokines activated by pyroptosis and the cGAS-STING pathway were released through the pores, thus inducing the disintegration of the tight junction between the intestinal epithelial cells and enhancing intestinal permeability. Metabonomics further confirmed that EGT can change the composition and content of phospholipids on the cell membrane, indicating the morphological changes of the intestinal epithelial cell membrane. In conclusion, this study highlights that EGT induced intestinal dysfunction via Caspase-1/NLRP3/GSDMD and cGAS-STING pathways.
Collapse
Affiliation(s)
- Xue Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Yan-Jun Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Yu Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Yuan-Fa Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Yong-Jiang Xu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, National Engineering Research Center for Functional Food, National Engineering Laboratory for Cereal Fermentation Technology, Collaborative Innovation Center of Food Safety and Quality Control, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| |
Collapse
|
15
|
Smith FM, Kosman DJ. Frataxin-deficient human brain microvascular endothelial cells lose polymerized actin and are paracellularly permeable -implications for blood-brain barrier integrity in Friedreich's Ataxia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.09.527936. [PMID: 36798283 PMCID: PMC9934603 DOI: 10.1101/2023.02.09.527936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Background Friedreich's Ataxia (FRDA) is the most prevalent inherited ataxia; the disease results from loss of Frataxin, an essential mitochondrial iron trafficking protein. FRDA presents as neurodegeneration of the dorsal root ganglion and cerebellar dentate nuclei, followed by brain iron accumulation in the latter. End stage disease includes cardiac fibrosis that contributes to hypertrophic cardiomyopathy. The microvasculature plays an essential barrier role in both the brain and heart, thus an investigation of this tissue system in FRDA is essential to the delineation of the cellular dysfunction in this genetic disorder. Here, we investigate brain microvascular endothelial cell integrity in FRDA in a model of the blood-brain barrier (BBB). Methods We used lentiviral mediated shRNA delivery to generate a novel FRDA model in immortalized human brain microvascular endothelial cells (hBMVEC) that compose the microcapillaries of the BBB. We verified known cellular pathophysiologies of FXN knockdown including increased oxidative stress, loss of energy metabolism, and increased cell size. Furthermore, we investigated cytoskeletal architecture including the abundance and organization of filamentous actin, and barrier physiology via transendothelial electrical resistance and fluorescent tracer flux. Results shFXN hBMVEC display the known FRDA cell morbidity including increased oxidative stress, decreased energy metabolism, and an increase in cell size. We demonstrate that shFXN hBMVEC have less overall filamentous actin, and that filamentous actin is lost at the cell membrane and cortical actin ring. Consistent with loss of cytoskeletal structure and anchorage, we found decreased barrier strength and increased paracellular tracer flux in the shFXN hBMVEC transwell model. Conclusion We identified that insufficient FXN levels in the hBMVEC BBB model causes changes in cytoskeletal architecture and increased barrier permeability, cell pathologies that may be related to patient brain iron accumulation, neuroinflammation, neurodegeneration, and stroke. Our findings implicate other barrier cells, e.g., the cardiac microvasculature, likely contributory also to disease pathology in FRDA.
Collapse
Affiliation(s)
- Frances M Smith
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, The University of New York at Buffalo
| | - Daniel J Kosman
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, The University of New York at Buffalo
| |
Collapse
|
16
|
ZO-1 Guides Tight Junction Assembly and Epithelial Morphogenesis via Cytoskeletal Tension-Dependent and -Independent Functions. Cells 2022; 11:cells11233775. [PMID: 36497035 PMCID: PMC9740252 DOI: 10.3390/cells11233775] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/08/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Formation and maintenance of tissue barriers require the coordination of cell mechanics and cell-cell junction assembly. Here, we combined methods to modulate ECM stiffness and to measure mechanical forces on adhesion complexes to investigate how tight junctions regulate cell mechanics and epithelial morphogenesis. We found that depletion of the tight junction adaptor ZO-1 disrupted junction assembly and morphogenesis in an ECM stiffness-dependent manner and led to a stiffness-dependant reorganisation of active myosin. Both junction formation and morphogenesis were rescued by inhibition of actomyosin contractility. ZO-1 depletion also impacted mechanical tension at cell-matrix and E-cadherin-based cell-cell adhesions. The effect on E-cadherin also depended on ECM stiffness and correlated with effects of ECM stiffness on actin cytoskeleton organisation. However, ZO-1 knockout also revealed tension-independent functions of ZO-1. ZO-1-deficient cells could assemble functional barriers at low tension, but their tight junctions remained corrupted with strongly reduced and discontinuous recruitment of junctional components. Our results thus reveal that reciprocal regulation between ZO-1 and cell mechanics controls tight junction assembly and epithelial morphogenesis, and that, in a second, tension-independent step, ZO-1 is required to assemble morphologically and structurally fully assembled and functionally normal tight junctions.
Collapse
|
17
|
Li DK, Chaudhari SN, Lee Y, Sojoodi M, Adhikari AA, Zukerberg L, Shroff S, Barrett SC, Tanabe K, Chung RT, Devlin AS. Inhibition of microbial deconjugation of micellar bile acids protects against intestinal permeability and liver injury. SCIENCE ADVANCES 2022; 8:eabo2794. [PMID: 36026454 PMCID: PMC9417178 DOI: 10.1126/sciadv.abo2794] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 07/13/2022] [Indexed: 06/15/2023]
Abstract
Altered host-microbe interactions and increased intestinal permeability have been implicated in disease pathogenesis. However, the mechanisms by which intestinal microbes affect epithelial barrier integrity remain unclear. Here, we investigate the impact of bacterial metabolism of host-produced bile acid (BA) metabolites on epithelial barrier integrity. We observe that rats fed a choline-deficient, l-amino acid-defined, high-fat diet (CDAHFD) exhibit reduced intestinal abundance of host-produced conjugated BAs at early time points, coinciding with increased gut permeability. We show that in vitro, conjugated BAs protect gut epithelial monolayers from damage caused by bacterially produced unconjugated BAs through micelle formation. We then demonstrate that inhibition of bacterial BA deconjugation with a small-molecule inhibitor prevents the development of pathologic intestinal permeability and hepatic inflammation in CDAHFD-fed rats. Our study identifies a signaling-independent, physicochemical mechanism for conjugated BA-mediated protection of epithelial barrier function and suggests that rational manipulation of microbial BA metabolism could be leveraged to regulate gut barrier integrity.
Collapse
Affiliation(s)
- Darrick K. Li
- Liver Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Snehal N. Chaudhari
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Yoojin Lee
- Liver Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mozhdeh Sojoodi
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Arijit A. Adhikari
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Lawrence Zukerberg
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Stuti Shroff
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephen Cole Barrett
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kenneth Tanabe
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Raymond T. Chung
- Liver Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - A. Sloan Devlin
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
18
|
Huang S, Tao R, Zhou J, Qian L, Wu J. Trans-10-Hydroxy-2-Decenoic Acid Alleviates Dextran Sulfate Sodium-Induced Colitis in Mice via Regulating the Inflammasome-Mediated Pyroptotic Pathway and Enhancing Colonic Barrier Function. Mol Nutr Food Res 2022; 66:e2100821. [PMID: 35373915 DOI: 10.1002/mnfr.202100821] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 01/31/2022] [Indexed: 12/24/2022]
Abstract
SCOPE The objective of this study is to explore the effects of 10-hydroxy-2-decenoic acid (10-HDA), the major fatty acid in royal jelly, on dextran sodium sulfate (DSS)-induced mice ulcerative colitis (UC) and its potential mechanism of action. METHODS AND RESULTS Forty male C57BL/6 mice are randomly divided into five experimental groups: control, DSS, DSS + 25 (or 100)mg kg-1 d-1 10-HDA, and DSS + 200 mg kg-1 d-1 mesalazine (ME). UC is induced in mice using 2.5% DSS in drinking water for 7 days. During the induction, these UC mice are orally administrated 10-HDA or ME per day. Meanwhile, lipopolysaccharide (LPS)/adenosine-triphosphate (ATP)-stimulated THP1 cells are used as a model to test the effects of 10-HDA. 10-HDA reduces DSS-induced pathological damage, reactive oxygen species (ROS) accumulation, neutrophil infiltration, and cytokine production in colonic tissue. Compared with the DSS group, the expressions of thioredoxin interacting protein (TXNIP), NOD-like receptor family pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC), cysteinyl aspartate specific proteinase-1 (Caspase-1), gasdermin-D (GSDMD), N-terminal domain of gasdermin-D (N-GSDMD), interleukin-1β (IL-1β), and interleukin-18 (IL-18) in the colon are decreased after administration of 10-HDA. 10-HDA also elevates the barrier integrity and the expressions of zonula occludens-1 (ZO-1) and Occludin in colonic epithelium exposed to DSS. In THP1 cells, the inflammasome-mediated pyroptosis induced by LPS/ATP is inhibited by 10-HDA pretreatment. CONCLUSION 10-HDA alleviates DSS-induced colitis by regulating the NLRP3 inflammasome-mediated pyroptotic pathway and enhancing colonic barrier function.
Collapse
Affiliation(s)
- Shanshan Huang
- Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University, School of Medicine, Shanghai, P. R. China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, P. R. China
| | - Ranran Tao
- Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University, School of Medicine, Shanghai, P. R. China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, P. R. China
| | - Jiefei Zhou
- Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University, School of Medicine, Shanghai, P. R. China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, P. R. China
| | - Linxi Qian
- Xinhua Hospital, Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University, School of Medicine, Shanghai, P. R. China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, P. R. China
| | - Jiang Wu
- Department of Nutrition, Huadong Hospital Affiliated to Fudan University, Shanghai, P. R. China
| |
Collapse
|
19
|
Kuo WT, Odenwald MA, Turner JR, Zuo L. Tight junction proteins occludin and ZO-1 as regulators of epithelial proliferation and survival. Ann N Y Acad Sci 2022; 1514:21-33. [PMID: 35580994 PMCID: PMC9427709 DOI: 10.1111/nyas.14798] [Citation(s) in RCA: 205] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Epithelial cells are the first line of mucosal defense. In the intestine, a single layer of epithelial cells must establish a selectively permeable barrier that supports nutrient absorption and waste secretion while preventing the leakage of potentially harmful luminal materials. Key to this is the tight junction, which seals the paracellular space and prevents unrestricted leakage. The tight junction is a protein complex established by interactions between members of the claudin, zonula occludens, and tight junction-associated MARVEL protein (TAMP) families. Claudins form the characteristic tight junction strands seen by freeze-fracture microscopy and create paracellular channels, but the functions of ZO-1 and occludin, founding members of the zonula occludens and TAMP families, respectively, are less well defined. Recent studies have revealed that these proteins have essential noncanonical (nonbarrier) functions that allow them to regulate epithelial apoptosis and proliferation, facilitate viral entry, and organize specialized epithelial structures. Surprisingly, neither is required for intestinal barrier function or overall health in the absence of exogenous stressors. Here, we provide a brief overview of ZO-1 and occludin canonical (barrier-related) functions, and a more detailed examination of their noncanonical functions.
Collapse
Affiliation(s)
- Wei-Ting Kuo
- Graduate Institute of Oral Biology, National Taiwan University, Taipei, Taiwan.,Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Li Zuo
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Anhui Medical University, Hefei, China
| |
Collapse
|
20
|
Zhang XZ, Huo HQ, Zhu YQ, Feng HY, Jiao J, Tan JX, Wang Y, Hu P, Xu ZF. Folic Acid Rescues Valproic Acid-Induced Morphogenesis Inhibition in Neural Rosettes Derived From Human Pluripotent Stem Cells. Front Cell Neurosci 2022; 16:888152. [PMID: 35651759 PMCID: PMC9148965 DOI: 10.3389/fncel.2022.888152] [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: 03/02/2022] [Accepted: 04/26/2022] [Indexed: 12/05/2022] Open
Abstract
The ability of human pluripotent stem cells (hPSCs) to specialize in neuroepithelial tissue makes them ideal candidates for use in the disease models of neural tube defects. In this study, we cultured hPSCs in suspension with modified neural induction method, and immunostaining was applied to detect important markers associated with cell fate and morphogenesis to verify the establishment of the neural tube model in vitro. We carried out the drug experiments to further investigate the toxicity of valproic acid (VPA) exposure and the potential protective effect of folic acid (FA). The results demonstrated that neural rosette undergoes cell fate speciation and lumen formation accompanied by a spatiotemporal shift in the expression patterns of cadherin, indicating the model was successfully established. The results showed that VPA caused morphogenesis inhibition of lumen formation by altering cytoskeletal function and cell polarization, which could be rescued by FA supplement.
Collapse
|
21
|
A Novel Herbal Extract Blend Product Prevents Particulate Matters-Induced Inflammation by Improving Gut Microbiota and Maintaining the Integrity of the Intestinal Barrier. Nutrients 2022; 14:nu14102010. [PMID: 35631153 PMCID: PMC9145798 DOI: 10.3390/nu14102010] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/26/2022] [Accepted: 05/07/2022] [Indexed: 02/05/2023] Open
Abstract
Air pollutants of PM2.5 can alter the composition of gut microbiota and lead to inflammation in the lung and gastrointestinal tract. The aim of this study was to evaluate the protective effect of a novel herbal extract blend, FC, composed of Lonicera japonica extract, Momordica grosvenori extract, and broccoli seed extract, on PM2.5-induced inflammation in the respiratory and intestinal tract. A549 cells and THP-1 cells, as well as C57BL/6 mice, were stimulated with PM2.5 to establish in vitro and in vivo exposure models. The models were treated with or without FC. The expression of inflammatory cytokines and tight junction proteins were studied. Proteomic analysis was performed to elucidate mechanisms. Mouse feces were collected for gut microbiota analysis. FC was shown to modulate the upregulation of pro-inflammatory cytokines mRNA expression in A549 and THP-1 cells and downregulated tight junction proteins mRNA expression in A549 cells due to PM2.5 stimulation. In animal models, the decreased expression of the anti-inflammatory factor il-10, tight junction protein ZO-1, and the elevated expression of COX-2 induced by PM2.5 were improved by FC intervention, which may be associated with zo-1 and cox-2 signaling pathways. In addition, FC was shown to improve the gut microbiota by increasing the abundance of beneficial bacteria.
Collapse
|
22
|
Inosine and D-Mannose Secreted by Drug-Resistant Klebsiella pneumoniae Affect Viability of Lung Epithelial Cells. Molecules 2022; 27:molecules27092994. [PMID: 35566345 PMCID: PMC9106066 DOI: 10.3390/molecules27092994] [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: 03/29/2022] [Revised: 04/30/2022] [Accepted: 05/03/2022] [Indexed: 11/29/2022] Open
Abstract
The antibiotic resistance rates of Klebsiella pneumoniae have been steadily increasing in recent years. Nevertheless, the metabolic features of the drug-resistant Klebsiella pneumoniae and its associated benefits for bacterial pathogenicity are far from expounded. This study aims to unravel the unique physiological and metabolic properties specific to drug-resistant K. pneumoniae. Using scanning electron microscopy (SEM), we observed a thicker extracellular mucus layer around a drug-resistant K. pneumonia strain (Kp-R) than a drug-sensitive K. pneumonia strain (Kp-S). Kp-R also produced more capsular polysaccharide (CPS) and biofilm, and appeared to have a significant competitive advantage when co-cultured with Kp-S. Moreover, Kp-R was easier to adhere to and invade A549 epithelial cells than Kp-S but caused less cell-viability damage according to cell counting kit-8 (CCK-8) tests. Immunofluorescence revealed that both Kp-R and Kp-S infection destroyed the tight junctions and F-actin of epithelial cells, while the damage caused by Kp-S was more severe than Kp-R. We detected the extracellular metabolites secreted by the two strains with UHPLC-Q-TOF MS to explore the critical secretion products. We identified 16 predominant compounds that were differentially expressed. Among them, inosine increased the viability of epithelial cells in a dose-dependent manner, and an A2AR antagonist can abolish such enhancement. D-mannose, which was secreted less in Kp-R, inhibited the viability of A549 cells in the range of low doses. These findings provide potential targets and research strategies for preventing and treating drug-resistant K. pneumoniae infections.
Collapse
|
23
|
Kabbesh H, Bulldan A, Konrad L, Scheiner-Bobis G. The Role of ZIP9 and Androgen Receptor in the Establishment of Tight Junctions between Adult Rat Sertoli Cells. BIOLOGY 2022; 11:biology11050668. [PMID: 35625396 PMCID: PMC9138102 DOI: 10.3390/biology11050668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 06/13/2023]
Abstract
The blood-testis barrier (BTB) is formed from tight junctions (TJs) between Sertoli cells. This dynamic structure, which establishes an immune-privileged environment protecting haploid germ cells formed in puberty from cells of the innate immune system, protects male fertility. Testosterone produced in Leydig cells is one of the main regulators of TJ protein expression and BTB dynamics. Nevertheless, although it has been assumed that testosterone effects on TJs and BTB are mediated through the classical androgen receptor (AR), newer results call the importance of this receptor into question. ZIP9, a recently identified androgen receptor of plasma membranes, mediates testosterone effects that promote the expression of TJ proteins and TJ formation in a rat Sertoli cell line that lacks the classical AR. Although these findings suggest that ZIP9 mediates these testosterone effects, participation of the classical AR in these events cannot be excluded. Here we used immortalized adult rat Sertoli cells that express both ZIP9 and AR and addressed the involvement of these receptors in the stimulation of TJ protein expression and TJ formation in response to testosterone and to the androgenic peptide IAPG that acts via ZIP9. We find that both testosterone and IAPG trigger the so-called non-classical signaling pathway of testosterone and stimulate the expression of TJ-associated proteins and TJ formation. Silencing classical AR expression had no effect on the responses, whereas silencing of ZIP9 expression completely blocked them. Our results demonstrate that ZIP9 is the sole androgen receptor involved in the regulation of TJ protein expression and TJ formation at the BTB.
Collapse
Affiliation(s)
- Hassan Kabbesh
- Institute for Veterinary Physiology and Biochemistry, School of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Str., D-35392 Giessen, Germany; (H.K.); (A.B.)
- Center of Gynecology and Obstetrics, Faculty of Medicine, Justus-Liebig-University Giessen, Feulgenstr. 10-12, D-35392 Giessen, Germany;
| | - Ahmed Bulldan
- Institute for Veterinary Physiology and Biochemistry, School of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Str., D-35392 Giessen, Germany; (H.K.); (A.B.)
| | - Lutz Konrad
- Center of Gynecology and Obstetrics, Faculty of Medicine, Justus-Liebig-University Giessen, Feulgenstr. 10-12, D-35392 Giessen, Germany;
| | - Georgios Scheiner-Bobis
- Institute for Veterinary Physiology and Biochemistry, School of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Str., D-35392 Giessen, Germany; (H.K.); (A.B.)
| |
Collapse
|
24
|
Presence of Natural Killer B Cells in Simian Immunodeficiency Virus-Infected Colon That Have Properties and Functions Similar to Those of Natural Killer Cells and B Cells but Are a Distinct Cell Population. J Virol 2022; 96:e0023522. [PMID: 35311549 PMCID: PMC9006943 DOI: 10.1128/jvi.00235-22] [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] [Indexed: 02/07/2023] Open
Abstract
There is low-level but significant mucosal inflammation in the gastrointestinal tract secondary to human immunodeficiency virus (HIV) infection that has long-term consequences for the infected host. This inflammation most likely originates from the immune response that appears as a consequence of HIV. Here, we show in an animal model of HIV that the chronically SIV-infected gut contains cytotoxic natural killer B cells that produce inflammatory cytokines and proliferate during infection.
Collapse
|
25
|
Yang HY, Liang ZH, Xie JL, Wu Q, Qin YY, Zhang SY, Tang GD. Gelsolin impairs barrier function in pancreatic ductal epithelial cells by actin filament depolymerization in hypertriglyceridemia‑induced pancreatitis in vitro. Exp Ther Med 2022; 23:290. [PMID: 35317441 PMCID: PMC8908475 DOI: 10.3892/etm.2022.11219] [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: 10/16/2021] [Accepted: 01/25/2022] [Indexed: 11/23/2022] Open
Abstract
Gelsolin (GSN) is a calcium-regulated actin-binding protein that can sever actin filaments. Notably, actin dynamics affect the structure and function of epithelial barriers. The present study investigated the role of GSN in the barrier function of pancreatic ductal epithelial cells (PDECs) in hypertriglyceridemia-induced pancreatitis (HTGP). The human PDEC cell line HPDE6-C7 underwent GSN knockdown and was treated with caerulein (CAE) + triglycerides (TG). Intracellular calcium levels and the actin filament network were analyzed under a fluorescence microscope. The expression levels of GSN, E-cadherin, nectin-2, ZO-1 and occludin were evaluated by reverse transcription-quantitative polymerase chain reaction and western blotting. Ultrastructural changes in tight junctions were observed by transmission electron microscopy. Furthermore, the permeability of PDECs was analyzed by fluorescein isothiocyanate-dextran fluorescence. The results revealed that CAE + TG increased intracellular calcium levels, actin filament depolymerization and GSN expression, and increased PDEC permeability by decreasing the expression levels of E-cadherin, nectin-2, ZO-1 and occludin compared with the control. Moreover, changes in these markers, with the exception of intracellular calcium levels, were reversed by silencing GSN. In conclusion, GSN may disrupt barrier function in PDECs by causing actin filament depolymerization in HTGP in vitro.
Collapse
Affiliation(s)
- Hui-Ying Yang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zhi-Hai Liang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jin-Lian Xie
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Qing Wu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530007, P.R. China
| | - Ying-Ying Qin
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Shi-Yu Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Guo-Du Tang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
26
|
Kuo WT, Zuo L, Odenwald MA, Madha S, Singh G, Gurniak CB, Abraham C, Turner JR. The Tight Junction Protein ZO-1 Is Dispensable for Barrier Function but Critical for Effective Mucosal Repair. Gastroenterology 2021; 161:1924-1939. [PMID: 34478742 PMCID: PMC8605999 DOI: 10.1053/j.gastro.2021.08.047] [Citation(s) in RCA: 251] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUNDS & AIMS Increased permeability is implicated in the pathogenesis of intestinal disease. In vitro and in vivo studies have linked down-regulation of the scaffolding protein ZO-1, encoded by the TJP1 gene, to increased tight junction permeability. This has not, however, been tested in vivo. Here, we assessed the contributions of ZO-1 to in vivo epithelial barrier function and mucosal homeostasis. METHODS Public Gene Expression Omnibus data sets and biopsy specimens from patients with inflammatory bowel disease (IBD) and healthy control individuals were analyzed. Tjp1f/f;vil-CreTg mice with intestinal epithelial-specific ZO-1 knockout (ZO-1KO.IEC) mice and Tjp1f/f mice littermates without Cre expression were studied using chemical and immune-mediated models of disease as well as colonic stem cell cultures. RESULTS ZO-1 transcript and protein expression were reduced in biopsy specimens from patients with IBD. Despite mildly increased intestinal permeability, ZO-1KO.IEC mice were healthy and did not develop spontaneous disease. ZO-1KO.IEC mice were, however, hypersensitive to mucosal insults and displayed defective repair. Furthermore, ZO-1-deficient colonic epithelia failed to up-regulate proliferation in response to damage in vivo or Wnt signaling in vitro. ZO-1 was associated with centrioles in interphase cells and mitotic spindle poles during division. In the absence of ZO-1, mitotic spindles failed to correctly orient, resulting in mitotic catastrophe and abortive proliferation. ZO-1 is, therefore, critical for up-regulation of epithelial proliferation and successful completion of mitosis. CONCLUSIONS ZO-1 makes critical, tight junction-independent contributions to Wnt signaling and mitotic spindle orientation. As a result, ZO-1 is essential for mucosal repair. We speculate that ZO-1 down-regulation may be one cause of ineffective mucosal healing in patients with IBD.
Collapse
Affiliation(s)
- Wei-Ting Kuo
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Li Zuo
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Anhui Medical University, Hefei, Anhui, China
| | | | - Shariq Madha
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Gurminder Singh
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Clara Abraham
- Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Department of Pathology, The University of Chicago, Chicago, Illinois.
| |
Collapse
|
27
|
Panwar S, Sharma S, Tripathi P. Role of Barrier Integrity and Dysfunctions in Maintaining the Healthy Gut and Their Health Outcomes. Front Physiol 2021; 12:715611. [PMID: 34630140 PMCID: PMC8497706 DOI: 10.3389/fphys.2021.715611] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/27/2021] [Indexed: 01/08/2023] Open
Abstract
Mucosal surface layers are the critical borders throughout epithelial membranes. These epithelial cells segregate luminal material from external environments. However, mucosal linings are also accountable for absorbing nutrients and requiring specific barrier permeability. These functional acts positioned the mucosal epithelium at the epicenter of communications concerning the mucosal immune coordination and foreign materials, such as dietary antigens and microbial metabolites. Current innovations have revealed that external stimuli can trigger several mechanisms regulated by intestinal mucosal barrier system. Crucial constituents of this epithelial boundary are physical intercellular structures known as tight junctions (TJs). TJs are composed of different types transmembrane proteins linked with cytoplasmic adaptors which helps in attachment to the adjacent cells. Disruption of this barrier has direct influence on healthy or diseased condition, as barrier dysfunctions have been interrelated with the initiation of inflammation, and pathogenic effects following metabolic complications. In this review we focus and overview the TJs structure, function and the diseases which are able to influence TJs during onset of disease. We also highlighted and discuss the role of phytochemicals evidenced to enhance the membrane permeability and integrity through restoring TJs levels.
Collapse
Affiliation(s)
- Shruti Panwar
- Infection and Immunology, Translational Health Science and Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| | - Sapna Sharma
- Gene Regulation Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Prabhanshu Tripathi
- Food Drug and Chemical Toxicology Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Toxicology Research, Lucknow, India
| |
Collapse
|
28
|
Skamrahl M, Pang H, Ferle M, Gottwald J, Rübeling A, Maraspini R, Honigmann A, Oswald TA, Janshoff A. Tight Junction ZO Proteins Maintain Tissue Fluidity, Ensuring Efficient Collective Cell Migration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100478. [PMID: 34382375 PMCID: PMC8498871 DOI: 10.1002/advs.202100478] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 06/18/2021] [Indexed: 06/01/2023]
Abstract
Tight junctions (TJs) are essential components of epithelial tissues connecting neighboring cells to provide protective barriers. While their general function to seal compartments is well understood, their role in collective cell migration is largely unexplored. Here, the importance of the TJ zonula occludens (ZO) proteins ZO1 and ZO2 for epithelial migration is investigated employing video microscopy in conjunction with velocimetry, segmentation, cell tracking, and atomic force microscopy/spectroscopy. The results indicate that ZO proteins are necessary for fast and coherent migration. In particular, ZO1 and 2 loss (dKD) induces actomyosin remodeling away from the central cortex towards the periphery of individual cells, resulting in altered viscoelastic properties. A tug-of-war emerges between two subpopulations of cells with distinct morphological and mechanical properties: 1) smaller and highly contractile cells with an outward bulging apical membrane, and 2) larger, flattened cells, which, due to tensile stress, display a higher proliferation rate. In response, the cell density increases, leading to crowding-induced jamming and more small cells over time. Co-cultures comprising wildtype and dKD cells migrate inefficiently due to phase separation based on differences in contractility rather than differential adhesion. This study shows that ZO proteins are necessary for efficient collective cell migration by maintaining tissue fluidity and controlling proliferation.
Collapse
Affiliation(s)
- Mark Skamrahl
- Institute of Physical ChemistryUniversity of GöttingenTammannstr. 6Göttingen37077Germany
| | - Hongtao Pang
- Institute of Physical ChemistryUniversity of GöttingenTammannstr. 6Göttingen37077Germany
| | - Maximilian Ferle
- Institute of Physical ChemistryUniversity of GöttingenTammannstr. 6Göttingen37077Germany
| | - Jannis Gottwald
- Institute of Physical ChemistryUniversity of GöttingenTammannstr. 6Göttingen37077Germany
| | - Angela Rübeling
- Institute of Organic and Biomolecular ChemistryUniversity of GöttingenTammannstr. 2Göttingen37077Germany
| | - Riccardo Maraspini
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstraße 108Dresden01307Germany
| | - Alf Honigmann
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstraße 108Dresden01307Germany
| | - Tabea A. Oswald
- Institute of Organic and Biomolecular ChemistryUniversity of GöttingenTammannstr. 2Göttingen37077Germany
| | - Andreas Janshoff
- Institute of Physical ChemistryUniversity of GöttingenTammannstr. 6Göttingen37077Germany
| |
Collapse
|
29
|
Vasquez CG, Vachharajani VT, Garzon-Coral C, Dunn AR. Physical basis for the determination of lumen shape in a simple epithelium. Nat Commun 2021; 12:5608. [PMID: 34556639 PMCID: PMC8460836 DOI: 10.1038/s41467-021-25050-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
The formation of a hollow lumen in a formerly solid mass of cells is a key developmental process whose dysregulation leads to diseases of the kidney and other organs. Hydrostatic pressure has been proposed to drive lumen expansion, a view that is supported by experiments in the mouse blastocyst. However, lumens formed in other tissues adopt irregular shapes with cell apical faces that are bowed inward, suggesting that pressure may not be the dominant contributor to lumen shape in all cases. Here we use live-cell imaging to study the physical mechanism of lumen formation in Madin-Darby Canine Kidney cell spheroids, a canonical cell-culture model for lumenogenesis. We find that in this system, lumen shape reflects basic geometrical considerations tied to the establishment of apico-basal polarity. A physical model incorporating both cell geometry and intraluminal pressure can account for our observations as well as cases in which pressure plays a dominant role.
Collapse
Affiliation(s)
| | | | | | - Alexander R Dunn
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
30
|
Li W, Yuan W, Zhang D, Cai S, Luo J, Zeng K. LCZ696 Possesses a Protective Effect Against Homocysteine (Hcy)-Induced Impairment of Blood-Brain Barrier (BBB) Integrity by Increasing Occludin, Mediated by the Inhibition of Egr-1. Neurotox Res 2021; 39:1981-1990. [PMID: 34542838 DOI: 10.1007/s12640-021-00414-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/24/2021] [Accepted: 09/03/2021] [Indexed: 12/14/2022]
Abstract
Homocysteine (Hcy) is a non-essential amino acid produced from methionine. It has been reported that high concentrations of Hcy are related to the pathogenesis of neurodegenerative diseases and induce the disruption of the blood-brain barrier (BBB) by triggering oxidative stress and inflammation. LCZ696 is a novel antihypertensive agent that has been recently reported to possess promising anti-inflammatory properties. However, whether it has a protective effect on the BBB disruption is still unknown. For the first time, in this study, we aim to investigate whether LCZ696 exerts anti-inflammatory effects on Hcy-induced injury in brain endothelial cells and explore its neuroprotective properties. In in vivo experiments, we found that treatment with LCZ696 ameliorated oxidative stress by reducing malondialdehyde (MDA) and increasing glutathione (GSH). Furthermore, LCZ696 downregulated the excessive release of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) at mRNA and protein levels. Importantly, it reversed the disruption of the BBB induced by Hcy stimulation. In the in vitro human brain microvascular endothelial cell (HBMVEC) experiments, compared to the control, the permeability of the endothelial monolayer was significantly enlarged, the expression level of occludin declined, and Egr-1 upregulated by the introduction of Hcy, and these were all reversed by the treatment with LCZ696. Lastly, we found that the protective effects of LCZ696 against Hcy-induced reduction of occludin and hyper-permeability of the endothelial monolayer were greatly abolished by the overexpression of Egr-1. Taken together, we found that LCZ696 protected against Hcy-induced impairment of BBB integrity by increasing the expression of occludin, all mediated by the inhibition of Egr-1.
Collapse
Affiliation(s)
- Wenfeng Li
- Department of Cardiology, The First Affiliated Hospital of Ji'nan University, Guangzhou, 510630, Guangdong, China.,Department of Cardiology, Ganzhou People's Hospital, Ganzhou, 341000, Jiangxi, China
| | - Wenjin Yuan
- Department of Cardiology, Ganzhou People's Hospital, Ganzhou, 341000, Jiangxi, China
| | - Dandan Zhang
- Department of Cardiology, Ganzhou People's Hospital, Ganzhou, 341000, Jiangxi, China
| | - Shuchun Cai
- Department of Cardiology, Ganzhou People's Hospital, Ganzhou, 341000, Jiangxi, China
| | - Jun Luo
- Department of Cardiology, Ganzhou People's Hospital, Ganzhou, 341000, Jiangxi, China
| | - Kanghua Zeng
- Department of Cardiology, Ganzhou People's Hospital, Ganzhou, 341000, Jiangxi, China.
| |
Collapse
|
31
|
Beneficial Effects of Transplanted Human Bone Marrow Endothelial Progenitors on Functional and Cellular Components of Blood-Spinal Cord Barrier in ALS Mice. eNeuro 2021; 8:ENEURO.0314-21.2021. [PMID: 34479980 PMCID: PMC8451202 DOI: 10.1523/eneuro.0314-21.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 12/18/2022] Open
Abstract
Convincing evidence of blood-spinal cord barrier (BSCB) alterations has been demonstrated in amyotrophic lateral sclerosis (ALS) and barrier repair is imperative to prevent motor neuron dysfunction. We showed benefits of human bone marrow-derived CD34+ cells (hBM34+) and endothelial progenitor cells (hBM-EPCs) intravenous transplantation into symptomatic G93A SOD1 mutant mice on barrier reparative processes. These gains likely occurred by replacement of damaged endothelial cells, prolonging motor neuron survival. However, additional investigations are needed to confirm the effects of administered cells on integrity of the microvascular endothelium. The aim of this study was to determine tight junction protein levels, capillary pericyte coverage, microvascular basement membrane, and endothelial filamentous actin (F-actin) status in spinal cord capillaries of G93A SOD1 mutant mice treated with human bone marrow-derived stem cells. Tight junction proteins were detected in the spinal cords of cell-treated versus non-treated mice via Western blotting at four weeks after transplant. Capillary pericyte, basement membrane laminin, and endothelial F-actin magnitudes were determined in cervical/lumbar spinal cord tissues in ALS mice, including controls, by immunohistochemistry and fluorescent staining. Results showed that cell-treated versus media-treated ALS mice substantially increased tight junction protein levels, capillary pericyte coverage, basement membrane laminin immunoexpressions, and endothelial cytoskeletal F-actin fluorescent expressions. The greatest benefits were detected in mice receiving hBM-EPCs versus hBM34+ cells. These study results support treatment with a specific cell type derived from human bone marrow toward BSCB repair in ALS. Thus, hBM-EPCs may be advanced for clinical applications as a cell-specific approach for ALS therapy through restored barrier integrity.
Collapse
|
32
|
Hwang D, Kim M, Kim S, Kwon MR, Kang YS, Kim D, Kang HC, Lim DS. AMOTL2 mono-ubiquitination by WWP1 promotes contact inhibition by facilitating LATS activation. Life Sci Alliance 2021; 4:4/10/e202000953. [PMID: 34404733 PMCID: PMC8372784 DOI: 10.26508/lsa.202000953] [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: 11/04/2020] [Revised: 08/06/2021] [Accepted: 08/06/2021] [Indexed: 11/24/2022] Open
Abstract
This work reveals a novel function of WWP1 E3 ligase in the mono-ubiquitination of AMOTL2, which enables the binding and activation of LATS kinases upon contact inhibition. Contact inhibition is a key cellular phenomenon that prevents cells from hyper-proliferating upon reaching confluence. Although not fully characterized, a critical driver of this process is the Hippo signaling pathway, whose downstream effector yes-associated protein plays pivotal roles in cell growth and differentiation. Here, we provide evidence that the E3 ligase WWP1 (WW-domain containing protein 1) mono-ubiquitinates AMOTL2 (angiomotin-like 2) at K347 and K408. Mono-ubiquitinated AMOTL2, in turn, interacts with the kinase LATS2, which facilitates recruitment of the upstream Hippo pathway component SAV1 and ultimately promotes yes-associated protein phosphorylation and subsequent cytoplasmic sequestration and/or degradation. Furthermore, contact inhibition induced by high cell density promoted the localization and stabilization of WWP1 at cell junctions, where it interacted with Crumbs polarity proteins. Notably, the Crumbs complex was functionally important for AMOTL2 mono-ubiquitination and LATS activation under high cell density conditions. These findings delineate a functionally important molecular mechanism in which AMOTL2 mono-ubiquitination by WWP1 at cell junctions and LATS activation are tightly coupled to upstream cell density cues.
Collapse
Affiliation(s)
- Daehee Hwang
- National Creative Research Center for Cell Plasticity, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Miju Kim
- National Creative Research Center for Cell Plasticity, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Soyeon Kim
- Genomic Instability Research Center, Ajou University School of Medicine, Suwon, Korea
| | - Mi Ra Kwon
- Genomic Instability Research Center, Ajou University School of Medicine, Suwon, Korea
| | - Ye-Seul Kang
- National Creative Research Center for Cell Plasticity, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Dahyun Kim
- National Creative Research Center for Cell Plasticity, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Ho-Chul Kang
- Genomic Instability Research Center, Ajou University School of Medicine, Suwon, Korea
| | - Dae-Sik Lim
- National Creative Research Center for Cell Plasticity, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| |
Collapse
|
33
|
Abstract
Mechanical forces have emerged as essential regulators of cell organization, proliferation, migration, and polarity to regulate cellular and tissue homeostasis. Changes in forces or loss of the cellular response to them can result in abnormal embryonic development and diseases. Over the past two decades, many efforts have been put in deciphering the molecular mechanisms that convert forces into biochemical signals, allowing for the identification of many mechanotransducer proteins. Here we discuss how PDZ proteins are emerging as new mechanotransducer proteins by altering their conformations or localizations upon force loads, leading to the formation of macromolecular modules tethering the cell membrane to the actin cytoskeleton.
Collapse
|
34
|
Park SY, Jang H, Kim SY, Kim D, Park Y, Kee SH. Expression of E-Cadherin in Epithelial Cancer Cells Increases Cell Motility and Directionality through the Localization of ZO-1 during Collective Cell Migration. Bioengineering (Basel) 2021; 8:bioengineering8050065. [PMID: 34064908 PMCID: PMC8151941 DOI: 10.3390/bioengineering8050065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 02/01/2023] Open
Abstract
Collective cell migration of epithelial tumor cells is one of the important factors for elucidating cancer metastasis and developing novel drugs for cancer treatment. Especially, new roles of E-cadherin in cancer migration and metastasis, beyond the epithelial–mesenchymal transition, have recently been unveiled. Here, we quantitatively examined cell motility using micropatterned free edge migration model with E-cadherin re-expressing EC96 cells derived from adenocarcinoma gastric (AGS) cell line. EC96 cells showed increased migration features such as the expansion of cell islands and straightforward movement compared to AGS cells. The function of tight junction proteins known to E-cadherin expression were evaluated for cell migration by knockdown using sh-RNA. Cell migration and straight movement of EC96 cells were reduced by knockdown of ZO-1 and claudin-7, to a lesser degree. Analysis of the migratory activity of boundary cells and inner cells shows that EC96 cell migration was primarily conducted by boundary cells, similar to leader cells in collective migration. Immunofluorescence analysis showed that tight junctions (TJs) of EC96 cells might play important roles in intracellular communication among boundary cells. ZO-1 is localized to the base of protruding lamellipodia and cell contact sites at the rear of cells, indicating that ZO-1 might be important for the interaction between traction and tensile forces. Overall, dynamic regulation of E-cadherin expression and localization by interaction with ZO-1 protein is one of the targets for elucidating the mechanism of collective migration of cancer metastasis.
Collapse
Affiliation(s)
- Song-Yi Park
- Department of Microbiology, College of Medicine, Korea University, Seoul 02841, Korea; (S.-Y.P.); (S.-Y.K.); (D.K.)
| | - Hwanseok Jang
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea; or
| | - Seon-Young Kim
- Department of Microbiology, College of Medicine, Korea University, Seoul 02841, Korea; (S.-Y.P.); (S.-Y.K.); (D.K.)
| | - Dasarang Kim
- Department of Microbiology, College of Medicine, Korea University, Seoul 02841, Korea; (S.-Y.P.); (S.-Y.K.); (D.K.)
| | - Yongdoo Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea; or
- Correspondence: (Y.P.); (S.-H.K.); Tel.: +82-2-2286-1460 (Y.P.); +82-2-2286-1165 (S.-H.K.)
| | - Sun-Ho Kee
- Department of Microbiology, College of Medicine, Korea University, Seoul 02841, Korea; (S.-Y.P.); (S.-Y.K.); (D.K.)
- Correspondence: (Y.P.); (S.-H.K.); Tel.: +82-2-2286-1460 (Y.P.); +82-2-2286-1165 (S.-H.K.)
| |
Collapse
|
35
|
Abstract
The apical junctional complexes (AJCs) of airway epithelial cells are a key component of the innate immune system by creating barriers to pathogens, inhaled allergens, and environmental particles. AJCs form between adjacent cells and consist of tight junctions (TJs) and adherens junctions (AJs). Respiratory viruses have been shown to target various components of the AJCs, leading to airway epithelial barrier dysfunction by different mechanisms. Virus-induced epithelial permeability may allow for allergens and bacterial pathogens to subsequently invade. In this review, we discuss the pathophysiologic mechanisms leading to disruption of AJCs and the potential ensuing ramifications. We focus on the following viruses that affect the pulmonary system: respiratory syncytial virus, rhinovirus, influenza viruses, immunodeficiency virus, and other viruses such as coxsackievirus, adenovirus, coronaviruses, measles, parainfluenza virus, bocavirus, and vaccinia virus. Understanding the mechanisms by which viruses target the AJC and impair barrier function may help design therapeutic innovations to treat these infections.
Collapse
Affiliation(s)
- Debra T Linfield
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Andjela Raduka
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, Ohio, USA
| | - Mahyar Aghapour
- Institute of Medical Microbiology, Otto-von-Guericke University, Magdeburg, Germany
| | - Fariba Rezaee
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, Ohio, USA.,Center for Pediatric Pulmonary Medicine, Cleveland, Ohio, USA
| |
Collapse
|
36
|
Hof L, Moreth T, Koch M, Liebisch T, Kurtz M, Tarnick J, Lissek SM, Verstegen MMA, van der Laan LJW, Huch M, Matthäus F, Stelzer EHK, Pampaloni F. Long-term live imaging and multiscale analysis identify heterogeneity and core principles of epithelial organoid morphogenesis. BMC Biol 2021; 19:37. [PMID: 33627108 PMCID: PMC7903752 DOI: 10.1186/s12915-021-00958-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 01/12/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Organoids are morphologically heterogeneous three-dimensional cell culture systems and serve as an ideal model for understanding the principles of collective cell behaviour in mammalian organs during development, homeostasis, regeneration, and pathogenesis. To investigate the underlying cell organisation principles of organoids, we imaged hundreds of pancreas and cholangiocarcinoma organoids in parallel using light sheet and bright-field microscopy for up to 7 days. RESULTS We quantified organoid behaviour at single-cell (microscale), individual-organoid (mesoscale), and entire-culture (macroscale) levels. At single-cell resolution, we monitored formation, monolayer polarisation, and degeneration and identified diverse behaviours, including lumen expansion and decline (size oscillation), migration, rotation, and multi-organoid fusion. Detailed individual organoid quantifications lead to a mechanical 3D agent-based model. A derived scaling law and simulations support the hypotheses that size oscillations depend on organoid properties and cell division dynamics, which is confirmed by bright-field microscopy analysis of entire cultures. CONCLUSION Our multiscale analysis provides a systematic picture of the diversity of cell organisation in organoids by identifying and quantifying the core regulatory principles of organoid morphogenesis.
Collapse
Affiliation(s)
- Lotta Hof
- Physical Biology Group, Buchmann Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Till Moreth
- Physical Biology Group, Buchmann Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Michael Koch
- Physical Biology Group, Buchmann Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Tim Liebisch
- Frankfurt Institute for Advanced Studies and Faculty of Biological Sciences, Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Marina Kurtz
- Department of Physics, Goethe Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Julia Tarnick
- Deanery of Biomedical Science, University of Edinburgh, Edinburgh, UK
| | - Susanna M Lissek
- Experimental Medicine and Therapy Research, University of Regensburg, Regensburg, Germany
| | - Monique M A Verstegen
- Department of Surgery, Erasmus MC - University Medical Center, Rotterdam, The Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC - University Medical Center, Rotterdam, The Netherlands
| | - Meritxell Huch
- The Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge, UK
- Present address: Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Franziska Matthäus
- Frankfurt Institute for Advanced Studies and Faculty of Biological Sciences, Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Ernst H K Stelzer
- Physical Biology Group, Buchmann Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Francesco Pampaloni
- Physical Biology Group, Buchmann Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany.
| |
Collapse
|
37
|
Xie W, Chen M, Zhai Z, Li H, Song T, Zhu Y, Dong D, Zhou P, Duan L, Zhang Y, Li D, Liu X, Zhou J, Liu M. HIV-1 exposure promotes PKG1-mediated phosphorylation and degradation of stathmin to increase epithelial barrier permeability. J Biol Chem 2021; 296:100644. [PMID: 33839152 PMCID: PMC8105298 DOI: 10.1016/j.jbc.2021.100644] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 03/29/2021] [Accepted: 04/05/2021] [Indexed: 01/11/2023] Open
Abstract
Exposure of mucosal epithelial cells to the human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein gp120 is known to disrupt epithelial cell junctions by impairing stathmin-mediated microtubule depolymerization. However, the pathological significance of this process and its underlying molecular mechanism remain unclear. Here we show that treatment of epithelial cells with pseudotyped HIV-1 viral particles or recombinant gp120 protein results in the activation of protein kinase G 1 (PKG1). Examination of epithelial cells by immunofluorescence microscopy reveals that PKG1 activation mediates the epithelial barrier damage upon HIV-1 exposure. Immunoprecipitation experiments show that PKG1 interacts with stathmin and phosphorylates stathmin at serine 63 in the presence of gp120. Immunoprecipitation and immunofluorescence microscopy further demonstrate that PKG1-mediated phosphorylation of stathmin promotes its autophagic degradation by enhancing the interaction between stathmin and the autophagy adaptor protein p62. Collectively, these results suggest that HIV-1 exposure exploits the PKG1/stathmin axis to affect the microtubule cytoskeleton and thereby perturbs epithelial cell junctions. Our findings reveal a novel molecular mechanism by which exposure to HIV-1 increases epithelial permeability, which has implications for the development of effective strategies to prevent mucosal HIV-1 transmission.
Collapse
Affiliation(s)
- Wei Xie
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Mingzhen Chen
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Zhaodong Zhai
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Hongjie Li
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Ting Song
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Yigao Zhu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Dan Dong
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Peng Zhou
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Liangwei Duan
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - You Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Dengwen Li
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Xinqi Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Jun Zhou
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China; State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China.
| | - Min Liu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China.
| |
Collapse
|
38
|
Doxorubicin increases permeability of murine small intestinal epithelium and cultured T84 monolayers. Sci Rep 2020; 10:21486. [PMID: 33293626 PMCID: PMC7722747 DOI: 10.1038/s41598-020-78473-1] [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: 06/16/2020] [Accepted: 11/23/2020] [Indexed: 12/17/2022] Open
Abstract
Enteric bacteria and/or their products are necessary for doxorubicin (DXR)-induced small intestine mucosal damage. While DXR does not induce gross loss of epithelium, others have shown elevated serum endotoxin after DXR administration. However, the mechanism of movement is unknown. We hypothesized that DXR treatment resulted in increased paracellular translocation of bacteria or bacterial products through the small intestinal epithelium. We measured permeability after DXR administration using transepithelial resistance and macromolecular flux and assessed tight junctional gene expression and protein localization both in vitro using T84 cells and ex vivo using murine jejunum. DXR treatment increased flux of 4 kDa dextrans in mouse jejenum, but increased flux of 4, 10 and 20 kDa dextrans in T84 cells. Following DXR, we observed increased permeability, both in vitro and ex vivo, independent of bacteria. DXR induced increased expression of Cldn2 and Cldn4 in murine small intestine but increased only CLDN2 expression in T84 cells. DXR treatment induced disorganization of tight junctional proteins. We conclude that DXR increases paracellular transit of small macromolecules, including bacterial products, through the epithelium, by altering expression of tight junctional components and dynamic loosening of cellular tight junctions.
Collapse
|
39
|
Ameliorative Effect of Sinapic Acid on Dextran Sodium Sulfate- (DSS-) Induced Ulcerative Colitis in Kunming (KM) Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8393504. [PMID: 33312339 PMCID: PMC7719534 DOI: 10.1155/2020/8393504] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/11/2019] [Accepted: 10/16/2019] [Indexed: 12/25/2022]
Abstract
Ulcerative colitis is a chronic gastrointestinal disease characterized by intestinal inflammation and serious mucosal damage. As a naturally hydroxycinnamic acid, sinapic acid (SA) has antioxidant, anticancer, and neuroprotective activities. We investigated the anticolitic effect and potential mechanisms of SA in DSS-induced colitis in Kunming (KM) mice. SA treatment significantly reduced body weight loss, colon shortening, and intestinal wall thickening in colitis mice. SA treatment also significantly reduced the histological infiltration of inflammatory cells and decreased myeloperoxidase (MPO) activity in the colons of colitis mice. The administration of SA attenuated oxidative damage by enhancing the activity of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and catalase and reduced the serum and colonic mRNA levels of proinflammatory cytokines in colitis mice. We used qRT-PCR and Western blotting assays and demonstrated that SA reduced the activation of the NLRP3 inflammasome and attenuated intestinal permeability by enhancing the expression of ZO-1, occludin, and claudin-1 in colitis mice. Here, we conclude that SA exhibits great anticolitic activity against DSS-induced colitis by enhancing the activity of antioxidant enzymes, reducing intestinal inflammation, and maintaining the intestinal barrier. Finally, we suggest that SA may be a safe adjuvant for the prevention of clinical colitis.
Collapse
|
40
|
The PDZ motif peptide of ZO-1 attenuates Pseudomonas aeruginosa LPS-induced airway inflammation. Sci Rep 2020; 10:19644. [PMID: 33184397 PMCID: PMC7665049 DOI: 10.1038/s41598-020-76883-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 11/02/2020] [Indexed: 11/24/2022] Open
Abstract
Pseudomonas aeruginosa is known to play a role in many human diseases. Therefore, examining the negative control mechanisms of tight junction protein ZO-1 on the exotoxin LPS of P. aeruginosa-induced diseases could be critical in the development of novel therapeutics. We found that ZO-1 expression dramatically decreased in inflammatory human lung tissues. Interestingly, PDZ1 deletion of the PDZ domain in the ZO-1 protein dramatically decreased LPS-induced F-actin formation and increased the expression of genes for pro-inflammatory cytokines, but not PDZ2 and PDZ3 of the ZO-1 protein. We also found that the consensus PDZ peptide (based on PDZ1) of ZO-1 down-regulates the expression of pro-inflammatory cytokine genes and F-actin formation; in contrast, the GG24,25AA mutant PDZ peptide cannot control these genes. LPS activates IL-8 secretion extracellularly in a time-dependent manner, while the secretion is inhibited by PDZ peptide. Whereas increased IL-8 secretion by LPS activates the CXCR2 receptor, overexpressed RGS12 negatively regulates LPS-induced CXCR2/IL-8 signaling. The PDZ peptide also decreases LPS-induced inflammatory cell populations, pro-inflammatory cytokine gene expression, and TEER in bronchoalveolar lavage fluid and cultured alveolar macrophages. Collectively, we suggest that the PDZ peptide may be a potential therapeutic for bacteria-induced respiratory diseases.
Collapse
|
41
|
Buglak DB, Kushner EJ, Marvin AP, Davis KL, Bautch VL. Excess centrosomes disrupt vascular lumenization and endothelial cell adherens junctions. Angiogenesis 2020; 23:567-575. [PMID: 32699963 PMCID: PMC7524686 DOI: 10.1007/s10456-020-09737-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 07/08/2020] [Indexed: 12/12/2022]
Abstract
Proper blood vessel formation requires coordinated changes in endothelial cell polarity and rearrangement of cell-cell junctions to form a functional lumen. One important regulator of cell polarity is the centrosome, which acts as a microtubule organizing center. Excess centrosomes perturb aspects of endothelial cell polarity linked to migration, but whether centrosome number influences apical-basal polarity and cell-cell junctions is unknown. Here, we show that excess centrosomes alter the apical-basal polarity of endothelial cells in angiogenic sprouts and disrupt endothelial cell-cell adherens junctions. Endothelial cells with excess centrosomes had narrower lumens in a 3D sprouting angiogenesis model, and zebrafish intersegmental vessels had reduced perfusion following centrosome overduplication. These results indicate that endothelial cell centrosome number regulates proper lumenization downstream of effects on apical-basal polarity and cell-cell junctions. Endothelial cells with excess centrosomes are prevalent in tumor vessels, suggesting how centrosomes may contribute to tumor vessel dysfunction.
Collapse
Affiliation(s)
- Danielle B Buglak
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Erich J Kushner
- Department of Biology, The University of North Carolina at Chapel Hill, CB#3280, Chapel Hill, NC, 27599, USA
- Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Allison P Marvin
- Department of Biology, The University of North Carolina at Chapel Hill, CB#3280, Chapel Hill, NC, 27599, USA
| | - Katy L Davis
- Department of Biology, The University of North Carolina at Chapel Hill, CB#3280, Chapel Hill, NC, 27599, USA
| | - Victoria L Bautch
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Biology, The University of North Carolina at Chapel Hill, CB#3280, Chapel Hill, NC, 27599, USA.
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
42
|
Xiong B, Liu M, Zhang C, Hao Y, Zhang P, Chen L, Tang X, Zhang H, Zhao Y. Alginate oligosaccharides enhance small intestine cell integrity and migration ability. Life Sci 2020; 258:118085. [DOI: 10.1016/j.lfs.2020.118085] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/10/2020] [Accepted: 07/08/2020] [Indexed: 01/27/2023]
|
43
|
Otani T, Furuse M. Tight Junction Structure and Function Revisited. Trends Cell Biol 2020; 30:805-817. [PMID: 32891490 DOI: 10.1016/j.tcb.2020.08.004] [Citation(s) in RCA: 380] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 12/14/2022]
Abstract
Tight junctions (TJs) are intercellular junctions critical for building the epithelial barrier and maintaining epithelial polarity. The claudin family of membrane proteins play central roles in TJ structure and function. However, recent findings have uncovered claudin-independent aspects of TJ structure and function, and additional players including junctional adhesion molecules (JAMs), membrane lipids, phase separation of the zonula occludens (ZO) family of scaffolding proteins, and mechanical force have been shown to play important roles in TJ structure and function. In this review, we discuss how these new findings have the potential to transform our understanding of TJ structure and function, and how the intricate network of TJ proteins and membrane lipids dynamically interact to drive TJ assembly.
Collapse
Affiliation(s)
- Tetsuhisa Otani
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi 444-8787, Japan; Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8787, Japan.
| | - Mikio Furuse
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Aichi 444-8787, Japan; Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8787, Japan
| |
Collapse
|
44
|
Formiga RDO, Alves Júnior EB, Vasconcelos RC, Guerra GCB, Antunes de Araújo A, de Carvalho TG, Garcia VB, de Araújo Junior RF, Gadelha FAAF, Vieira GC, Sobral MV, Barbosa Filho JM, Spiller F, Batista LM. p-Cymene and Rosmarinic Acid Ameliorate TNBS-Induced Intestinal Inflammation Upkeeping ZO-1 and MUC-2: Role of Antioxidant System and Immunomodulation. Int J Mol Sci 2020; 21:E5870. [PMID: 32824269 PMCID: PMC7461622 DOI: 10.3390/ijms21165870] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/04/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Abstract
p-Cymene (p-C) and rosmarinic acid (RA) are secondary metabolites that are present in medicinal herbs and Mediterranean spices that have promising anti-inflammatory properties. This study aimed to evaluate their intestinal anti-inflammatory activity in the trinitrobenzene sulphonic acid (TNBS)-induced colitis model in rats. p-C and RA (25-200 mg/kg) oral administration reduced the macroscopic lesion score, ulcerative area, intestinal weight/length ratio, and diarrheal index in TNBS-treated animals. Both compounds (200 mg/kg) decreased malondialdehyde (MDA) and myeloperoxidase (MPO), restored glutathione (GSH) levels, and enhanced fluorescence intensity of superoxide dismutase (SOD). They also decreased interleukin (IL)-1β and tumor necrosis factor (TNF)-α, and maintained IL-10 basal levels. Furthermore, they modulated T cell populations (cluster of differentiation (CD)4+, CD8+, or CD3+CD4+CD25+) analyzed from the spleen, mesenteric lymph nodes, and colon samples, and also decreased cyclooxigenase 2 (COX-2), interferon (IFN)-γ, inducible nitric oxide synthase (iNOS), and nuclear transcription factor kappa B subunit p65 (NFκB-p65) mRNA transcription, but only p-C interfered in the suppressor of cytokine signaling 3 (SOCS3) expression in inflamed colons. An increase in gene expression and positive cells immunostained for mucin type 2 (MUC-2) and zonula occludens 1 (ZO-1) was observed. Altogether, these results indicate intestinal anti-inflammatory activity of p-C and RA involving the cytoprotection of the intestinal barrier, maintaining the mucus layer, and preserving communicating junctions, as well as through modulation of the antioxidant and immunomodulatory systems.
Collapse
Affiliation(s)
- Rodrigo de Oliveira Formiga
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa 58051970, Brazil; (R.d.O.F); (E.B.A.J.); (F.A.A.F.G.); (G.C.V.); (M.V.S.); (J.M.B.F.)
| | - Edvaldo Balbino Alves Júnior
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa 58051970, Brazil; (R.d.O.F); (E.B.A.J.); (F.A.A.F.G.); (G.C.V.); (M.V.S.); (J.M.B.F.)
| | - Roseane Carvalho Vasconcelos
- Department of Biophysics and Pharmacology, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59064-741, Brazil; (R.C.V); (G.C.B.G.); (A.A.d.A.)
| | - Gerlane Coelho Bernardo Guerra
- Department of Biophysics and Pharmacology, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59064-741, Brazil; (R.C.V); (G.C.B.G.); (A.A.d.A.)
| | - Aurigena Antunes de Araújo
- Department of Biophysics and Pharmacology, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59064-741, Brazil; (R.C.V); (G.C.B.G.); (A.A.d.A.)
| | - Thaís Gomes de Carvalho
- Department of Morphology, Histology and Basic Pathology, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59064-741, Brazil; (T.G.d.C.); (V.B.G.); (R.F.d.A.J.)
| | - Vinícius Barreto Garcia
- Department of Morphology, Histology and Basic Pathology, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59064-741, Brazil; (T.G.d.C.); (V.B.G.); (R.F.d.A.J.)
| | - Raimundo Fernandes de Araújo Junior
- Department of Morphology, Histology and Basic Pathology, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59064-741, Brazil; (T.G.d.C.); (V.B.G.); (R.F.d.A.J.)
| | - Francisco Allysson Assis Ferreira Gadelha
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa 58051970, Brazil; (R.d.O.F); (E.B.A.J.); (F.A.A.F.G.); (G.C.V.); (M.V.S.); (J.M.B.F.)
| | - Giciane Carvalho Vieira
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa 58051970, Brazil; (R.d.O.F); (E.B.A.J.); (F.A.A.F.G.); (G.C.V.); (M.V.S.); (J.M.B.F.)
| | - Marianna Vieira Sobral
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa 58051970, Brazil; (R.d.O.F); (E.B.A.J.); (F.A.A.F.G.); (G.C.V.); (M.V.S.); (J.M.B.F.)
| | - José Maria Barbosa Filho
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa 58051970, Brazil; (R.d.O.F); (E.B.A.J.); (F.A.A.F.G.); (G.C.V.); (M.V.S.); (J.M.B.F.)
| | - Fernando Spiller
- Department of Pharmacology, Federal University of Santa Catarina (UFSC), Florianópolis 88037-000, Brazil;
| | - Leônia Maria Batista
- Postgraduate Program in Natural and Synthetic Bioactive Products, Health Sciences Center, Federal University of Paraíba (UFPB), João Pessoa 58051970, Brazil; (R.d.O.F); (E.B.A.J.); (F.A.A.F.G.); (G.C.V.); (M.V.S.); (J.M.B.F.)
| |
Collapse
|
45
|
Viola JM, Porter CM, Gupta A, Alibekova M, Prahl LS, Hughes AJ. Guiding Cell Network Assembly using Shape-Morphing Hydrogels. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002195. [PMID: 32578300 PMCID: PMC7950730 DOI: 10.1002/adma.202002195] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/30/2020] [Indexed: 05/11/2023]
Abstract
Forces and relative movement between cells and extracellular matrix (ECM) are crucial to the self-organization of tissues during development. However, the spatial range over which these dynamics can be controlled in engineering approaches is limited, impeding progress toward the construction of large, structurally mature tissues. Herein, shape-morphing materials called "kinomorphs" that rationally control the shape and size of multicellular networks are described. Kinomorphs are sheets of ECM that change their shape, size, and density depending on patterns of cell contractility within them. It is shown that these changes can manipulate structure-forming behaviors of epithelial cells in many spatial locations at once. Kinomorphs are built using a new photolithographic technology to pattern single cells into ECM sheets that are >10× larger than previously described. These patterns are designed to partially mimic the branch geometry of the embryonic kidney epithelial network. Origami-inspired simulations are then used to predict changes in kinomorph shapes. Last, kinomorph dynamics are shown to provide a centimeter-scale program that sets specific spatial locations in which ≈50 µm-diameter epithelial tubules form by cell coalescence and structural maturation. The kinomorphs may significantly advance organ-scale tissue construction by extending the spatial range of cell self-organization in emerging model systems such as organoids.
Collapse
Affiliation(s)
- John M Viola
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Catherine M Porter
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ananya Gupta
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mariia Alibekova
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Louis S Prahl
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Alex J Hughes
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
46
|
Zhang S, Lee JM, Ashok AA, Jung HS. Action of Actomyosin Contraction With Shh Modulation Drive Epithelial Folding in the Circumvallate Papilla. Front Physiol 2020; 11:936. [PMID: 32848868 PMCID: PMC7411262 DOI: 10.3389/fphys.2020.00936] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/13/2020] [Indexed: 11/13/2022] Open
Abstract
The mouse tongue possesses three types of gustatory papillae: large circumvallate papillae (CVP), foliate papillae (FOP) and fungiform papillae (FFP). Although CVP is the largest papilla and contain a high density of taste buds, little is known about CVP development. Their transition from placode to dome-shape is particularly ambiguous. Understanding this phase is crucial since dome-shaped morphology is essential for proper localization of the imminent nerve fibers and taste buds. Here, we report actomyosin-dependent apical and basal constriction of epithelial cells during dynamic epithelial folding. Furthermore, actomyosin-dependent basal constriction requires focal adhesion kinase to guide dome-shape formation. Sonic hedgehog (Shh) is closely associated with the differentiation or survival of the neurons in CVP ganglion and cytoskeletal alteration in trench epithelial cells which regulate CVP morphogenesis. Our results demonstrate the CVP morphogenesis mechanism from placode to dome-shape by actomyosin-dependent cell shape change and suggest roles that Shh may play in trench and stromal core formation during CVP development.
Collapse
Affiliation(s)
- Sushan Zhang
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Brain Korea 21 PLUS Project, Taste Research Center, College of Dentistry, Yonsei University, Seoul, South Korea
| | - Jong-Min Lee
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Brain Korea 21 PLUS Project, Taste Research Center, College of Dentistry, Yonsei University, Seoul, South Korea
| | - Adpaikar Anish Ashok
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Brain Korea 21 PLUS Project, Taste Research Center, College of Dentistry, Yonsei University, Seoul, South Korea
| | - Han-Sung Jung
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Brain Korea 21 PLUS Project, Taste Research Center, College of Dentistry, Yonsei University, Seoul, South Korea
| |
Collapse
|
47
|
Hebeda CB, Sandri S, Benis CM, de Paula-Silva M, Loiola RA, Reutelingsperger C, Perretti M, Farsky SHP. Annexin A1/Formyl Peptide Receptor Pathway Controls Uterine Receptivity to the Blastocyst. Cells 2020; 9:cells9051188. [PMID: 32403233 PMCID: PMC7291299 DOI: 10.3390/cells9051188] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/22/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022] Open
Abstract
Embryo implantation into the uterine wall is a highly modulated, complex process. We previously demonstrated that Annexin A1 (AnxA1), which is a protein secreted by epithelial and inflammatory cells in the uterine microenvironment, controls embryo implantation in vivo. Here, we decipher the effects of recombinant AnxA1 in this phenomenon by using human trophoblast cell (BeWo) spheroids and uterine epithelial cells (Ishikawa; IK). AnxA1-treated IK cells demonstrated greater levels of spheroid adherence and upregulation of the tight junction molecules claudin-1 and zona occludens-1, as well as the glycoprotein mucin-1 (Muc-1). The latter effect of AnxA1 was not mediated through IL-6 secreted from IK cells, a known inducer of Muc-1 expression. Rather, these effects of AnxA1 involved activation of the formyl peptide receptors FPR1 and FPR2, as pharmacological blockade of FPR1 or FPR1/FPR2 abrogated such responses. The downstream actions of AnxA1 were mediated through the ERK1/2 phosphorylation pathway and F-actin polymerization in IK cells, as blockade of ERK1/2 phosphorylation reversed AnxA1-induced Muc-1 and claudin-1 expression. Moreover, FPR2 activation by AnxA1 induced vascular endothelial growth factor (VEGF) secretion by IK cells, and the supernatant of AnxA1-treated IK cells evoked angiogenesis in vitro. In conclusion, these data highlight the role of the AnxA1/FPR1/FPR2 pathway in uterine epithelial control of blastocyst implantation.
Collapse
Affiliation(s)
- Cristina B. Hebeda
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo CEP 05508-000, Brazil; (C.B.H.); (S.S.); (C.M.B.); (M.d.P.-S.); (R.A.L.)
| | - Silvana Sandri
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo CEP 05508-000, Brazil; (C.B.H.); (S.S.); (C.M.B.); (M.d.P.-S.); (R.A.L.)
| | - Cláudia M. Benis
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo CEP 05508-000, Brazil; (C.B.H.); (S.S.); (C.M.B.); (M.d.P.-S.); (R.A.L.)
| | - Marina de Paula-Silva
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo CEP 05508-000, Brazil; (C.B.H.); (S.S.); (C.M.B.); (M.d.P.-S.); (R.A.L.)
| | - Rodrigo A. Loiola
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo CEP 05508-000, Brazil; (C.B.H.); (S.S.); (C.M.B.); (M.d.P.-S.); (R.A.L.)
| | - Chris Reutelingsperger
- Faculty of Health, Medicine and Life Sciences, Part of Maastricht University Medical Center, Part of Maastricht University, 6211 LK Maastricht, The Netherlands;
| | - Mauro Perretti
- The William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK;
| | - Sandra H. P. Farsky
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo CEP 05508-000, Brazil; (C.B.H.); (S.S.); (C.M.B.); (M.d.P.-S.); (R.A.L.)
- Correspondence: ; Tel.: +55-(11)-3091-2197
| |
Collapse
|
48
|
Pradhan-Sundd T, Kosar K, Saggi H, Zhang R, Vats R, Cornuet P, Green S, Singh S, Zeng G, Sundd P, Nejak-Bowen K. Wnt/β-Catenin Signaling Plays a Protective Role in the Mdr2 Knockout Murine Model of Cholestatic Liver Disease. Hepatology 2020; 71:1732-1749. [PMID: 31489648 PMCID: PMC7058521 DOI: 10.1002/hep.30927] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 08/15/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIMS The Wnt/β-catenin signaling pathway has a well-described role in liver pathobiology. Its suppression was recently shown to decrease bile acid (BA) synthesis, thus preventing the development of cholestatic liver injury and fibrosis after bile duct ligation (BDL). APPROACH AND RESULTS To generalize these observations, we suppressed β-catenin in Mdr2 knockout (KO) mice, which develop sclerosing cholangitis due to regurgitation of BA from leaky ducts. When β-catenin was knocked down (KD) in KO for 2 weeks, hepatic and biliary injury were exacerbated in comparison to KO given placebo, as shown by serum biochemistry, ductular reaction, inflammation, and fibrosis. Simultaneously, KO/KD livers displayed increased oxidative stress and senescence and an impaired regenerative response. Although the total liver BA levels were similar between KO/KD and KO, there was significant dysregulation of BA transporters and BA detoxification/synthesis enzymes in KO/KD compared with KO alone. Multiphoton intravital microscopy revealed a mixing of blood and bile in the sinusoids, and validated the presence of increased serum BA in KO/KD mice. Although hepatocyte junctions were intact, KO/KD livers had significant canalicular defects, which resulted from loss of hepatocyte polarity. Thus, in contrast to the protective effect of β-catenin KD in BDL model, β-catenin KD in Mdr2 KO aggravated rather than alleviated injury by interfering with expression of BA transporters, hepatocyte polarity, canalicular structure, and the regenerative response. CONCLUSIONS The resulting imbalance between ongoing injury and restitution led to worsening of the Mdr2 KO phenotype, suggesting caution in targeting β-catenin globally for all cholestatic conditions.
Collapse
Affiliation(s)
| | - Karis Kosar
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - Harvinder Saggi
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - Rong Zhang
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - Ravi Vats
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA
| | - Pamela Cornuet
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | | | - Sucha Singh
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - Gang Zeng
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - Prithu Sundd
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA
| | - Kari Nejak-Bowen
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
49
|
Liu Y, Zhang Y, Li T, Han J, Wang Y. The tight junction protein TJP1 regulates the feeding-modulated hepatic circadian clock. Nat Commun 2020; 11:589. [PMID: 32001717 PMCID: PMC6992704 DOI: 10.1038/s41467-020-14470-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/10/2020] [Indexed: 01/05/2023] Open
Abstract
Circadian clocks in the suprachiasmatic nucleus and peripheral tissues orchestrate behavioral and physiological activities of mammals in response to environmental cues. In the liver, the circadian clock is also modulated by feeding. However, the molecular mechanisms involved are unclear. Here, we show that TJP1 (tight junction protein 1) functions as a mediator of mTOR (mechanistic target of rapamycin) to modulate the hepatic circadian clock. TJP1 interacts with PER1 (period circadian regulator 1) and prevents its nuclear translocation. During feeding, mTOR phosphorylates TJP1 and attenuates its association with PER1, thereby enhancing nuclear shuttling of PER1 to dampen circadian oscillation. Therefore, our results provide a previously uncharacterized mechanistic insight into how feeding modulates the hepatic circadian clock. The circadian clock regulates rhythms of physiology and metabolism in response to environmental cues such as food intake. Here, the authors show that tight junction protein 1 (TJP1) interacts with period 1 and modulates its nuclear translocation in a mTOR-dependent manner.
Collapse
Affiliation(s)
- Yi Liu
- MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuanyuan Zhang
- MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Tong Li
- MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jinbo Han
- MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yiguo Wang
- MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
50
|
Singh AP, Mia MB, Saxena RK. Acid-functionalized single-walled carbon nanotubes alter epithelial tight junctions and enhance paracellular permeability. J Biosci 2020. [DOI: 10.1007/s12038-020-9989-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|