1
|
Bretová K, Svobodová V, Dubový P. Changes in Cx43 and AQP4 Proteins, and the Capture of 3 kDa Dextran in Subpial Astrocytes of the Rat Medial Prefrontal Cortex after Both Sham Surgery and Sciatic Nerve Injury. Int J Mol Sci 2024; 25:10989. [PMID: 39456773 PMCID: PMC11507206 DOI: 10.3390/ijms252010989] [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: 09/17/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
A subpopulation of astrocytes on the brain's surface, known as subpial astrocytes, constitutes the "glia limitans superficialis" (GLS), which is an interface between the brain parenchyma and the cerebrospinal fluid (CSF) in the subpial space. Changes in connexin-43 (Cx43) and aquaporin-4 (AQP4) proteins in subpial astrocytes were examined in the medial prefrontal cortex at postoperative day 1, 3, 7, 14, and 21 after sham operation and sciatic nerve compression (SNC). In addition, we tested the altered uptake of TRITC-conjugated 3 kDa dextran by reactive subpial astrocytes. Cellular immunofluorescence (IF) detection and image analysis were used to examine changes in Cx43 and AQP4 protein levels, as well as TRITC-conjugated 3 kDa dextran, in subpial astrocytes. The intensity of Cx43-IF was significantly increased, but AQP4-IF decreased in subpial astrocytes of sham- and SNC-operated rats during all survival periods compared to naïve controls. Similarly, the uptake of 3 kDa dextran in the GLS was reduced following both sham and SNC operations. The results suggest that both sciatic nerve injury and peripheral tissue injury alone can induce changes in subpial astrocytes related to the spread of their reactivity across the cortical surface mediated by increased amounts of gap junctions. At the same time, water transport and solute uptake were impaired in subpial astrocytes.
Collapse
Affiliation(s)
| | | | - Petr Dubový
- Cellular and Molecular Neurobiology Research Group, Department of Anatomy, Faculty of Medicine, Masaryk University, CZ-62500 Brno, Czech Republic; (K.B.)
| |
Collapse
|
2
|
Shamul JG, Wang Z, Gong H, Ou W, White AM, Moniz-Garcia DP, Gu S, Clyne AM, Quiñones-Hinojosa A, He X. Meta-analysis of the make-up and properties of in vitro models of the healthy and diseased blood-brain barrier. Nat Biomed Eng 2024:10.1038/s41551-024-01250-2. [PMID: 39304761 DOI: 10.1038/s41551-024-01250-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/08/2024] [Indexed: 09/22/2024]
Abstract
In vitro models of the human blood-brain barrier (BBB) are increasingly used to develop therapeutics that can cross the BBB for treating diseases of the central nervous system. Here we report a meta-analysis of the make-up and properties of transwell and microfluidic models of the healthy BBB and of BBBs in glioblastoma, Alzheimer's disease, Parkinson's disease and inflammatory diseases. We found that the type of model, the culture method (static or dynamic), the cell types and cell ratios, and the biomaterials employed as extracellular matrix are all crucial to recapitulate the low permeability and high expression of tight-junction proteins of the BBB, and to obtain high trans-endothelial electrical resistance. Specifically, for models of the healthy BBB, the inclusion of endothelial cells and pericytes as well as physiological shear stresses (~10-20 dyne cm-2) are necessary, and when astrocytes are added, astrocytes or pericytes should outnumber endothelial cells. We expect this meta-analysis to facilitate the design of increasingly physiological models of the BBB.
Collapse
Affiliation(s)
- James G Shamul
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- RNA Mediated Gene Regulation Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Zhiyuan Wang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Hyeyeon Gong
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Wenquan Ou
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Alisa M White
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | | | - Shuo Gu
- RNA Mediated Gene Regulation Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Alisa Morss Clyne
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA
- Brain and Behavior Institute, University of Maryland, College Park, MD, USA
| | | | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA.
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA.
- Brain and Behavior Institute, University of Maryland, College Park, MD, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
3
|
Zanatta D, Betanzos A, Azuara-Liceaga E, Montaño S, Orozco E. Entamoeba histolytica: EhADH, an Alix Protein, Participates in Several Virulence Events through Its Different Domains. Int J Mol Sci 2024; 25:7609. [PMID: 39062867 PMCID: PMC11277477 DOI: 10.3390/ijms25147609] [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: 06/03/2024] [Revised: 07/02/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Entamoeba histolytica is the protozoan causative of human amoebiasis. The EhADH adhesin (687 aa) is a protein involved in tissue invasion, phagocytosis and host-cell lysis. EhADH adheres to the prey and follows its arrival to the multivesicular bodies. It is an accessory protein of the endosomal sorting complexes required for transport (ESCRT) machinery. Here, to study the role of different parts of EhADH during virulence events, we produced trophozoites overexpressing the three domains of EhADH, Bro1 (1-400 aa), Linker (246-446 aa) and Adh (444-687 aa) to evaluate their role in virulence. The TrophozBro11-400 slightly increased adherence and phagocytosis, but these trophozoites showed a higher ability to destroy cell monolayers, augment the permeability of cultured epithelial cells and mouse colon, and produce more damage to hamster livers. The TrophozLinker226-446 also increased the virulence properties, but with lower effect than the TrophozBro11-400. In addition, this fragment participates in cholesterol transport and GTPase binding. Interestingly, the TrophozAdh444-687 produced the highest effect on adherence and phagocytosis, but it poorly influenced the monolayers destruction; nevertheless, they augmented the colon and liver damage. To identify the protein partners of each domain, we used recombinant peptides. Pull-down assays and mass spectrometry showed that Bro1 domain interplays with EhADH, Gal/GalNAc lectin, EhCPs, ESCRT machinery components and cytoskeleton proteins. While EhADH, ubiquitin, EhRabB, EhNPC1 and EhHSP70 were associated to the Linker domain, and EhADH, EhHSP70, EhPrx and metabolic enzymes interacted to the Adh domain. The diverse protein association confirms that EhADH is a versatile molecule with multiple functions probably given by its capacity to form distinct molecular complexes.
Collapse
Affiliation(s)
- Dxinegueela Zanatta
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies of National Polytechnic Institute, Mexico City 07360, Mexico;
| | - Abigail Betanzos
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies of National Polytechnic Institute, Mexico City 07360, Mexico;
| | - Elisa Azuara-Liceaga
- Postgraduate in Genomic Sciences, Autonomous University of Mexico City, Mexico City 03100, Mexico;
| | - Sarita Montaño
- Laboratory of Bioinformatics and Molecular Simulation, Faculty of Biological Chemistry Sciences, Autonomous University of Sinaloa, Sinaloa 80030, Mexico;
| | - Esther Orozco
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies of National Polytechnic Institute, Mexico City 07360, Mexico;
| |
Collapse
|
4
|
Schimetz J, Shah P, Keese C, Dehnert C, Detweiler M, Michael S, Toniatti-Yanulavich C, Xu X, Padilha EC. Automated measurement of transepithelial electrical resistance (TEER) in 96-well transwells using ECIS TEER96: Single and multiple time point assessments. SLAS Technol 2024; 29:100116. [PMID: 37923083 DOI: 10.1016/j.slast.2023.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Transepithelial electrical resistance (TEER) is a widely used technique for quantifying the permeability of epithelial and endothelial cell layers. However, traditional methods of measuring TEER are limited to single timepoint measurements and can subject cells to an altered environment during the measurement. Here, we assessed the validity of TEER measurements by the ECIS TEER96 device, which is designed to take continuous TEER measurements of a cell culture system in a standard laboratory incubator. We found that the instrument accurately measures TEER across TEER values ranging from 10 to 2050 Ω*cm2 and is more accurate than the manual epithelial voltohmmeter electrode at high TEER values. Furthermore, the high-resolution measurements provided by the device allowed for a unique insight into the mechanisms and kinetics of cells in vitro. To demonstrate the continuous measurement capability of the device, we tracked the formation of an MDCKI cell monolayer until TEER plateaued. Furthermore, we treated Caco-2 monolayers with different concentrations of DMSO and the antimicrobial and surfactant compound benzethonium chloride to measure disruptions to barrier integrity. Treatment of both compounds resulted in concentration-dependent loss of barrier integrity. Our results suggest that the ECIS TEER96 device is a reliable and convenient option for measuring TEER in cell cultures and can provide valuable insights into the behavior of cells in vitro. This technology will be especially useful for increasing throughput of drug permeability assays, inflammation studies, and gaining better understanding of disease states in a cell culture system.
Collapse
Affiliation(s)
- Jacob Schimetz
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Dr, Rockville, MD 20850
| | - Pranav Shah
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Dr, Rockville, MD 20850
| | | | | | | | - Sam Michael
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Dr, Rockville, MD 20850
| | | | - Xin Xu
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Dr, Rockville, MD 20850
| | - Elias C Padilha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Dr, Rockville, MD 20850.
| |
Collapse
|
5
|
Kamakura S, Hayase J, Kohda A, Iwakiri Y, Chishiki K, Izaki T, Sumimoto H. TMEM25 is a Par3-binding protein that attenuates claudin assembly during tight junction development. EMBO Rep 2024; 25:144-167. [PMID: 38177906 PMCID: PMC10897455 DOI: 10.1038/s44319-023-00018-0] [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: 01/10/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 01/06/2024] Open
Abstract
The tight junction (TJ) in epithelial cells is formed by integral membrane proteins and cytoplasmic scaffolding proteins. The former contains the claudin family proteins with four transmembrane segments, while the latter includes Par3, a PDZ domain-containing adaptor that organizes TJ formation. Here we show the single membrane-spanning protein TMEM25 localizes to TJs in epithelial cells and binds to Par3 via a PDZ-mediated interaction with its C-terminal cytoplasmic tail. TJ development during epithelial cell polarization is accelerated by depletion of TMEM25, and delayed by overexpression of TMEM25 but not by that of a C-terminally deleted protein, indicating a regulatory role of TMEM25. TMEM25 associates via its N-terminal extracellular domain with claudin-1 and claudin-2 to suppress their cis- and trans-oligomerizations, both of which participate in TJ strand formation. Furthermore, Par3 attenuates TMEM25-claudin association via binding to TMEM25, implying its ability to affect claudin oligomerization. Thus, the TJ protein TMEM25 appears to negatively regulate claudin assembly in TJ formation, which regulation is modulated by its interaction with Par3.
Collapse
Affiliation(s)
- Sachiko Kamakura
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Junya Hayase
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Akira Kohda
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yuko Iwakiri
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kanako Chishiki
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Tomoko Izaki
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Hideki Sumimoto
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan.
| |
Collapse
|
6
|
Deniz Derman I, Yeo M, Castaneda DC, Callender M, Horvath M, Mo Z, Xiong R, Fleming E, Chen P, Peeples ME, Palucka K, Oh J, Ozbolat IT. High-throughput bioprinting of the nasal epithelium using patient-derived nasal epithelial cells. Biofabrication 2023; 15:044103. [PMID: 37536321 PMCID: PMC10424246 DOI: 10.1088/1758-5090/aced23] [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: 02/02/2023] [Revised: 07/20/2023] [Accepted: 08/03/2023] [Indexed: 08/05/2023]
Abstract
Progenitor human nasal epithelial cells (hNECs) are an essential cell source for the reconstruction of the respiratory pseudostratified columnar epithelium composed of multiple cell types in the context of infection studies and disease modeling. Hitherto, manual seeding has been the dominant method for creating nasal epithelial tissue models through biofabrication. However, this approach has limitations in terms of achieving the intricate three-dimensional (3D) structure of the natural nasal epithelium. 3D bioprinting has been utilized to reconstruct various epithelial tissue models, such as cutaneous, intestinal, alveolar, and bronchial epithelium, but there has been no attempt to use of 3D bioprinting technologies for reconstruction of the nasal epithelium. In this study, for the first time, we demonstrate the reconstruction of the nasal epithelium with the use of primary hNECs deposited on Transwell inserts via droplet-based bioprinting (DBB), which enabled high-throughput fabrication of the nasal epithelium in Transwell inserts of 24-well plates. DBB of progenitor hNECs ranging from one-tenth to one-half of the cell seeding density employed during the conventional cell seeding approach enabled a high degree of differentiation with the presence of cilia and tight-junctions over a 4 weeks air-liquid interface culture. Single cell RNA sequencing of these cultures identified five major epithelial cells populations, including basal, suprabasal, goblet, club, and ciliated cells. These cultures recapitulated the pseudostratified columnar epithelial architecture present in the native nasal epithelium and were permissive to respiratory virus infection. These results denote the potential of 3D bioprinting for high-throughput fabrication of nasal epithelial tissue models not only for infection studies but also for other purposes, such as disease modeling, immunological studies, and drug screening.
Collapse
Affiliation(s)
- I Deniz Derman
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, United States of America
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, United States of America
| | - Miji Yeo
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, United States of America
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, United States of America
| | | | - Megan Callender
- The Jackson Laboratory, Farmington, CT 06032, United States of America
| | - Mian Horvath
- The Jackson Laboratory, Farmington, CT 06032, United States of America
| | - Zengshuo Mo
- The Jackson Laboratory, Farmington, CT 06032, United States of America
| | - Ruoyun Xiong
- The Jackson Laboratory, Farmington, CT 06032, United States of America
| | - Elizabeth Fleming
- The Jackson Laboratory, Farmington, CT 06032, United States of America
| | - Phylip Chen
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, United States of America
| | - Mark E Peeples
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, United States of America
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH 43210, United States of America
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, United States of America
| | - Karolina Palucka
- The Jackson Laboratory, Farmington, CT 06032, United States of America
| | - Julia Oh
- The Jackson Laboratory, Farmington, CT 06032, United States of America
| | - Ibrahim T Ozbolat
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, United States of America
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, United States of America
- Biomedical Engineering Department, Penn State University, University Park, PA 16802, United States of America
- Materials Research Institute, Penn State University, University Park, PA 16802, United States of America
- Cancer Institute, Penn State University, University Park, PA 16802, United States of America
- Neurosurgery Department, Penn State University, University Park, PA 16802, United States of America
- Department of Medical Oncology, Cukurova University, Adana, Turkey
- Biotechnology Research and Application Center, Cukurova University, Adana, Turkey
| |
Collapse
|
7
|
Higashi T, Stephenson RE, Schwayer C, Huljev K, Higashi AY, Heisenberg CP, Chiba H, Miller AL. ZnUMBA - a live imaging method to detect local barrier breaches. J Cell Sci 2023; 136:jcs260668. [PMID: 37461809 PMCID: PMC10445723 DOI: 10.1242/jcs.260668] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 07/06/2023] [Indexed: 08/09/2023] Open
Abstract
Epithelial barrier function is commonly analyzed using transepithelial electrical resistance, which measures ion flux across a monolayer, or by adding traceable macromolecules and monitoring their passage across the monolayer. Although these methods measure changes in global barrier function, they lack the sensitivity needed to detect local or transient barrier breaches, and they do not reveal the location of barrier leaks. Therefore, we previously developed a method that we named the zinc-based ultrasensitive microscopic barrier assay (ZnUMBA), which overcomes these limitations, allowing for detection of local tight junction leaks with high spatiotemporal resolution. Here, we present expanded applications for ZnUMBA. ZnUMBA can be used in Xenopus embryos to measure the dynamics of barrier restoration and actin accumulation following laser injury. ZnUMBA can also be effectively utilized in developing zebrafish embryos as well as cultured monolayers of Madin-Darby canine kidney (MDCK) II epithelial cells. ZnUMBA is a powerful and flexible method that, with minimal optimization, can be applied to multiple systems to measure dynamic changes in barrier function with spatiotemporal precision.
Collapse
Affiliation(s)
- Tomohito Higashi
- Department of Basic Pathology, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Rachel E. Stephenson
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Cornelia Schwayer
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Karla Huljev
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria
| | - Atsuko Y. Higashi
- Department of Basic Pathology, Fukushima Medical University, Fukushima 960-1295, Japan
- Department of Nephrology and Hypertension, Fukushima Medical University, Fukushima 960-1295, Japan
| | | | - Hideki Chiba
- Department of Basic Pathology, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Ann L. Miller
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
8
|
Min J, Yang S, Cai Y, Vanderwall DR, Wu Z, Li S, Liu S, Liu B, Wang J, Ding Y, Chen J, Jiang C, Wren JD, Csiszar A, Ungvari Z, Greco C, Kanie T, Peng J, Zhang XA. Tetraspanin Tspan8 restrains interferon signaling to stabilize intestinal epithelium by directing endocytosis of interferon receptor. Cell Mol Life Sci 2023; 80:154. [PMID: 37204469 PMCID: PMC10484302 DOI: 10.1007/s00018-023-04803-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/25/2023] [Accepted: 05/08/2023] [Indexed: 05/20/2023]
Abstract
Inflammation can impair intestinal barrier, while increased epithelial permeability can lead to inflammation. In this study, we found that the expression of Tspan8, a tetraspanin expressed specifically in epithelial cells, is downregulated in mouse model of ulcerative disease (UC) but correlated with those of cell-cell junction components, such as claudins and E-cadherin, suggesting that Tspan8 supports intestinal epithelial barrier. Tspan8 removal increases intestinal epithelial permeability and upregulates IFN-γ-Stat1 signaling. We also demonstrated that Tspan8 coalesces with lipid rafts and facilitates IFNγ-R1 localization at or near lipid rafts. As IFN-γ induces its receptor undergoing clathrin- or lipid raft-dependent endocytosis and IFN-γR endocytosis plays an important role in Jak-Stat1 signaling, our analysis on IFN-γR endocytosis revealed that Tspan8 silencing impairs lipid raft-mediated but promotes clathrin-mediated endocytosis of IFN-γR1, leading to increased Stat1 signaling. These changes in IFN-γR1 endocytosis upon Tspan8 silencing correlates with fewer lipid raft component GM1 at the cell surface and more clathrin heavy chain in the cells. Our findings indicate that Tspan8 determines the IFN-γR1 endocytosis route, to restrain Stat1 signaling, stabilize intestine epithelium, and subsequently prevent intestine from inflammation. Our finding also implies that Tspan8 is needed for proper endocytosis through lipid rafts.
Collapse
Affiliation(s)
- Jiang Min
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Shenglan Yang
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Yang Cai
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - David R Vanderwall
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Zhiping Wu
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Shuping Li
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Songlan Liu
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Beibei Liu
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Jie Wang
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Yingjun Ding
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Junxiong Chen
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Chao Jiang
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | | | - Anna Csiszar
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Zoltan Ungvari
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Céline Greco
- Department of Pain and Palliative Care Unit, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Tomoharu Kanie
- University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Xin A Zhang
- University of Oklahoma Health Sciences Center, Oklahoma City, USA.
| |
Collapse
|
9
|
Łach A, Wnuk A, Wójtowicz AK. Experimental Models to Study the Functions of the Blood-Brain Barrier. Bioengineering (Basel) 2023; 10:bioengineering10050519. [PMID: 37237588 DOI: 10.3390/bioengineering10050519] [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: 02/23/2023] [Revised: 04/07/2023] [Accepted: 04/22/2023] [Indexed: 05/28/2023] Open
Abstract
The purpose of this paper was to discuss the achievements of in vitro modeling in terms of the blood-brain barrier [BBB] and to create a clear overview of this research area, which is useful in research planning. The text was divided into three main parts. The first part describes the BBB as a functional structure, its constitution, cellular and noncellular components, mechanisms of functioning and importance for the central nervous system, in terms of both protection and nourishment. The second part is an overview of parameters important in terms of establishing and maintaining a barrier phenotype that allows for formulating criteria of evaluation of the BBB in vitro models. The third and last part discusses certain techniques for developing the BBB in vitro models. It describes subsequent research approaches and models, as they underwent change alongside technological advancement. On the one hand, we discuss possibilities and limitations of different research approaches: primary cultures vs. cell lines and monocultures vs. multicultures. On the other hand, we review advantages and disadvantages of specific models, such as models-on-a-chip, 3D models or microfluidic models. We not only attempt to state the usefulness of specific models in different kinds of research on the BBB but also emphasize the significance of this area of research for advancement of neuroscience and the pharmaceutical industry.
Collapse
Affiliation(s)
- Andrzej Łach
- Laboratory of Neuropharmacology and Epigenetics, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland
- Department of Nutrition, Animal Biotechnology and Fisheries, Faculty of Animal Sciences, University of Agriculture, 30-059 Kraków, Poland
| | - Agnieszka Wnuk
- Laboratory of Neuropharmacology and Epigenetics, Department of Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland
| | - Anna Katarzyna Wójtowicz
- Department of Nutrition, Animal Biotechnology and Fisheries, Faculty of Animal Sciences, University of Agriculture, 30-059 Kraków, Poland
| |
Collapse
|
10
|
Wang M, Wang L, Gong Y, Li Q, Shao Y, Li X. Study on the Effects of Different anti-VEGF drugs on Fibrovascular Membranes of Proliferative Diabetic Retinopathy. Photodiagnosis Photodyn Ther 2023; 42:103530. [PMID: 37060987 DOI: 10.1016/j.pdpdt.2023.103530] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/21/2023] [Accepted: 03/21/2023] [Indexed: 04/17/2023]
Abstract
PURPOSE To investigate the effects of different anti-VEGF drugs on fibrovascular membranes (FVM) in proliferative diabetic retinopathy (PDR). In addition, in vitro model was used to simulate the intraocular fibroblasts barrier to explore the penetration of different anti-VEGF drugs. METHODS 24 eyes from 24 PDR patients with FVM were recruited for this prospective observational study. The patients were randomized to receive one of three anti-VEGF drugs (Ranibizumab, Conbercept, or Aflibercept). Then neovascular structures were assessed by optical coherence tomography angiography (OCTA) before intravitreal injection (pre-IVT) and 1, 2, and 3 days after intravitreal injection (post-IVT). The changes in vessels area (VSA), vessels percentage area (VPA), junction density (JD), and average lacunarity (AL) were analyzed by using the image processing software Angiotool. In vitro penetrating model with fibroblasts barrier was used to compare the effects of the three drugs on human retinal vascular endothelial cells (HRVECs) over 3 days by Cell proliferation measurement. Moreover, the drug concentrations in the penetrating model were detected by liquid chromatography-mass spectrometry (LC-MS). RESULTS The VSA, VPA, and JD all decreased, while the AL increased in Ranibizumab group(n=8), Conbercept group (n=8), and Aflibercept group (n=8) within 3 days (P<0.05). Meanwhile, under the condition of the same amount of substance, the inhibition effect of Ranibizumab on HRVEC was the strongest in the penetrating model evaluated by CCK8 absorbance experiments of HRVECs (FCCK8=6.493, PCCK8= 0.0051), and the number of transmembrane molecules in the Ranibizumab group was also the largest within 3 days (F=8.209, P=0.0006) among the three groups. CONCLUSION Angiotool is feasible to reconstruct the neovascular structure on the FVM in OCTA images. The three different anti-VEGF drugs can significantly reduce the vascular area and density on the proliferating membranes, and there is no significant difference in the anti-neovascularization among the three drugs clinically. However, small molecule drug is more penetrating and move faster across membranes in vitro cell model. CLINICAL TRIAL REGISTRATION This trial is registered with the Chinese Clinical Trial Registry (http://www.chictr.org.cn, registration number ChiCTR2300067476).
Collapse
Affiliation(s)
- Manqiao Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin 300384, China; Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin 300384, China; Eye Institute and School of Optometry, Tianjin 300384, China; Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Linni Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin 300384, China; Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin 300384, China; Eye Institute and School of Optometry, Tianjin 300384, China; Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Yi Gong
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin 300384, China; Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin 300384, China; Eye Institute and School of Optometry, Tianjin 300384, China; Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Qingbo Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin 300384, China; Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin 300384, China; Eye Institute and School of Optometry, Tianjin 300384, China; Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Yan Shao
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin 300384, China; Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin 300384, China; Eye Institute and School of Optometry, Tianjin 300384, China; Tianjin Medical University Eye Hospital, Tianjin 300384, China.
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin 300384, China; Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin 300384, China; Eye Institute and School of Optometry, Tianjin 300384, China; Tianjin Medical University Eye Hospital, Tianjin 300384, China.
| |
Collapse
|
11
|
Derman ID, Yeo M, Castaneda DC, Callender M, Horvath M, Mo Z, Xiong R, Fleming E, Chen P, Peeples ME, Palucka K, Oh J, Ozbolat IT. High-Throughput Bioprinting of the Nasal Epithelium using Patient-derived Nasal Epithelial Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.29.534723. [PMID: 37034627 PMCID: PMC10081172 DOI: 10.1101/2023.03.29.534723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Human nasal epithelial cells (hNECs) are an essential cell source for the reconstruction of the respiratory pseudostratified columnar epithelium composed of multiple cell types in the context of infection studies and disease modeling. Hitherto, manual seeding has been the dominant method for creating nasal epithelial tissue models. However, the manual approach is slow, low-throughput and has limitations in terms of achieving the intricate 3D structure of the natural nasal epithelium in a uniform manner. 3D Bioprinting has been utilized to reconstruct various epithelial tissue models, such as cutaneous, intestinal, alveolar, and bronchial epithelium, but there has been no attempt to use of 3D bioprinting technologies for reconstruction of the nasal epithelium. In this study, for the first time, we demonstrate the reconstruction of the nasal epithelium with the use of primary hNECs deposited on Transwell inserts via droplet-based bioprinting (DBB), which enabled high-throughput fabrication of the nasal epithelium in Transwell inserts of 24-well plates. DBB of nasal progenitor cells ranging from one-tenth to one-half of the cell seeding density employed during the conventional cell seeding approach enabled a high degree of differentiation with the presence of cilia and tight-junctions over a 4-week air-liquid interface culture. Single cell RNA sequencing of these cultures identified five major epithelial cells populations, including basal, suprabasal, goblet, club, and ciliated cells. These cultures recapitulated the pseudostratified columnar epithelial architecture present in the native nasal epithelium and were permissive to respiratory virus infection. These results denote the potential of 3D bioprinting for high-throughput fabrication of nasal epithelial tissue models not only for infection studies but also for other purposes such as disease modeling, immunological studies, and drug screening.
Collapse
|
12
|
Nguyen HLT, Peng G, Trujillo-Paez JV, Yue H, Ikutama R, Takahashi M, Umehara Y, Okumura K, Ogawa H, Ikeda S, Niyonsaba F. The Antimicrobial Peptide AMP-IBP5 Suppresses Dermatitis-like Lesions in a Mouse Model of Atopic Dermatitis through the Low-Density Lipoprotein Receptor-Related Protein-1 Receptor. Int J Mol Sci 2023; 24:ijms24065200. [PMID: 36982275 PMCID: PMC10049508 DOI: 10.3390/ijms24065200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 03/07/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
The antimicrobial peptide derived from insulin-like growth factor-binding protein 5 (AMP-IBP5) exhibits antimicrobial activities and immunomodulatory functions in keratinocytes and fibroblasts. However, its role in regulating skin barrier function remains unclear. Here, we investigated the effects of AMP-IBP5 on the skin barrier and its role in the pathogenesis of atopic dermatitis (AD). 2,4-Dinitrochlorobenzene was used to induce AD-like skin inflammation. Transepithelial electrical resistance and permeability assays were used to investigate tight junction (TJ) barrier function in normal human epidermal keratinocytes and mice. AMP-IBP5 increased the expression of TJ-related proteins and their distribution along the intercellular borders. AMP-IBP5 also improved TJ barrier function through activation of the atypical protein kinase C and Rac1 pathways. In AD mice, AMP-IBP5 ameliorated dermatitis-like symptoms restored the expression of TJ-related proteins, suppressed the expression of inflammatory and pruritic cytokines, and improved skin barrier function. Interestingly, the ability of AMP-IBP5 to alleviate inflammation and improve skin barrier function in AD mice was abolished in mice treated with an antagonist of the low-density lipoprotein receptor-related protein-1 (LRP1) receptor. Collectively, these findings indicate that AMP-IBP5 may ameliorate AD-like inflammation and enhance skin barrier function through LRP1, suggesting a possible role for AMP-IBP5 in the treatment of AD.
Collapse
Affiliation(s)
- Hai Le Thanh Nguyen
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Ge Peng
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Juan Valentin Trujillo-Paez
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Hainan Yue
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Risa Ikutama
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Miho Takahashi
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yoshie Umehara
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Ko Okumura
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Hideoki Ogawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Shigaku Ikeda
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Dermatology and Allergology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - François Niyonsaba
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
- Faculty of International Liberal Arts, Juntendo University, Bunkyo-ku, Tokyo 113-8421, Japan
- Correspondence: ; Tel.: +81-3-5802-1591; Fax: +81-3-3813-5512
| |
Collapse
|
13
|
Gulsun T, Izat N, Sahin S. Influence of permeability enhancers on the paracellular permeability of metformin hydrochloride and furosemide across Caco-2 cells. Can J Physiol Pharmacol 2022; 101:185-199. [PMID: 36459686 DOI: 10.1139/cjpp-2022-0265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Permeability enhancers can affect absorption of paracellularly transported drugs. This study aims to evaluate effects of permeability enhancers (chitosan, methyl-β -cyclodextrin, sodium caprate, sodium lauryl sulfate, etc.) on the permeability of paracellularly absorbed furosemide and metformin hydrochloride. Methyl thiazole tetrazolium bromide test was carried out to determine the drug concentrations in permeability study. Trans-epithelial electrical resistance (TEER) values determined to assess the integrity of tight junctions. Permeability enhancers were applied at different concentrations alone, in dual/triple combinations. Permeability was determined using human colorectal adenocarcinoma (Caco-2) cells (TEER > 400 Ω·cm2). Permeability enhancers have no significant effect (<2-fold; p > 0.05) on the permeability of furosemide (1.80 × 10-5 ± 4.55 × 10-7 cm/s); however, metformin permeability (1.36 × 10-5 ± 1.25 × 10-6 cm/s) increased significantly (p < 0.05) with 0.3% and 0.5% (w/v) chitosan (2.0- and 2.7-fold, respectively), 1% methyl-β -cyclodextrin (w/v) (3.5-fold), 10 and 20 µmol/L sodium caprate (2.2- and 2.8-fold, respectively), and 0.012% sodium lauryl sulfate (w/v) (1.9-fold). Furosemide permeability increased significantly (p < 0.05) with chitosan-sodium lauryl sulfate combination (1.7-fold), and all triple combinations (1.4- to 1.9-fold). Chitosan containing dual/triple combinations resulted in significant increase (p < 0.05) in metformin permeability (1.7 to 2.8-fold). All results indicated that absorption of furosemide and metformin can be improved by the combination of permeability enhancers. Therefore, it can be evaluated for the formulation of development strategies containing furosemide and metformin by the pharmaceutical industry.
Collapse
Affiliation(s)
- Tugba Gulsun
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Turkey
| | - Nihan Izat
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Turkey
| | - Selma Sahin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Turkey
| |
Collapse
|
14
|
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: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
15
|
Garimano N, Scalise ML, Gómez F, Amaral MM, Ibarra C. Intestinal mucus-derived metabolites modulate virulence of a clade 8 enterohemorrhagic Escherichia coli O157:H7. Front Cell Infect Microbiol 2022; 12:975173. [PMID: 36004327 PMCID: PMC9393340 DOI: 10.3389/fcimb.2022.975173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 07/20/2022] [Indexed: 11/24/2022] Open
Abstract
The human colonic mucus is mainly composed of mucins, which are highly glycosylated proteins. The normal commensal colonic microbiota has mucolytic activity and is capable of releasing the monosaccharides contained in mucins, which can then be used as carbon sources by pathogens such as Enterohemorrhagic Escherichia coli (EHEC). EHEC can regulate the expression of some of its virulence factors through environmental sensing of mucus-derived sugars, but its implications regarding its main virulence factor, Shiga toxin type 2 (Stx2), among others, remain unknown. In the present work, we have studied the effects of five of the most abundant mucolytic activity-derived sugars, Fucose (L-Fucose), Galactose (D-Galactose), N-Gal (N-acetyl-galactosamine), NANA (N-Acetyl-Neuraminic Acid) and NAG (N-Acetyl-D-Glucosamine) on EHEC growth, adhesion to epithelial colonic cells (HCT-8), and Stx2 production and translocation across a polarized HCT-8 monolayer. We found that bacterial growth was maximum when using NAG and NANA compared to Galactose, Fucose or N-Gal, and that EHEC adhesion was inhibited regardless of the metabolite used. On the other hand, Stx2 production was enhanced when using NAG and inhibited with the rest of the metabolites, whilst Stx2 translocation was only enhanced when using NANA, and this increase occurred only through the transcellular route. Overall, this study provides insights on the influence of the commensal microbiota on the pathogenicity of E. coli O157:H7, helping to identify favorable intestinal environments for the development of severe disease.
Collapse
|
16
|
Moysidou CM, Withers AM, Nisbet AJ, Price DRG, Bryant CE, Cantacessi C, Owens RM. Investigation of Host-Microbe-Parasite Interactions in an In Vitro 3D Model of the Vertebrate Gut. Adv Biol (Weinh) 2022; 6:e2200015. [PMID: 35652159 DOI: 10.1002/adbi.202200015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/23/2022] [Indexed: 01/28/2023]
Abstract
In vitro models of the gut-microbiome axis are in high demand. Conventionally, intestinal monolayers grown on Transwell setups are used to test the effects of commensals/pathogens on the barrier integrity, both under homeostatic and pathophysiological conditions. While such models remain valuable for deepening the understanding of host-microbe interactions, often, they lack key biological components that mediate this intricate crosstalk. Here, a 3D in vitro model of the vertebrate intestinal epithelium, interfaced with immune cells surviving in culture for over 3 weeks, is developed and applied to proof-of-concept studies of host-microbe interactions. More specifically, the establishment of stable host-microbe cocultures is described and functional and morphological changes in the intestinal barrier induced by the presence of commensal bacteria are shown. Finally, evidence is provided that the 3D vertebrate gut models can be used as platforms to test host-microbe-parasite interactions. Exposure of gut-immune-bacteria cocultures to helminth "excretory/secretory products" induces in vivo-like up-/down-regulation of certain cytokines. These findings support the robustness of the modular in vitro cell systems for investigating the dynamics of host-microbe crosstalk and pave the way toward new approaches for systems biology studies of pathogens that cannot be maintained in vitro, including parasitic helminths.
Collapse
Affiliation(s)
- Chrysanthi-Maria Moysidou
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, West Cambridge Site, CB3 0AS, UK
| | - Aimee M Withers
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, West Cambridge Site, CB3 0AS, UK
| | - Alasdair J Nisbet
- Moredun Research Institute, Pentlands Science Park, Edinburgh, EH26 0PZ, UK
| | - Daniel R G Price
- Moredun Research Institute, Pentlands Science Park, Edinburgh, EH26 0PZ, UK
| | - Clare E Bryant
- Department of Veterinary Medicine, Cambridge Veterinary School, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - Cinzia Cantacessi
- Department of Veterinary Medicine, Cambridge Veterinary School, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, West Cambridge Site, CB3 0AS, UK
| |
Collapse
|
17
|
Richter JF, Hildner M, Mrowka R, Schmauder R. Probing the leak pathway: Live-cell imaging of macromolecule passage through epithelia. Ann N Y Acad Sci 2022; 1516:151-161. [PMID: 35766317 DOI: 10.1111/nyas.14847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Epithelia compartmentalize multicellular organisms and provide interfacing between the inside and outside. Apart from regulating the exchange of solutes, uptake of nutrients, and excretion of waste products, their major function is to prevent uncontrolled access of foreign material to immune-competent compartments. Progress in understanding this barrier function toward larger solutes and its possible defects, as can be seen in a variety of diseases, is largely hampered by a lack of methods to spatiotemporally resolve transepithelial passage of macromolecules. Using different cell culture epithelia, we applied biotinylated dextran tracers carrying an acceptor fluorophore. These bind to cell-adherent avidin carrying donor fluorophore at the basolateral membranes of single-layered epithelial sheets. Confocal fluorescence microscopy was applied to living epithelia in order to image apical-to-basolateral tracer passage as a Förster resonance energy transfer signal of the fluorescent dextran-avidin pair over time. Stimulated macromolecule passage using barrier-perturbing agents proved its effectiveness for the leak imaging method presented herein. Over hours of imaging, spontaneous leaks were rare, occurring transiently on the scale of minutes and for the most part associated with rearranging cell junctions. The discussed approach to leak imaging is expected to promote the understanding of epithelial barriers, particularly, the nature and dynamics of the epithelial cell leak pathway.
Collapse
Affiliation(s)
- Jan F Richter
- Jena University Hospital, Institute of Anatomy II, Friedrich Schiller University Jena, Jena, Germany
| | - Markus Hildner
- Jena University Hospital, Institute of Anatomy II, Friedrich Schiller University Jena, Jena, Germany
| | - Ralf Mrowka
- Jena University Hospital, KIM III Department of Experimental Nephrology, Friedrich Schiller University Jena, Jena, Germany
| | - Ralf Schmauder
- Jena University Hospital, Institute of Physiology II, Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
18
|
Lansdell TA, Chambers LC, Dorrance AM. Endothelial Cells and the Cerebral Circulation. Compr Physiol 2022; 12:3449-3508. [PMID: 35766836 DOI: 10.1002/cphy.c210015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Endothelial cells form the innermost layer of all blood vessels and are the only vascular component that remains throughout all vascular segments. The cerebral vasculature has several unique properties not found in the peripheral circulation; this requires that the cerebral endothelium be considered as a unique entity. Cerebral endothelial cells perform several functions vital for brain health. The cerebral vasculature is responsible for protecting the brain from external threats carried in the blood. The endothelial cells are central to this requirement as they form the basis of the blood-brain barrier. The endothelium also regulates fibrinolysis, thrombosis, platelet activation, vascular permeability, metabolism, catabolism, inflammation, and white cell trafficking. Endothelial cells regulate the changes in vascular structure caused by angiogenesis and artery remodeling. Further, the endothelium contributes to vascular tone, allowing proper perfusion of the brain which has high energy demands and no energy stores. In this article, we discuss the basic anatomy and physiology of the cerebral endothelium. Where appropriate, we discuss the detrimental effects of high blood pressure on the cerebral endothelium and the contribution of cerebrovascular disease endothelial dysfunction and dementia. © 2022 American Physiological Society. Compr Physiol 12:3449-3508, 2022.
Collapse
Affiliation(s)
- Theresa A Lansdell
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Laura C Chambers
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, 48824, USA
| |
Collapse
|
19
|
Mo Y, Zhang X, Lao Y, Wang B, Li X, Zheng Y, Ding W. Fentanyl alleviates intestinal mucosal barrier damage in rats with severe acute pancreatitis by inhibiting the MMP-9/FasL/Fas pathway. Immunopharmacol Immunotoxicol 2022; 44:757-765. [PMID: 35616237 DOI: 10.1080/08923973.2022.2082304] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Fentanyl is an analgesic used against pancreatitis-related pain, while whether it ameliorates severe acute pancreatitis (SAP) has yet to be checked. The present study aims to determine fentanyl-delivered effect on SAP and the mechanism underlying this effect. METHODS Rat SAP models were established, following fentanyl treatment. The serum activity of amylase (AMY), lipase (LIP) and diamine oxidase (DAO) was detected by enzyme-linked immunosorbent assay. Histological examination was performed in the pancreatic and intestinal tissues with hematoxylin-eosin staining. After transfection with matrix metalloproteinase (MMP)9 overexpression plasmids, Caco-2 monolayers were treated with fentanyl and subsequently exposed to lipopolysaccharide (LPS). The transepithelial electrical resistance (TEER) value was determined in rat intestinal mucosa through an Ussing chamber assisted by Analyze & Acquire, and in Caco-2 cell monolayers through a voltohmmeter. Intestinal mucosa and paracellular permeabilities were determined by fluorescein isothiocyanate (FITC)-labeled dextran assay. The expressions of ZO-1, Occludin, MMP9, Fas and Fas ligand (FasL) in rat intestinal mucosa and/or Caco-2 monolayers were analyzed by qRT-PCR or/and western blot. RESULTS Fentanyl alleviated SAP-related histological alterations in the pancreas and intestines, reduced the elevated levels of SAP-related AMY, LIP and DAO, but promoted the levels of ZO-1 and Occludin. In SAP rats and Caco-2 monolayers, SAP-related or LPS-induced TEER value decreases, permeability increases, and increases in the expressions of MMP9, Fas and FasL were reversed partly by fentanyl. Notably, MMP9 overexpression could reverse the above fentanyl-delivered in vitro effects. CONCLUSION Fentanyl alleviates intestinal mucosal barrier damage in rats with SAP by inhibiting the MMP9/FasL/Fas pathway.
Collapse
Affiliation(s)
- Yunchao Mo
- Clinical Pharmacy, Central People's Hospital of Zhanjiang
| | - Xiangdong Zhang
- Surgical Intensive Care Unit, Central People's Hospital of Zhanjiang
| | - Yongguang Lao
- Surgical Intensive Care Unit, Central People's Hospital of Zhanjiang
| | - Bizhu Wang
- Pharmacy Department, Central People's Hospital of Zhanjiang
| | - Xinmei Li
- Surgical Intensive Care Unit, Central People's Hospital of Zhanjiang
| | - Yuhong Zheng
- Surgical Intensive Care Unit, Central People's Hospital of Zhanjiang
| | - Weihua Ding
- Surgical Intensive Care Unit, Central People's Hospital of Zhanjiang
| |
Collapse
|
20
|
Guler S, Kayali HA, Sadan EO, Sen B, Subasi E. Half-Sandwich Arene Ruthenium(II) Thiosemicarbazone Complexes: Evaluation of Anticancer Effect on Primary and Metastatic Ovarian Cancer Cell Lines. Front Pharmacol 2022; 13:882756. [PMID: 35620291 PMCID: PMC9128756 DOI: 10.3389/fphar.2022.882756] [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: 02/24/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
In this study, we describe the synthesis, characterization and antiproliferative activity of three organo-ruthenium(II) half-sandwich complexes [RuCl(η6-p-cym)(N,S-L)]Cl (I, II, and III). To form these complexes, three thiosemicarbazone ligands (TSCs) were synthesized; L = 5-nitro-2-carboxyaldehyde-thiophen-N-methyl-thiosemicarbazone, (L1); 2-acetyl-5-bromo-thiophen-N-methyl-thiosemicarbazone, (L2) and 2-acetyl-5-bromo-thiophen-N,N-dimethyl-thiosemicarbazone, (L3). The isolated compounds were analyzed using spectroscopic techniques such as elemental analysis, conductance measurements, FT-IR, 1H NMR spectroscopy, MALDI-TOF mass spectrometry, and single-crystal XRD. Our results demonstrated that the synthesized thiosemicarbazone ligands (TSCs) are bound to the metal ion as a bidentate ligand that coordinates through the thiocarbonyl sulfur and azomethine nitrogen atoms in all complexes (I, II, and III). The X-ray crystal structures of L1 and L2 revealed that both compounds are crystallized in the triclinic crystal system with space group P-1. The biological potency of newly synthesized TSC ligands (L1, L2, and L3) and their corresponding ruthenium complexes (I, II, and III) were investigated on human primary ovarian (A2780) and human metastatic ovarian (OVCAR-3) cell lines. To get detailed information respecting antitumor properties, cytotoxicity, DNA/BSA binding affinity, cellular uptake, DNA binding competition, and trans-epithelial resistance measurement assays were performed. Our results demonstrate that newly synthesized ruthenium(II) complexes possess potential biological activity. Moreover, we observe that the ruthenium complexes reported here show anticancer activity on primary (A2780) and metastatic (OVCAR-3) ovarian cancer cells.
Collapse
Affiliation(s)
- Seminay Guler
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Hulya Ayar Kayali
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
- Department of Chemistry, Division of Biochemistry, Faculty of Science, Dokuz Eylul University, İzmir, Turkey
| | - Egemen Orkun Sadan
- Institute of Science and Technology, Dokuz Eylul University, Izmir, Turkey
| | - Betul Sen
- Department of Physics, Faculty of Science, Dokuz Eylul University, Izmir, Turkey
| | - Elif Subasi
- Department of Chemistry, Faculty of Science, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
21
|
Breaking through the barrier: Modelling and exploiting the physical microenvironment to enhance drug transport and efficacy. Adv Drug Deliv Rev 2022; 184:114183. [PMID: 35278523 DOI: 10.1016/j.addr.2022.114183] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/03/2022] [Accepted: 03/06/2022] [Indexed: 02/08/2023]
Abstract
Pharmaceutical compounds are the main pillar in the treatment of various illnesses. To administer these drugs in the therapeutic setting, multiple routes of administration have been defined, including ingestion, inhalation, and injection. After administration, drugs need to find their way to the intended target for high effectiveness, and this penetration is greatly dependent on obstacles the drugs encounter along their path. Key hurdles include the physical barriers that are present within the body and knowledge of those is indispensable for progress in the development of drugs with increased therapeutic efficacy. In this review, we examine several important physical barriers, such as the blood-brain barrier, the gut-mucosal barrier, and the extracellular matrix barrier, and evaluate their influence on drug transport and efficacy. We explore various in vitro model systems that aid in understanding how parameters within the barrier model affect drug transfer and therapeutic effect. We conclude that physical barriers in the body restrict the quantity of drugs that can pass through, mainly as a consequence of the barrier architecture. In addition, the specific physical properties of the tissue can trigger intracellular changes, altering cell behavior in response to drugs. Though the barriers negatively influence drug distribution, physical stimulation of the surrounding environment may also be exploited as a mechanism to control drug release. This drug delivery approach is explored in this review as a potential alternative to the conventional ways of delivering therapeutics.
Collapse
|
22
|
Parajuli P, Gokulan K, Khare S. Preclinical In Vitro Model to Assess the Changes in Permeability and Cytotoxicity of Polarized Intestinal Epithelial Cells during Exposure Mimicking Oral or Intravenous Routes: An Example of Arsenite Exposure. Int J Mol Sci 2022; 23:ijms23094851. [PMID: 35563241 PMCID: PMC9101442 DOI: 10.3390/ijms23094851] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 12/04/2022] Open
Abstract
The gastrointestinal tract (GIT) is exposed to xenobiotics, including drugs, through both: local (oral) and systemic routes. Despite the advances in drug discovery and in vitro pre-clinical models, there is a lack of appropriate translational models to distinguish the impact of these routes of exposure. Changes in intestinal permeability has been observed in different gastrointestinal and systemic diseases. This study utilized one such xenobiotic, arsenic, to which more than 200 million people around the globe are exposed via their food, drinking water, work environment, soil, and air. The purpose of this study was to establish an in vitro model to mimic gastrointestinal tract exposure to xenobiotics via oral or intravenous routes. To achieve this, we compared the route (mimicking oral and intravenous exposure to GIT and the dose response (using threshold approach) of trivalent and pentavalent inorganic arsenic species on the permeability of in vitro cultured polarized T84 cells, an example of intestinal epithelial cells. Arsenic treatment to polarized T84 cells via the apical and basolateral compartment of the trans-well system reflected oral or intravenous routes of exposure in vivo, respectively. Sodium arsenite, sodium arsenate, dimethyl arsenic acid sodium salt (DMAV), and disodium methyl arsonate hydrate (MMAV) were assessed for their effects on intestinal permeability by measuring the change in trans-epithelial electrical resistance (TEER) of T-84 cells. Polarized T-84 cells exposed to 12.8 µM of sodium arsenite from the basolateral side showed a marked reduction in TEER. Cytotoxicity of sodium arsenite, as measured by release of lactate dehydrogenase (LDH), was increased when cells were exposed via the basolateral side. The mRNA expression of genes related to cell junctions in T-84 cells was analyzed after exposure with sodium arsenite for 72 h. Changes in TEER correlated with mRNA expression of focal-adhesion-, tight-junction- and gap-junction-related genes (upregulation of Jam2, Itgb3 and Notch4 genes and downregulation of Cldn2, Cldn3, Gjb1, and Gjb2). Overall, exposure to sodium arsenite from the basolateral side was found to have a differential effect on monolayer permeability and on cell-junction-related genes as compared to apical exposure. Most importantly, this study established a preclinical human-relevant in vitro translational model to assess the changes in permeability and cytotoxicity during exposure, mimicking oral or intravenous routes.
Collapse
|
23
|
Pitsalidis C, Pappa AM, Boys AJ, Fu Y, Moysidou CM, van Niekerk D, Saez J, Savva A, Iandolo D, Owens RM. Organic Bioelectronics for In Vitro Systems. Chem Rev 2021; 122:4700-4790. [PMID: 34910876 DOI: 10.1021/acs.chemrev.1c00539] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bioelectronics have made strides in improving clinical diagnostics and precision medicine. The potential of bioelectronics for bidirectional interfacing with biology through continuous, label-free monitoring on one side and precise control of biological activity on the other has extended their application scope to in vitro systems. The advent of microfluidics and the considerable advances in reliability and complexity of in vitro models promise to eventually significantly reduce or replace animal studies, currently the gold standard in drug discovery and toxicology testing. Bioelectronics are anticipated to play a major role in this transition offering a much needed technology to push forward the drug discovery paradigm. Organic electronic materials, notably conjugated polymers, having demonstrated technological maturity in fields such as solar cells and light emitting diodes given their outstanding characteristics and versatility in processing, are the obvious route forward for bioelectronics due to their biomimetic nature, among other merits. This review highlights the advances in conjugated polymers for interfacing with biological tissue in vitro, aiming ultimately to develop next generation in vitro systems. We showcase in vitro interfacing across multiple length scales, involving biological models of varying complexity, from cell components to complex 3D cell cultures. The state of the art, the possibilities, and the challenges of conjugated polymers toward clinical translation of in vitro systems are also discussed throughout.
Collapse
Affiliation(s)
- Charalampos Pitsalidis
- Department of Physics, Khalifa University of Science and Technology, P.O. Box 127788, Abu Dhabi 127788, UAE.,Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| | - Anna-Maria Pappa
- Department of Biomedical Engineering, Khalifa University of Science and Technology, P.O. Box 127788, Abu Dhabi 127788, UAE
| | - Alexander J Boys
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| | - Ying Fu
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.,Department of Pure and Applied Chemistry, Technology and Innovation Centre, University of Strathclyde, Glasgow G1 1RD, U.K
| | - Chrysanthi-Maria Moysidou
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| | - Douglas van Niekerk
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| | - Janire Saez
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K.,Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Avenida Miguel de Unamuno, 3, 01006 Vitoria-Gasteiz, Spain.,Ikerbasque, Basque Foundation for Science, E-48011 Bilbao, Spain
| | - Achilleas Savva
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| | - Donata Iandolo
- INSERM, U1059 Sainbiose, Université Jean Monnet, Mines Saint-Étienne, Université de Lyon, 42023 Saint-Étienne, France
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge Philippa Fawcett Drive, Cambridge CB3 0AS, U.K
| |
Collapse
|
24
|
Rohhimi W, Tan JW, Liew KY, Jacquet A, Harith HH, Israf DA, Tham CL. Zerumbone attenuates house dust mite extract-induced epithelial barrier dysfunction in 16HBE14o- cells. Immunopharmacol Immunotoxicol 2021; 43:813-824. [PMID: 34694946 DOI: 10.1080/08923973.2021.1992633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
CONTEXT The airway epithelial barrier can be disrupted by house dust mite (HDM) allergens leading to allergic airway inflammation. Zerumbone, a natural monocyclic sesquiterpene, was previously found to possess anti-asthmatic effect by modulating Th1/Th2 cytokines. However, the protective role of zerumbone on epithelial barrier function remains to be fully explored. OBJECTIVE To investigate the effect of zerumbone on HDM extract-induced airway epithelial barrier dysfunction. MATERIALS AND METHODS Human bronchial epithelial cells 16HBE14o- were incubated with 100 μg/mL HDM extract and treated with non-cytotoxic concentrations of zerumbone (6.25 μM, 12.5 μM, and 25 μM) for 24 h. The epithelial junctional integrity and permeability were evaluated through transepithelial electrical resistance (TEER) and fluorescein isothiocynate (FITC)-Dextran permeability assays, respectively. The localization of junctional proteins, occludin and zona occludens (ZO)-1, was studied using immunofluorescence (IF) while the protein expression was measured by western blot. RESULTS Zerumbone inhibited changes in junctional integrity (6.25 μM, p ≤ .05; 12.5 μM, p ≤ .001; 25 μM, p ≤ .001) and permeability (6.25 μM, p ≤ .05; 12.5 μM, p ≤ .01; 25 μM, p ≤ .001) triggered by HDM extract in a concentration-dependent manner. This protective effect could be explained by the preservation of occludin (12.5 μM, p ≤ .01 and 25 μM, p ≤ .001) and ZO-1 (12.5 μM, p ≤ .05 and 25 μM, p ≤ .001) localization, rather than the prevention of their cleavage. DISCUSSION AND CONCLUSION Zerumbone attenuates HDM extract-induced epithelial barrier dysfunction which supports its potential application for the treatment of inflammation-driven airway diseases such as asthma.
Collapse
Affiliation(s)
- Wafda Rohhimi
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Ji Wei Tan
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia.,School of Science, Monash University Malaysia, Subang Jaya, Malaysia
| | - Kong Yen Liew
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Alain Jacquet
- Center of Excellence in Vaccine Research and Development, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Hanis Hazeera Harith
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Daud Ahmad Israf
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Chau Ling Tham
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
25
|
Prolonged sub-lethal exposure to galaxolide (HHCB) and tonalide (AHTN) promotes the metastatic potential of glioblastoma tumor spheroids. Neurotoxicology 2021; 87:219-230. [PMID: 34687775 DOI: 10.1016/j.neuro.2021.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 10/06/2021] [Accepted: 10/18/2021] [Indexed: 02/08/2023]
Abstract
Galaxolide and tonalide are well-known polycyclic musks whose intensive use without limitations in numerous cleaning, hygiene, and personal care products has resulted in widespread direct human exposure via absorption, inhalation, and oral ingestion. Latest data shows that long-term, low-dose exposure to toxic chemicals can induce unpredictable harmful effects in a variety of living systems, however, interactions between synthetic musks and brain tumours remain largely unexplored. Glioblastoma (GB) accounts for nearly half of all tumours of the central nervous system and is characterized by very poor prognosis. The aims of this study were (1) to investigate the potential effect of long-term (20-generation) single and combined application of galaxolide and tonalide at sub-lethal doses (5-2.5 u M) on the angiogenesis, invasion, and migration of human U87 cells or tumour spheroids, and (2) to explore the underlying molecular mechanisms. Random amplified polymorphic DNA assays revealed significant DNA damage and increased total mutation load in galaxolide- and/or tonalide-treated U87 cells. In those same groups, we also detected remarkable tumour spheroid invasion and up-regulation of both HIF1-α/VEGF/MMP9 and IL6/JAK2/STAT3 signals, known to have important roles in hypoxia-related angiogenesis and/or proliferation. Prolonged musk treatment further altered angio-miRNA expression in a manner consistent with poor prognosis in GB. We also detected significant over-expression of the genes Slug, Snail, ZEB1, and Vimentin, which are biomarkers of epithelial to mesenchymal transition. In addition, matrigel, transwell, and wound healing assays clearly showed that long-term sub-lethal exposure to galaxolide and/or tonalide induced invasion and migration proposing a high metastatic potential. Our results suggest that assessing expression of HIF-1a, VEGF, STAT3, and the miR-17-92 cluster in biopsy samples of GB patients who have a history of possible long-term exposure to galaxolide or tonalide could be beneficial for deciding a therapy regime. Additionally, we recommend that extensively-used hygiene and cleaning materials be selected from synthetic musk-free products, especially when used in palliative care processes for GB patients.
Collapse
|
26
|
Gallegos-Alcalá P, Jiménez M, Cervantes-García D, Salinas E. The Keratinocyte as a Crucial Cell in the Predisposition, Onset, Progression, Therapy and Study of the Atopic Dermatitis. Int J Mol Sci 2021; 22:ijms221910661. [PMID: 34639001 PMCID: PMC8509070 DOI: 10.3390/ijms221910661] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 12/24/2022] Open
Abstract
The keratinocyte (KC) is the main functional and structural component of the epidermis, the most external layer of the skin that is highly specialized in defense against external agents, prevention of leakage of body fluids and retention of internal water within the cells. Altered epidermal barrier and aberrant KC differentiation are involved in the pathophysiology of several skin diseases, such as atopic dermatitis (AD). AD is a chronic inflammatory disease characterized by cutaneous and systemic immune dysregulation and skin microbiota dysbiosis. Nevertheless, the pathological mechanisms of this complex disease remain largely unknown. In this review, we summarize current knowledge about the participation of the KC in different aspects of the AD. We provide an overview of the genetic predisposing and environmental factors, inflammatory molecules and signaling pathways of the KC that participate in the physiopathology of the AD. We also analyze the link among the KC, the microbiota and the inflammatory response underlying acute and chronic skin AD lesions.
Collapse
Affiliation(s)
- Pamela Gallegos-Alcalá
- Department of Microbiology, Center of Basic Science, Autonomous University of Aguascalientes, Aguascalientes 20100, Mexico; (P.G.-A.); (M.J.); (D.C.-G.)
| | - Mariela Jiménez
- Department of Microbiology, Center of Basic Science, Autonomous University of Aguascalientes, Aguascalientes 20100, Mexico; (P.G.-A.); (M.J.); (D.C.-G.)
| | - Daniel Cervantes-García
- Department of Microbiology, Center of Basic Science, Autonomous University of Aguascalientes, Aguascalientes 20100, Mexico; (P.G.-A.); (M.J.); (D.C.-G.)
- National Council of Science and Technology, Ciudad de México 03940, Mexico
| | - Eva Salinas
- Department of Microbiology, Center of Basic Science, Autonomous University of Aguascalientes, Aguascalientes 20100, Mexico; (P.G.-A.); (M.J.); (D.C.-G.)
- Correspondence: ; Tel.: +52-449-9108424
| |
Collapse
|
27
|
Brayden DJ, Maher S. Transient Permeation Enhancer® (TPE®) technology for oral delivery of octreotide: a technological evaluation. Expert Opin Drug Deliv 2021; 18:1501-1512. [PMID: 34128734 DOI: 10.1080/17425247.2021.1942838] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The FDA approval of oral semaglutide for type 2 diabetes (2019) and oral octreotide for acromegaly (2020) is evidence that selected niche peptides can be administered orally if formulated with selected intestinal permeation enhancers. AREAS COVERED We evaluated the oral octreotide formulation, MYCAPSSA® (Chiasma Pharmaceuticals, Needham, MA, USA). An outline of the current standard of care in acromegaly and the benefits of oral octreotide versus depot injections is provided. We discuss the Transient Permeation Enhancer (TPE®) technology used and detail the safety and efficacy data from animal models and clinical trials. EXPERT OPINION TPE® is an oily suspension of octreotide that includes a number of excipients that can transiently alter epithelial barrier integrity by opening of intestinal epithelial tight junctions arising from transcellular perturbation. Phase I studies using 20 mg octreotide capsules yielded a relative oral bioavailability of ~0.7% and primary endpoints were achieved in two Phase III studies. The oral octreotide dose required to achieve these endpoints was over 200 times that of the 0.1 mg immediate-release subcutaneous injection, a reminder of the difficulty in achieving oral absorption of macromolecules. Many acromegaly patients will prefer a convenient twice-daily oral formulation of octreotide compared to monthly depot injections.
Collapse
Affiliation(s)
- David J Brayden
- University College Dublin (UCD) School of Veterinary Medicine, UCD, Belfield, Dublin 4, Ireland.,UCD Conway Institute of Biotechnology, UCD, Belfield, Dublin 4, Ireland.,CÚRAM, the SFI Research Centre for Medical Devices, UCD, Belfield, Dublin 4, Ireland
| | - Sam Maher
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, St. Stephen's Green, Dublin 2, Ireland
| |
Collapse
|
28
|
Effect of Graphene and Graphene Oxide on Airway Barrier and Differential Phosphorylation of Proteins in Tight and Adherens Junction Pathways. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:nano11051283. [PMID: 34068174 PMCID: PMC8152977 DOI: 10.3390/nano11051283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/09/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022]
Abstract
Via inhalation we are continuously exposed to environmental and occupational irritants which can induce adverse health effects, such as irritant-induced asthma (IIA). The airway epithelium forms the first barrier encountered by these agents. We investigated the effect of environmental and occupational irritants on the airway epithelial barrier in vitro. The airway epithelial barrier was mimicked using a coculture model, consisting of bronchial epithelial cells (16HBE) and monocytes (THP-1) seeded on the apical side of a permeable support, and human lung microvascular endothelial cells (HLMVEC) grown on the basal side. Upon exposure to graphene (G) and graphene oxide (GO) in a suspension with fetal calf serum (FCS), ammonium persulfate (AP), sodium persulfate (SP) and hypochlorite (ClO−), the transepithelial electrical resistance (TEER) and flux of fluorescent labelled dextran (FD4-flux), was determined. Exposure to graphene nanoparticles (GNPs) induced an immediate negative effect on the epithelial barrier, whereas ClO− only had a negative impact after 24 h of exposure. AP and SP did not affect the barrier properties. The tight junctions (TJ) network showed less connected zonula occludens 1 (ZO-1) and occludin staining in GNP-exposed cocultures. Functional analysis of the phosphoproteomic data indicated that proteins in the adherens junction (AJ) and TJ pathways showed an altered phosphorylation due to GNP exposure. To conclude, the negative effect of GNPs on the epithelial barrier can be explained by the slightly altered the TJ organization which could be caused by alterations in the phosphorylation level of proteins in the AJ and TJ pathway.
Collapse
|
29
|
Park SK, Yeon SH, Choi MR, Choi SH, Lee SB, Rha KS, Kim YM. Urban Particulate Matters May Affect Endoplasmic Reticulum Stress and Tight Junction Disruption in Nasal Epithelial Cells. Am J Rhinol Allergy 2021; 35:817-829. [PMID: 33736454 DOI: 10.1177/19458924211004006] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Exposure to airborne urban particulate matter (UPM) has been closely related to the development and aggravation of respiratory disease, including sinonasal disorders. OBJECTIVE The aims of this study were to investigate the effect of UPM on nasal epithelial tight junctions (TJs) and mucosal barrier function and delineate the underlying mechanism by using both in vitro and in vivo models. METHODS In this study, human nasal epithelial cells (hNECs) and BALB/c mice were exposed to UPMs. UPM 1648a and 1649 b were employed. TJ and endoplasmic reticulum (ER) stress marker expression was measured using western blot analysis and immunofluorescence. TJ integrity and nasal epithelial barrier function were evaluated by transepithelial electric resistance (TER) and paracellular flux. In addition, the effects of N-acetyl-L-cysteine (NAC) on UPM-induced nasal epithelial cells were investigated. RESULTS UPM significantly impaired the nasal epithelial barrier, as demonstrated by decreased protein expression of TJ and ER stress markers in human nasal epithelial cells. This finding was in parallel to reduced transepithelial electrical resistance and increased fluorescein isothiocyanate-dextran permeability. Pretreatment with NAC decreased the degree of UPM-mediated ER stress and restored nasal epithelial barrier disruption in human nasal epithelial cells (hNEC) and the nasal mucosa of experimental animals. CONCLUSION These data suggest that UPMs may induce nasal epithelial barrier dysfunction by targeting TJs and ER stress could be related in this process. Based on these results, we suggest that suppression of this process with an inhibitor targeting ER stress responses could represent a novel promising therapeutic target in UPM-induced sinonasal disease.
Collapse
Affiliation(s)
- Soo Kyoung Park
- Department of Otorhinolaryngology-Head and Neck Surgery, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Sun Hee Yeon
- Department of Otorhinolaryngology-Head and Neck Surgery, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Mi-Ra Choi
- Department of Otorhinolaryngology-Head and Neck Surgery, Chungnam National University School of Medicine, Daejeon, Republic of Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Seung Hyeon Choi
- Department of Otorhinolaryngology-Head and Neck Surgery, Chungnam National University School of Medicine, Daejeon, Republic of Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Sung Bok Lee
- Department of Ophthalmology, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Ki-Sang Rha
- Department of Otorhinolaryngology-Head and Neck Surgery, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Yong Min Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Chungnam National University School of Medicine, Daejeon, Republic of Korea.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| |
Collapse
|
30
|
Raut B, Chen LJ, Hori T, Kaji H. An Open-Source Add-On EVOM ® Device for Real-Time Transepithelial/Endothelial Electrical Resistance Measurements in Multiple Transwell Samples. MICROMACHINES 2021; 12:282. [PMID: 33800233 PMCID: PMC8000980 DOI: 10.3390/mi12030282] [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] [Received: 01/21/2021] [Revised: 02/24/2021] [Accepted: 03/04/2021] [Indexed: 11/23/2022]
Abstract
This study provides design of a low-cost and open source add-on device that enhances the functionality of the popular EVOM® instrument for transepithelial/endothelial electrical resistance (TEER) measurement. The original EVOM® instrument is designed for measuring TEER in transwell samples manually using a pair of Ag/AgCl electrodes. The inconsistency in electrode placement, temperature variation, and a typically large (12-24 h) time interval between measurements result in large data variabilities. Thus, to solve the current limitation of the EVOM® instrument, we built an add-on device using a custom designed electronic board and a 3D printed electrode holder that allowed automated TEER measurements in multiple transwell samples. To demonstrate the functionality of the device prototype, we monitored TEER in 4 transwell samples containing retinal cells (ARPE-19) for 67 h. Furthermore, by monitoring temperature of the cell culture medium, we were able to detect fluctuations in TEER due to temperature change after the medium change process, and were able to correct the data offset. Although we demonstrated the use of our add-on device on EVOM® instrument only, the concept (multiplexing using digitally controlled relays) and hardware (custom data logger) presented here can be applied to more advanced TEER instruments to improve the performance of those devices.
Collapse
Affiliation(s)
- Bibek Raut
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-01 Aramaki, Aoba-ku, Sendai 980-8579, Japan; (B.R.); (L.-J.C.); (T.H.)
| | - Li-Jiun Chen
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-01 Aramaki, Aoba-ku, Sendai 980-8579, Japan; (B.R.); (L.-J.C.); (T.H.)
| | - Takeshi Hori
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-01 Aramaki, Aoba-ku, Sendai 980-8579, Japan; (B.R.); (L.-J.C.); (T.H.)
| | - Hirokazu Kaji
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-01 Aramaki, Aoba-ku, Sendai 980-8579, Japan; (B.R.); (L.-J.C.); (T.H.)
- Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, 6-6-01 Aramaki, Aoba-ku, Sendai 980-8579, Japan
| |
Collapse
|
31
|
Shrestha J, Ryan ST, Mills O, Zhand S, Razavi Bazaz S, Hansbro PM, Ghadiri M, Ebrahimi Warkiani M. A 3D-printed microfluidic platform for simulating the effects of CPAP on the nasal epithelium. Biofabrication 2021; 13. [PMID: 33561837 DOI: 10.1088/1758-5090/abe4c1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/09/2021] [Indexed: 02/01/2023]
Abstract
Obstructive sleep apnoea (OSA) is a chronic disorder that involves a decrease or complete cessation of airflow during sleep. It occurs when the muscles supporting the soft tissues in the throat relax during sleep, causing narrowing or closure of the upper airway. Sleep apnoea is a serious medical condition with an increased risk of cardiovascular complications and impaired quality of life. Continuous positive airway pressure (CPAP) is the most effective treatment for moderate to severe cases of OSA and is effective in mild sleep apnoea. However, CPAP therapy is associated with the development of several nasal side effects and is inconvenient for the user, leading to low compliance rates. The effects of CPAP treatment on the upper respiratory system, as well as the pathogenesis of side effects, are incompletely understood and not adequately researched. To better understand the effects of CPAP treatment on the upper respiratory system, we developed an in vitro 3D-printed microfluidic platform. A nasal epithelial cell line, RPMI 2650, was then exposed to certain conditions to mimic the in-vivo environment. To create these conditions, the microfluidic device was utilized to expose nasal epithelial cells grown and differentiated at the air-liquid interface. The airflow was similar to what is experienced with CPAP, with pressure ranging between 0-20 cm of H20. Cells exposed to pressure showed decreased barrier integrity, change in cellular shape, and increased cell death (lactate dehydrogenase release into media) compared to unstressed cells. Stressed cells also showed increased secretions of inflammatory markers IL-6 and IL-8 and had increased production of ATP. Our results suggest that stress induced by airflow leads to structural, metabolic, and inflammatory changes in the nasal epithelium, which may be responsible for developing nasal side-effects following CPAP treatment.
Collapse
Affiliation(s)
- Jesus Shrestha
- University of Technology Sydney, 15 Broadway, Sydney, New South Wales, 2007, AUSTRALIA
| | - Sean Thomas Ryan
- The University of Sydney, 15 Broadway, Sydney, New South Wales, 2006, AUSTRALIA
| | - Oliver Mills
- The University of Sydney, Camperdown, Sydney, New South Wales, 2006, AUSTRALIA
| | - Sareh Zhand
- University of Technology Sydney, 15 Broadway, Sydney, New South Wales, 2007, AUSTRALIA
| | - Sajad Razavi Bazaz
- School of Biomedical Engineering, University of Technology Sydney, 15 Broadway, Sydney, New South Wales, 2007, AUSTRALIA
| | | | - Maliheh Ghadiri
- The University of Sydney, Camperdown, Sydney, New South Wales, 2006, AUSTRALIA
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney Faculty of Engineering and Information Technology, 15 Broadway, Sydney, New South Wales, 2007, AUSTRALIA
| |
Collapse
|
32
|
Shini S, Aland RC, Bryden WL. Avian intestinal ultrastructure changes provide insight into the pathogenesis of enteric diseases and probiotic mode of action. Sci Rep 2021; 11:167. [PMID: 33420315 PMCID: PMC7794591 DOI: 10.1038/s41598-020-80714-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/24/2020] [Indexed: 01/27/2023] Open
Abstract
Epithelial damage and loss of barrier integrity occur following intestinal infections in humans and animals. Gut health was evaluated by electron microscopy in an avian model that exposed birds to subclinical necrotic enteritis (NE) and fed them a diet supplemented with the probiotic Bacillus amyloliquefaciens strain H57 (H57). Scanning electron microscopy of ileal mucosa revealed significant villus damage, including focal erosions of epithelial cells and villous atrophy, while transmission electron microscopy demonstrated severe enterocyte damage and loss of cellular integrity in NE-exposed birds. In particular, mitochondria were morphologically altered, appearing irregular in shape or swollen, and containing electron-lucent regions of matrix and damaged cristae. Apical junctional complexes between adjacent enterocytes were significantly shorter, and the adherens junction was saccular, suggesting loss of epithelial integrity in NE birds. Segmented filamentous bacteria attached to villi, which play an important role in intestinal immunity, were more numerous in birds exposed to NE. The results suggest that mitochondrial damage may be an important initiator of NE pathogenesis, while H57 maintains epithelium and improves the integrity of intestinal mucosa. Potential actions of H57 are discussed that further define the mechanisms responsible for probiotic bacteria’s role in maintaining gut health.
Collapse
Affiliation(s)
- Shaniko Shini
- School of Agriculture and Food Sciences, University of Queensland, Gatton, QLD, 4343, Australia.
| | - R Claire Aland
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD, 4071, Australia
| | - Wayne L Bryden
- School of Agriculture and Food Sciences, University of Queensland, Gatton, QLD, 4343, Australia
| |
Collapse
|
33
|
Ferrari E, Palma C, Vesentini S, Occhetta P, Rasponi M. Integrating Biosensors in Organs-on-Chip Devices: A Perspective on Current Strategies to Monitor Microphysiological Systems. BIOSENSORS 2020; 10:E110. [PMID: 32872228 PMCID: PMC7558092 DOI: 10.3390/bios10090110] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 01/20/2023]
Abstract
Organs-on-chip (OoC), often referred to as microphysiological systems (MPS), are advanced in vitro tools able to replicate essential functions of human organs. Owing to their unprecedented ability to recapitulate key features of the native cellular environments, they represent promising tools for tissue engineering and drug screening applications. The achievement of proper functionalities within OoC is crucial; to this purpose, several parameters (e.g., chemical, physical) need to be assessed. Currently, most approaches rely on off-chip analysis and imaging techniques. However, the urgent demand for continuous, noninvasive, and real-time monitoring of tissue constructs requires the direct integration of biosensors. In this review, we focus on recent strategies to miniaturize and embed biosensing systems into organs-on-chip platforms. Biosensors for monitoring biological models with metabolic activities, models with tissue barrier functions, as well as models with electromechanical properties will be described and critically evaluated. In addition, multisensor integration within multiorgan platforms will be further reviewed and discussed.
Collapse
Affiliation(s)
| | | | | | | | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133 Milano, Italy; (E.F.); (C.P.); (S.V.); (P.O.)
| |
Collapse
|
34
|
Husain A, Ganesan A, Machacek M, Cerveny L, Kubat P, Ghazal B, Zimcik P, Makhseed S. Dually directional glycosylated phthalocyanines as extracellular red-emitting fluorescent probes. Dalton Trans 2020; 49:9605-9617. [PMID: 32542251 DOI: 10.1039/d0dt01180k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The development of new non-aggregated phthalocyanines bearing multivalent saccharide moieties on their macrocyclic rims is of great interest. Many characteristics, including water-solubility, non-toxicity and others, can be feasibly obtained by these amphiphiles which can be considered as a key solution for demonstrating highly efficient photoactive materials in water. Herein, a family of five newly prepared dually directional Zn(ii) containing phthalocyanines (PcG1-4) and azaphthalocyanine (AzaPcG1) glycoconjugates is described. The unique spatial arrangement of the glucoside units based on peripherally hexadeca-(PcG1) and nonperipherally octa-(PcG4) macrocycles provides a fully monomeric behaviour along with a high fluorescence (ΦF∼ 0.21) in aqueous solution. These amphiphiles were characterized by low toxicity, and an extremely low cellular uptake was obtained due to the highly polar nature of the glucoside substituents. Accordingly, their potential as suitable photoactive chromophores for red-emitting extracellular fluorescent probes has been confirmed upon the evaluation of paracellular transport using a layer of MDCKII cells with the permeability coefficient fully comparable with an established evaluator of the integrity of the monolayer.
Collapse
Affiliation(s)
- Ali Husain
- Department of Chemistry, Kuwait University, P.O. Box 5969, Safat, 13060, Kuwait.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Vazquez-Carretero MD, García-Miranda P, Balda MS, Matter K, Ilundáin AA, Peral MJ. Proper E-cadherin membrane location in colon requires Dab2 and it modifies by inflammation and cancer. J Cell Physiol 2020; 236:1083-1093. [PMID: 32617970 DOI: 10.1002/jcp.29917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/03/2020] [Accepted: 06/21/2020] [Indexed: 12/30/2022]
Abstract
We reported that Disabled-2 (Dab2) is located at the apical membrane in suckling rat intestine. Here, we discovered that, in colon of suckling and adult mouse and of adult human, Dab2 is only at lateral crypt cell membrane and colocalized with E-cadherin. Dab2 depletion in Caco-2 cells led to E-cadherin internalization indicating that its membrane location requires Dab2. In mice, we found that 3 days of dextran sulfate sodium-induced colitis increased Dab2/E-cadherin colocalization, which was decreased as colitis progressed to 6 and 9 days. In agreement, Dab2/E-cadherin colocalization increased in human mild and severe ulcerative colitis and in polyps, being reduced in colon adenocarcinomas, which even showed epithelial Dab2 absence and E-cadherin delocalization. Epithelial Dab2 decrement preceded that of E-cadherin. We suggest that Dab2, by inhibiting E-cadherin internalization, stabilizes adherens junctions, and its absence from the epithelium may contribute to development of colon inflammation and cancer.
Collapse
Affiliation(s)
| | - Pablo García-Miranda
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - María S Balda
- Department of Cell Biology, Institute of Ophthalmology, University College London, London, UK
| | - Karl Matter
- Department of Cell Biology, Institute of Ophthalmology, University College London, London, UK
| | - Anunciación A Ilundáin
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - María J Peral
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
36
|
An organoid model to assay the role of CFTR in the human epididymis epithelium. Cell Tissue Res 2020; 381:327-336. [PMID: 32377875 DOI: 10.1007/s00441-020-03208-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/20/2020] [Indexed: 12/12/2022]
Abstract
Organoid cultures derived from primary human tissues facilitate the study of disease processes and the development of new therapeutics. Most men with cystic fibrosis (CF) are infertile due to defects in the epididymis and vas deferens; however, the causative mechanisms are still unclear. We used human epididymis epithelial cell (HEE) organoids and polarized HEE cell cultures to assay the CF transmembrane conductance regulator (CFTR) in the human epididymis. 3D HEE organoids and polarized 2D HEE cell cultures on membrane inserts were established from human caput epididymis. Single-cell RNA sequencing (scRNA-seq) was performed to map cell type-specific gene expression in the organoids. Using forskolin (FSK) to activate CFTR and inhibitor CFTRinh172 to block its activity, we assessed how CFTR contributes to organoid swelling and epithelial barrier function. The scRNA-seq data showed key caput epididymis cell types present in HEE organoid cultures. FSK at 10 μM induced HEE organoid swelling by 20% at 16 h, while 5 and 10 μM CFTRinh172 treatment significantly reduced HEE organoid size. In transepithelial resistance (TER) measurements, FSK reduced TER, while inhibition of CFTR increased TER; also, depletion of CFTR with specific siRNAs significantly increased TER. FSK treatment significantly increased the flux of 4-kDa but not 70-kDa dextran, suggesting activation of CFTR mainly enhances transcellular diffusion. We have demonstrated that CFTR contributes to the maintenance of HEE cell TER and that cultured HEE organoids are a useful model to investigate human epididymis function. These results facilitate progress in elucidating how CFTR-dependent cellular processes impair fertility in CF.
Collapse
|
37
|
Degroote J, Vergauwen H, Wang W, Van Ginneken C, De Smet S, Michiels J. Changes of the glutathione redox system during the weaning transition in piglets, in relation to small intestinal morphology and barrier function. J Anim Sci Biotechnol 2020; 11:45. [PMID: 32337030 PMCID: PMC7178753 DOI: 10.1186/s40104-020-00440-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 02/24/2020] [Indexed: 12/12/2022] Open
Abstract
Background Weaning is known to result in barrier dysfunction and villus atrophy in the immediate post-weaning phase, and the magnitude of these responses is hypothesized to correlate with changes in the glutathione (GSH) redox system. Therefore, these parameters were simultaneously measured throughout the weaning phase, in piglets differing in birth weight category and weaning age, as these pre-weaning factors are important determinants for the weaning transition. Low birth weight (LBW) and normal birth weight (NBW) littermates were assigned to one of three weaning treatments; i.e. weaning at 3 weeks of age (3w), weaning at 4 weeks of age (4w) and removal from the sow at 3 d of age and fed a milk replacer until weaning at 3 weeks of age (3d3w). For each of these treatments, six LBW and six NBW piglets were euthanized at 0, 2, 5, 12 or 28 d post-weaning piglets, adding up 180 piglets. Results Weaning increased the glutathione peroxidase activity on d 5 post-weaning in plasma, and duodenal and jejunal mucosa. Small intestinal glutathione-S-transferase activity gradually increased until d 12 post-weaning, and this was combined with a progressive rise of mucosal GSH up till d 12 post-weaning. Oxidation of the GSH redox status (GSH/GSSG Eh) was only observed in the small intestinal mucosa of 3d3w weaned piglets at d 5 post-weaning. These piglets also demonstrated increased fluorescein isothiocyanate dextran (FD4) and horseradish peroxidase fluxes in the duodenum and distal jejunum during the experiment, and specifically demonstrated increased FD4 fluxes at d 2 to d 5 post-weaning. On the other hand, profound villus atrophy was observed during the weaning transition for all weaning treatments. Finally, LBW and NBW piglets did not demonstrate notable differences in GSH redox status, small intestinal barrier function and histo-morphology throughout the experiment. Conclusion Although moderate changes in the GSH redox system were observed upon weaning, the GSH redox status remained at a steady state level in 3w and 4w weaned piglets and was therefore not associated with weaning induced villus atrophy. Conversely, 3d3w weaned piglets demonstrated GSH redox imbalance in the small intestinal mucosa, and this co-occurred with a temporal malfunction of their intestinal barrier function.
Collapse
Affiliation(s)
- Jeroen Degroote
- 1Laboratory for Animal Nutrition and Animal Product Quality (LANUPRO), Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Block F, Campus Coupure, Coupure Links 653, 9000 Ghent, Belgium
| | - Hans Vergauwen
- 2Laboratory of Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Biomedical, Pharmaceutical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Belgium
| | - Wei Wang
- 1Laboratory for Animal Nutrition and Animal Product Quality (LANUPRO), Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Block F, Campus Coupure, Coupure Links 653, 9000 Ghent, Belgium
| | - Chris Van Ginneken
- 2Laboratory of Applied Veterinary Morphology, Department of Veterinary Sciences, Faculty of Biomedical, Pharmaceutical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Belgium
| | - Stefaan De Smet
- 1Laboratory for Animal Nutrition and Animal Product Quality (LANUPRO), Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Block F, Campus Coupure, Coupure Links 653, 9000 Ghent, Belgium
| | - Joris Michiels
- 1Laboratory for Animal Nutrition and Animal Product Quality (LANUPRO), Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Block F, Campus Coupure, Coupure Links 653, 9000 Ghent, Belgium
| |
Collapse
|
38
|
Garimano N, Amaral MM, Ibarra C. Endocytosis, Cytotoxicity, and Translocation of Shiga Toxin-2 Are Stimulated by Infection of Human Intestinal (HCT-8) Monolayers With an Hypervirulent E. coli O157:H7 Lacking stx2 Gene. Front Cell Infect Microbiol 2019; 9:396. [PMID: 31824869 PMCID: PMC6881261 DOI: 10.3389/fcimb.2019.00396] [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: 08/27/2019] [Accepted: 11/05/2019] [Indexed: 11/13/2022] Open
Abstract
Shiga toxin-producing Escherichia coli (STEC) strains are responsible for multiple clinical syndromes, including hemolytic uremic syndrome (HUS). E. coli O157:H7 is the most prevalent serotype associated with HUS and produces a variety of virulence factors being Stx2 the responsible of the most HUS severe cases. After intestinal colonization by STEC, Stx2 is released into the intestinal lumen, translocated to the circulatory system and then binds to its receptor, globotriaosylceramide (Gb3), in target cells. Thus, Stx2 passage through the colonic epithelial barrier is a key step in order to produce disease, being its mechanisms still poorly understood. We have previously reported that STEC interaction with the human colonic mucosa enhanced Stx2 production. In the present work, we have demonstrated that infection with O157:H7Δstx2, a mutant unable to produce Stx2, enhanced either Stx2 cytotoxicity on an intestinal cell line (HCT-8), or translocation across HCT-8 monolayers. Moreover, we found that translocation was enhanced by both paracellular and transcellular pathways. Using specific endocytosis inhibitors, we have further demonstrated that the main mechanisms implicated on Stx2 endocytosis and translocation, either when O157:H7Δstx2 was present or not, were Gb3-dependent, but dynamin-independent. On the other hand, dynamin dependent endocytosis and macropinocytosis became more relevant only when O157:H7Δstx2 infection was present. Overall, this study highlights the effects of STEC infection on the intestinal epithelial cell host and the mechanisms underlying Stx2 endocytosis, cytotoxic activity and translocation, in the aim of finding new tools toward a therapeutic approach.
Collapse
Affiliation(s)
- Nicolás Garimano
- Laboratorio de Fisiopatogenia, Departamento de Fisiología, Facultad de Medicina, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay-CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Marta Amaral
- Laboratorio de Fisiopatogenia, Departamento de Fisiología, Facultad de Medicina, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay-CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Cristina Ibarra
- Laboratorio de Fisiopatogenia, Departamento de Fisiología, Facultad de Medicina, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO Houssay-CONICET), Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
39
|
Ruffner MA, Song L, Maurer K, Shi L, Carroll MC, Wang JX, Muir AB, Spergel JM, Sullivan KE. Toll-like receptor 2 stimulation augments esophageal barrier integrity. Allergy 2019; 74:2449-2460. [PMID: 31267532 PMCID: PMC7083217 DOI: 10.1111/all.13968] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 04/26/2019] [Accepted: 05/13/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Germline-encoded innate immune pattern recognition receptors (PRR) are expressed at epithelial surfaces and modulate epithelial defenses. Evidence suggests that stimulation of the Toll-like receptor (TLR) family of PRR may regulate epithelial barrier integrity by upregulating tight junction (TJ) complex protein expression, but it is not known whether this mechanism is utilized in esophageal epithelial cells. TJ complex proteins maintain intact barrier function and are dysregulated in atopic disorders including eosinophilic esophagitis. METHODS Pattern recognition receptors expression was assessed in EoE and control primary esophageal epithelial cells, demonstrating robust expression of TLR2 and TLR3. The three-dimensional air-liquid interface culture (ALI) model was used to test whether TLR2 or TLR3 stimulation alters epithelial barrier function using an in vitro model of human epithelium. Transepithelial electrical resistance (TEER) and FITC-Dextran permeability were evaluated to assess membrane permeability. ALI cultures were evaluated by histology, immunohistochemistry, Western blotting, and chromatin immunoprecipitation (ChIP). RESULTS TLR3 stimulation did not change TEER in the ALI model. TLR2 stimulation increased TEER (1.28- to 1.31-fold) and decreased paracellular permeability to FITC-Dextran, and this effect was abolished by treatment with anti-TLR2 blocking antibody. TJ complex proteins claudin-1 and zonula occludens-1 were upregulated following TLR2 stimulation, and ChIP assay demonstrated altered histone 4 acetyl binding at the TJP1 enhancer and CLDN1 enhancer and promoter following zymosan treatment, implying the occurrence of durable chromatin changes. CONCLUSIONS Our findings implicate the TLR2 pathway as a potential regulator of esophageal epithelial barrier function and suggest that downstream chromatin modifications are associated with this effect.
Collapse
Affiliation(s)
- Melanie A Ruffner
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA, USA
| | - Li Song
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kelly Maurer
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lihua Shi
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Margaret C Carroll
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Joshua X Wang
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Amanda B Muir
- Department of Pediatrics, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jonathan M Spergel
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA, USA
| | - Kathleen E Sullivan
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
40
|
Cong X, Kong W. Endothelial tight junctions and their regulatory signaling pathways in vascular homeostasis and disease. Cell Signal 2019; 66:109485. [PMID: 31770579 DOI: 10.1016/j.cellsig.2019.109485] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/21/2019] [Accepted: 11/21/2019] [Indexed: 12/13/2022]
Abstract
Endothelial tight junctions (TJs) regulate the transport of water, ions, and molecules through the paracellular pathway, serving as an important barrier in blood vessels and maintaining vascular homeostasis. In endothelial cells (ECs), TJs are highly dynamic structures that respond to multiple external stimuli and pathological conditions. Alterations in the expression, distribution, and structure of endothelial TJs may lead to many related vascular diseases and pathologies. In this review, we provide an overview of the assessment methods used to evaluate endothelial TJ barrier function both in vitro and in vivo and describe the composition of endothelial TJs in diverse vascular systems and ECs. More importantly, the direct phosphorylation and dephosphorylation of TJ proteins by intracellular kinases and phosphatases, as well as the signaling pathways involved in the regulation of TJs, including and the protein kinase C (PKC), PKA, PKG, Ras homolog gene family member A (RhoA), mitogen-activated protein kinase (MAPK), phosphatidylinositol 3-kinase (PI3K)/Akt, and Wnt/β-catenin pathways, are discussed. With great advances in this area, targeting endothelial TJs may provide novel treatment for TJ-related vascular pathologies.
Collapse
Affiliation(s)
- Xin Cong
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China.
| | - Wei Kong
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China.
| |
Collapse
|
41
|
Das B, Okamoto K, Rabalais J, Kozan PA, Marchelletta RR, McGeough MD, Durali N, Go M, Barrett KE, Das S, Sivagnanam M. Enteroids expressing a disease-associated mutant of EpCAM are a model for congenital tufting enteropathy. Am J Physiol Gastrointest Liver Physiol 2019; 317:G580-G591. [PMID: 31433211 PMCID: PMC6879886 DOI: 10.1152/ajpgi.00098.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Congenital tufting enteropathy (CTE) is an autosomal recessive disease characterized by severe intestinal failure in infancy and mutations in the epithelial cell adhesion molecule (EPCAM) gene. Previous studies of CTE in mice expressing mutant EpCAM show neonatal lethality. Hence, to study the cellular, molecular, and physiological alterations that result from EpCAM mutation, a tamoxifen-inducible mutant EpCAM enteroid model has been generated. The presence of mutant EpCAM in the model was confirmed at both mRNA and protein levels. Immunofluorescence microscopy demonstrated the reduced expression of mutant EpCAM. Mutant enteroids had reduced budding potential as well as significantly decreased mRNA expression for epithelial lineage markers (Mucin 2, lysozyme, sucrase-isomaltase), proliferation marker Ki67, and secretory pathway transcription factors (Atoh1, Hnf1b). Significantly decreased numbers of Paneth and goblet cells were confirmed by staining. These findings were correlated with intestinal tissue from CTE patients and the mutant mice model that had significantly fewer Paneth and goblet cells than in healthy counterparts. FITC-dextran studies demonstrated significantly impaired barrier function in monolayers derived from mutant enteroids compared with control monolayers. In conclusion, we have established an ex vivo CTE model. The role of EpCAM in the budding potential, differentiation, and barrier function of enteroids is noted. Our study establishes new facets of EpCAM biology that will aid in understanding the pathophysiology of CTE and role of EpCAM in health and disease.NEW & NOTEWORTHY Here, we develop a novel ex vivo enteroid model for congenital tufting enteropathy (CTE) based on epithelial cell adhesion molecule (EPCAM) gene mutations found in patients. With this model we demonstrate the role of EpCAM in maintaining the functional homeostasis of the intestinal epithelium, including differentiation, proliferation, and barrier integrity. This study further establishes a new direction in EpCAM biology that will help in understanding the detailed pathophysiology of CTE and role of EpCAM.
Collapse
Affiliation(s)
- Barun Das
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Kevin Okamoto
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - John Rabalais
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Philip A. Kozan
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | | | - Matthew D. McGeough
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Nassim Durali
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Maria Go
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Kim E. Barrett
- 2Department of Medicine, University of California, San Diego, La Jolla, California
| | - Soumita Das
- 3Department of Pathology, University of California, San Diego, La Jolla, California
| | - Mamata Sivagnanam
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California,4Rady Children’s Hospital, San Diego, California
| |
Collapse
|
42
|
Antonescu IE, Rasmussen KF, Neuhoff S, Fretté X, Karlgren M, Bergström CAS, Nielsen CU, Steffansen B. The Permeation of Acamprosate Is Predominantly Caused by Paracellular Diffusion across Caco-2 Cell Monolayers: A Paracellular Modeling Approach. Mol Pharm 2019; 16:4636-4650. [DOI: 10.1021/acs.molpharmaceut.9b00733] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | | | | | | | - Maria Karlgren
- Department of Pharmacy, Uppsala University, P.O. Box 580, SE-751 23 Uppsala, Sweden
| | | | | | | |
Collapse
|
43
|
A unified in vitro test system for the assessment of tight junction modulators. Eur J Pharm Biopharm 2019; 142:353-363. [DOI: 10.1016/j.ejpb.2019.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/28/2019] [Accepted: 07/02/2019] [Indexed: 11/23/2022]
|
44
|
Kim N, Han DH, Suh MW, Lee JH, Oh SH, Park MK. Effect of lipopolysaccharide on diesel exhaust particle-induced junctional dysfunction in primary human nasal epithelial cells. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 248:736-742. [PMID: 30849591 DOI: 10.1016/j.envpol.2019.02.082] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 02/11/2019] [Accepted: 02/23/2019] [Indexed: 06/09/2023]
Abstract
OBJECTIVES Tight junctions (TJs) in the epithelium play a critical role in the formation of a paracellular epithelial barrier against the extracellular environment. Diesel exhaust particles (DEPs) disrupt the epithelial barrier. The aim of this study was to investigate how DEPs disrupt the epithelial barrier and whether Toll-like receptor 4 (TLR4) is involved in DEP-induced epithelial barrier dysfunction in primary human nasal epithelial (PHNE) cells. METHODS PHNE cells were cultured at an air-liquid interface (ALI) to create a fully differentiated in vivo-like model of the epithelium and then exposed to DEPs (particulate matter <4 μm) or lipopolysaccharide (LPS) alone (mono-exposure) and DEPs plus LPS (co-exposure) at the apical side of the PHNE. TJ formation and integrity were monitored by measuring transepithelial electric resistance (TEER) and fluorescently labeled dextran permeability. The expression of TJ proteins was assessed by confocal microscopy and a biochemical assay. RESULTS PHNE cell viability was reduced in a time- and dose-dependent manner following DEP exposure. TEER was significantly decreased at ALI day 20 but not at day 12 following DEP exposure. The dextran permeability of the PHNE was significantly increased at both ALI day 12 and day 20 following DEP exposure. The increased dextran permeability recovered to that of the control following co-exposure to DEPs plus LPS. In the presence of DEPs, the membrane expression of myosin light chain kinase (MLCK) was dramatically increased, and the expression of occludin, ZO1, claudin-1, and E-cadherin was significantly decreased. Co-exposure to DEPs plus LPS significantly reduced membrane MLCK, claudin-1, and E-cadherin but increased occludin and ZO1 expression at ALI day 12. CONCLUSION The activation of TLR4 by LPS inhibits MLCK trafficking to the plasma membrane, and this increased during DEP exposure, resulting in increased occludin expression at the plasma membrane that partially recovered TJ barrier dysfunction following DEP exposure.
Collapse
Affiliation(s)
- Nahyun Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Doo Hee Han
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Myung-Whan Suh
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea; Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Jun Ho Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea; Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Seung-Ha Oh
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea; Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Moo Kyun Park
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea; Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul, Republic of Korea.
| |
Collapse
|
45
|
Wegener J. Measuring the Permeability of Endothelial Cell Monolayers: Teaching New Tricks to an Old Dog. Biophys J 2019; 116:1377-1379. [PMID: 30975456 DOI: 10.1016/j.bpj.2019.03.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 02/06/2023] Open
Affiliation(s)
- Joachim Wegener
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, Regensburg, Germany; Fraunhofer Research Institution for Microsystems and Solid State Technologies EMFT, Munich, Germany.
| |
Collapse
|
46
|
Stone NE, Nunnally AE, Jimenez V, Cope EK, Sahl JW, Sheridan K, Hornstra HM, Vinocur J, Settles EW, Headley KC, Williamson CHD, Rideout JR, Bolyen E, Caporaso JG, Terriquez J, Monroy FP, Busch JD, Keim P, Wagner DM. Domestic canines do not display evidence of gut microbial dysbiosis in the presence of Clostridioides (Clostridium) difficile, despite cellular susceptibility to its toxins. Anaerobe 2019; 58:53-72. [PMID: 30946985 DOI: 10.1016/j.anaerobe.2019.03.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 03/11/2019] [Accepted: 03/25/2019] [Indexed: 12/14/2022]
Abstract
Clostridioides difficile infection (CDI) is an emerging public health threat and C. difficile is the most common cause of antimicrobial-associated diarrhea worldwide and the leading cause of hospital-associated infections in the US, yet the burden of community-acquired infections (CAI) is poorly understood. Characterizing C. difficile isolated from canines is important for understanding the role that canines may play in CAI. In addition, several studies have suggested that canines carry toxigenic C. difficile asymptomatically, which may imply that there are mechanisms responsible for resistance to CDI in canines that could be exploited to help combat human CDI. To assess the virulence potential of canine-derived C. difficile, we tested whether toxins TcdA and TcdB (hereafter toxins) derived from a canine isolate were capable of causing tight junction disruptions to colonic epithelial cells. Additionally, we addressed whether major differences exist between human and canine cells regarding C. difficile pathogenicity by exposing them to identical toxins. We then examined the canine gut microbiome associated with C. difficile carriage using 16S rRNA gene sequencing and searched for deviations from homeostasis as an indicator of CDI. Finally, we queried 16S rRNA gene sequences for bacterial taxa that may be associated with resistance to CDI in canines. Clostridioides difficile isolated from a canine produced toxins that reduced tight junction integrity in both human and canine cells in vitro. However, canine guts were not dysbiotic in the presence of C. difficile. These findings support asymptomatic carriage in canines and, furthermore, suggest that there are features of the gut microbiome and/or a canine-specific immune response that may protect canines against CDI. We identified two biologically relevant bacteria that may aid in CDI resistance in canines: 1) Clostridium hiranonis, which synthesizes secondary bile acids that have been shown to provide resistance to CDI in mice; and 2) Sphingobacterium faecium, which produces sphingophospholipids that may be associated with regulating homeostasis in the canine gut. Our findings suggest that canines may be cryptic reservoirs for C. difficile and, furthermore, that mechanisms of CDI resistance in the canine gut could provide insights into targeted therapeutics for human CDI.
Collapse
Affiliation(s)
- Nathan E Stone
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, 86011, USA.
| | - Amalee E Nunnally
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, 86011, USA.
| | - Victor Jimenez
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, 86011, USA.
| | - Emily K Cope
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, 86011, USA.
| | - Jason W Sahl
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, 86011, USA; Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, 86011, USA.
| | - Krystal Sheridan
- Translational Genomics Research Institute, Flagstaff, AZ, 86001, USA.
| | - Heidie M Hornstra
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, 86011, USA.
| | - Jacob Vinocur
- Northern Arizona Healthcare, Flagstaff Medical Center, Flagstaff, AZ, 86001, USA.
| | - Erik W Settles
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, 86011, USA; Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, 86011, USA.
| | - Kyle C Headley
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, 86011, USA.
| | - Charles H D Williamson
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, 86011, USA.
| | - Jai Ram Rideout
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, 86011, USA.
| | - Evan Bolyen
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, 86011, USA.
| | - J Gregory Caporaso
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, 86011, USA.
| | - Joel Terriquez
- Northern Arizona Healthcare, Flagstaff Medical Center, Flagstaff, AZ, 86001, USA.
| | - Fernando P Monroy
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, 86011, USA.
| | - Joseph D Busch
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, 86011, USA.
| | - Paul Keim
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, 86011, USA; Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, 86011, USA; Translational Genomics Research Institute, Flagstaff, AZ, 86001, USA
| | - David M Wagner
- Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, 86011, USA; Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, 86011, USA.
| |
Collapse
|
47
|
Søgaard PP, Ito N, Sato N, Fujita Y, Matter K, Itoh Y. Epithelial polarization in 3D matrix requires DDR1 signaling to regulate actomyosin contractility. Life Sci Alliance 2019; 2:2/1/e201800276. [PMID: 30760555 PMCID: PMC6374992 DOI: 10.26508/lsa.201800276] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/07/2019] [Accepted: 02/08/2019] [Indexed: 01/19/2023] Open
Abstract
For epithelial cells to establish epithelial polarity in a 3D matrix, signaling of a collagen receptor tyrosine kinase, DDR1, plays a crucial role. DDR1 signaling controls actomyosin contractility at the cell–cell junction through suppression of ROCK activity. Epithelial cells form sheets and tubules in various epithelial organs and establish apicobasal polarity and asymmetric vesicle transport to provide functionality in these structures. However, the molecular mechanisms that allow epithelial cells to establish polarity are not clearly understood. Here, we present evidence that the kinase activity of the receptor tyrosine kinase for collagen, discoidin domain receptor 1 (DDR1), is required for efficient establishment of epithelial polarity, proper asymmetric protein secretion, and execution of morphogenic programs. Lack of DDR1 protein or inhibition of DDR1 kinase activity disturbed tubulogenesis, cystogenesis, and the establishment of epithelial polarity and caused defects in the polarized localization of membrane-type 1 matrix metalloproteinase (MT1-MMP), GP135, primary cilia, laminin, and the Golgi apparatus. Disturbed epithelial polarity and cystogenesis upon DDR1 inhibition was caused by excess ROCK (rho-associated, coiled-coil-containing protein kinase)-driven actomyosin contractility, and pharmacological inhibition of ROCK was sufficient to correct these defects. Our data indicate that a DDR1-ROCK signaling axis is essential for the efficient establishment of epithelial polarity.
Collapse
Affiliation(s)
| | - Noriko Ito
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Nanami Sato
- Institute for Genetic Medicine, Division of Molecular Oncology, Hokkaido University, Sapporo, Japan
| | - Yasuyuki Fujita
- Institute for Genetic Medicine, Division of Molecular Oncology, Hokkaido University, Sapporo, Japan
| | - Karl Matter
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Yoshifumi Itoh
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
48
|
Intestinal Drug Absorption Enhancement by Aloe vera Gel and Whole Leaf Extract: In Vitro Investigations into the Mechanisms of Action. Pharmaceutics 2019; 11:pharmaceutics11010036. [PMID: 30669246 PMCID: PMC6359586 DOI: 10.3390/pharmaceutics11010036] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/12/2018] [Accepted: 12/13/2018] [Indexed: 12/30/2022] Open
Abstract
The co-administration of absorption enhancing agents with macromolecular drugs (e.g., protein and peptide drugs) has been identified as a means to improve the oral bioavailability of these drugs. Absorption-enhancing agents of natural origins have received a great deal of attention due to their sustainable production, in support of green chemistry. In previous studies, certain parts of the Aloe vera leaf (e.g., gel and whole leaf extract) have shown a potential to enhance drug permeation across the intestinal epithelial barrier. The mechanism of the drug-absorption-enhancement action and the capacity for absorption-enhancement of the A. vera gel and whole leaf, were investigated in this study. A clear decrease in transepithelial electrical resistance (TEER) of Caco-2 cell monolayers exposed to A. vera gel and wholeleaf extract, in various concentrations, indicated the opening of tight junctions between the epithelial cells. The transport of Fluorescein isothiocyanate (FITC)-dextran, with a molecular weight of 4 kDa (FD-4), could be enhanced across the Caco-2 cell monolayers, by the A. vera gel and whole-leaf extract, but not the FITC-dextran with larger molecular weights (i.e., 10, 20, and 40 kDa), which indicated a limited drug absorption enhancement capacity, in terms of the molecular size. Accumulation of FD-4 between the Caco-2 cells (and not within the cells), after treatment with the A. vera gel and whole-leaf extract was shown with a confocal laser scanning microscopy (CLSM) imaging, indicating that the paracellular transport of FD-4 occurred after the interaction of the A. vera gel and whole-leaf extract, with the epithelial cell monolayers. Furthermore, changes in the F-actin distribution in the cytoskeleton of the Caco-2 cell monolayers was observed by means of a fluorescence staining, which confirmed tight junction modulation as the mechanism of action for the absorption enhancement effect of the A. vera gel and whole-leaf extract.
Collapse
|
49
|
Feng R, Chen Q, Zhou P, Wang Y, Yan H. Nanoparticles based on disulfide-containing poly(β-amino ester) and zwitterionic fluorocarbon surfactant as a redox-responsive drug carrier for brain tumor treatment. NANOTECHNOLOGY 2018; 29:495101. [PMID: 30211689 DOI: 10.1088/1361-6528/aae122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Malignant brain tumors are often characterized by rapid growth, high invasiveness and poor prognosis. Current methods for brain tumor treatment are dramatically limited because of their inability to cross the blood-brain barrier (BBB) and enter the tumor cells. In this study, we prepared redox-responsive nanoparticles based on disulfide-containing poly(β-amino ester) (ssPBAE) and a zwitterionic fluorocarbon surfactant (Intechem-02) that has a carboxybetaine moiety in molecular structure, and preliminarily evaluated their potential as a drug carrier for brain tumor treatment. These nanoparticles, named as ssPBAEI, had a regular spherical shape and a small size below 50 nm with a relative narrow distribution. Doxorubicin (DOX), as a model chemotherapeutic drug, was efficiently encapsulated into ssPBAEI nanoparticles with a loading content of 25.4%. DOX-loaded ssPBAEI nanoparticles (ssPBAEI/DOX) showed significant redox-responsive in vitro release property and successfully carried DOX across a BBB model, monolayer of human brain capillary endothelial hCMEC/D3 cells. In human glioma LN229 cells, ssPBAEI/DOX nanoparticles were efficiently internalized and DOX was successfully released afterwards, thus significantly inhibited cell growth and induced cell apoptosis. In summary, this nanoparticle system based on ssPBAE and Intechem-02 showed a great potential as a drug carrier for brain tumor treatment.
Collapse
Affiliation(s)
- Ruoyang Feng
- Graduate School of Tianjin Medical University, Tianjin 300070, People's Republic of China
| | | | | | | | | |
Collapse
|
50
|
Shi F, Steuer A, Zhuang J, Kolb JF. Bioimpedance Analysis of Epithelial Monolayers after Exposure to Nanosecond Pulsed Electric Fields. IEEE Trans Biomed Eng 2018; 66:2010-2021. [PMID: 30452351 DOI: 10.1109/tbme.2018.2882299] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Exposures to pulsed electric fields (PEFs) are known to affect cell membranes and consequently also cell-cell interactions as well as associated characteristics. Bioimpedance analysis offers direct and non-invasive insights into structural and functional changes of cell membranes and extracellular matrices through a rigorous evaluation of electrical parameters. Accordingly, the multi-frequency impedance of confluent monolayers of rat liver epithelial WB-F344 cells was monitored in situ before and after exposure to nanosecond PEFs (nsPEFs). The results were fitted by two Cole models in series to obtain the Cole parameters for the monolayer. For an interpretation of the results, dielectric parameters, were correlated with changes of the TJ protein zonula occludens (ZO-1) and the paracellular permeability of the monolayer Cole parameters in general change as a function of pulse number and time. The findings demonstrate that impedance analysis is an effective method to monitor changes of TJs cell-cell contacts and paracellular permeability and relate them to exposure parameters.
Collapse
|