1
|
Chakraborty R, Ray P, Barik S, Banik O, Mahapatra C, Banoth E, Kumar P. A Review on Microengineering of Epithelial Barriers for Biomedical and Pharmaceutical Research. ACS APPLIED BIO MATERIALS 2024; 7:8107-8125. [PMID: 39565389 DOI: 10.1021/acsabm.4c00813] [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] [Indexed: 11/21/2024]
Abstract
Epithelial tissue forms a barrier around the human body and visceral organs, providing protection, permeation, sensation, and secretion. It is vital for our sustenance as it protects the tissue from harm and injury by restricting the entry of foreign bodies inside. Furthermore, it is a strong barrier to drugs, nutrients, and other essential deliverables. This layer also houses a large consortium of microbes, which thrive in tandem with human tissue, providing several health benefits. Moreover, the complex interplay of the microbiome with the barrier tissue is poorly understood. Therefore, replicating these barrier tissues on microdevices to generate physiological and pathophysiological models has been a huge interest for researchers over the last few decades. The artificially engineered reconstruction of these epithelial cellular barriers on microdevices could help underpin the host-microbe interaction, generating a physiological understanding of the tissue, tissue remodeling, receptor-based selective diffusion, drug testing, and others. In addition, these devices could reduce the burden of animal sacrifices for similar research and minimize the failure rate in drug discovery due to the use of primary human cells and others. This review discusses the nature of the epithelial barrier at different tissue sites, the recent developments in creating engineered barrier models, and their applications in pathophysiology, host-microbe interactions, drug discovery, and cytotoxicity. The review aims to provide know-how and knowledge behind engineered epithelial barrier tissue to bioengineers, biotechnologists, and scientists in allied fields.
Collapse
Affiliation(s)
- Ruchira Chakraborty
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Pragyan Ray
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Swagatika Barik
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
- Opto-Biomedical Microsystem Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Sector-1, Rourkela 769008, Odisha, India
| | - Oindrila Banik
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
- Opto-Biomedical Microsystem Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Sector-1, Rourkela 769008, Odisha, India
| | - Chinmaya Mahapatra
- Department of Biotechnology, National Institute of Technology, Raipur-492010 Chhattisgarh, India
| | - Earu Banoth
- Opto-Biomedical Microsystem Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Sector-1, Rourkela 769008, Odisha, India
| | - Prasoon Kumar
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
| |
Collapse
|
2
|
Huang X, Fei Y, Qiu X, Qian T, Shang Q, Cui J, Song Y, Sheng S, Xiao W, Yu Q, Wang T, Wang X. MiR-625-5p is a potential therapeutic target in sepsis by regulating CXCL16/CXCR6 axis and endothelial barrier. Int Immunopharmacol 2024; 137:112508. [PMID: 38889512 DOI: 10.1016/j.intimp.2024.112508] [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: 01/01/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND MicroRNA plays an important role in the progression of sepsis. We found a significant increase of in miR-625-5p expression in the blood of patients with sepsis, and lipopolysaccharide (LPS)-stimulated EA.hy926 cells. To date, little is known about the specific biological function of miR-625-5p in sepsis. METHODS Changes in miR-625-5p expression were verified through quantitative real-time polymerase chain reaction in 45 patients with sepsis or septic shock and 30 healthy subjects. In vitro, EA.hy926 cells were treated with LPS. Transendothelial electrical resistance assay and FITC-dextran were used in evaluating endothelial barrier function. RESULTS Herein, patients with sepsis or septic shock had significantly higher miR-625-5p expression levels, chemokine (C-X-C motif) ligand 16 (CXCL16) levels, and glycocalyx components than the healthy controls, and miR-625-5p level was positively correlated with disease. Kaplan-Meier analysis demonstrated a strong association between miR-625-5p level and 28-day mortality. Furthermore, the miR-625-5p inhibitor significantly alleviated LPS-induced endothelial barrier injury in vitro. Then, miR-625-5p positively regulated CXCL16 and down-regulated miR-625-5p attenuated CXCL16 transcription and expression in EA.hy926 cells. CXCL16 knockout significantly alleviated vascular barrier dysfunction in the LPS-induced EA.hy926 cells. sCXCL16 treatment in EA.hy926 cells significantly increased endothelial hyperpermeability by disrupting endothelial glycocalyx, tight junction proteins, and adherens junction proteins through the modulation of C-X-C chemokine receptor type 6 (CXCR6). CONCLUSIONS Increase in miR-625-5p level may be an effective biomarker for predicting 28-day mortality in patients with sepsis/septic shock. miR-625-5p is a critical pathogenic factor for endothelial barrier dysfunction in LPS-induced EA.hy926 cells because it activates the CXCL16/CXCR6 axis.
Collapse
Affiliation(s)
- Xiao Huang
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yuxin Fei
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xiaoyu Qiu
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Pulmonary and Critical Care Medicine, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Tiantian Qian
- Department of Respiratory Medicine, Ji'nan Zhangqiu District People's Hospital, No. 1920 Mingshuihuiquan Road, Ji'nan, 250200, Shandong, China
| | - Quanmei Shang
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jinfeng Cui
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yutong Song
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Shurui Sheng
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Wenhan Xiao
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Qilin Yu
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Tao Wang
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Xiaozhi Wang
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
3
|
Zhu W, Liu X, Luo L, Huang X, Wang X. Interaction between mitochondrial homeostasis and barrier function in lipopolysaccharide-induced endothelial cell injury. Int J Exp Pathol 2023; 104:272-282. [PMID: 37828780 PMCID: PMC10652695 DOI: 10.1111/iep.12495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/20/2023] [Accepted: 09/24/2023] [Indexed: 10/14/2023] Open
Abstract
This study aimed to investigate the effects of mitochondrial homeostasis on lipopolysaccharide (LPS)-induced endothelial cell barrier function and the mechanisms that underlie these effects. Cells were treated with LPS or oligomycin (mitochondrial adenosine triphosphate synthase inhibitor) and the mitochondrial morphology, mitochondrial reactive oxygen species (mtROS), and mitochondrial membrane potential (ΔΨm) were evaluated. Moreover, the shedding of glycocalyx-heparan sulphate (HS), the levels of HS-specific degrading enzyme heparanase (HPA), and the expression of occludin and zonula occludens (ZO-1) of Tight Junctions (TJ)s, which are mediated by myosin light chain phosphorylation (p-MLC), were assessed. Examining the changes in mitochondrial homeostasis showed that adding heparinase III, which is an exogenous HPA, can destroy the integrity of glycocalyx. LPS simultaneously increased mitochondrial swelling, mtROS, and ΔΨm. Without oligomycin effects, HS, HPA levels, and p-MLC were found to be elevated, and the destruction of occludin and ZO-1 increased. Heparinase III not only damaged the glycocalyx by increasing HS shedding but also increased mitochondrial swelling and mtROS and decreased ΔΨm. Mitochondrial homeostasis is involved in LPS-induced endothelial cell barrier dysfunction by aggravating HPA and p-MLC levels. In turn, the integrated glycocalyx protects mitochondrial homeostasis.
Collapse
Affiliation(s)
- Weiwei Zhu
- Department of Intensive Care UnitBinzhou Medical University HospitalBinzhouChina
| | - Xiaojing Liu
- Department of Intensive Care UnitBinzhou Medical University HospitalBinzhouChina
| | - Liqing Luo
- Department of HematologyBinzhou Medical University HospitalBinzhouChina
| | - Xiao Huang
- Department of Intensive Care UnitBinzhou Medical University HospitalBinzhouChina
| | - Xiaozhi Wang
- Department of Intensive Care UnitBinzhou Medical University HospitalBinzhouChina
| |
Collapse
|
4
|
Zimmermann CS, Snow RF, Wilson-Mifsud B, LeRoy K, Boeckman C, Huang E, Mathesius CA, Roper JM, Hurley BP. Evaluation of an in vitro experimental platform of human polarized intestinal epithelial monolayers for the hazard assessment of insecticidal proteins. Food Chem Toxicol 2023; 181:114106. [PMID: 37852351 DOI: 10.1016/j.fct.2023.114106] [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: 08/31/2023] [Revised: 10/13/2023] [Accepted: 10/14/2023] [Indexed: 10/20/2023]
Abstract
Previous work demonstrated the utility of using human-derived intestinal epithelial cell (IEC) lines cultured as polarized monolayers on Transwell® filters to differentiate between hazardous and non-hazardous proteins. The current study seeks to further resolve appropriate concentrations for evaluating proteins of unknown hazard potential using the IEC experimental platform and leverages these parameters for evaluating the potential toxicity of insecticidal proteins characteristic of those expressed in genetically modified (GM) agricultural biotechnology crops. To establish optimal test protein concentrations, effects of several known hazardous (C. perfringens epsilon toxin, Listeriolysin O, Phaseolus vulgaris erythroagglutinin, E. coli Shiga toxin 1, C. difficile Toxin B and wheat germ agglutinin) and non-hazardous (Ara-h2, β-lactoglobulin, fibronectin and Rubisco) proteins on IEC barrier integrity and cell viability were evaluated at concentration ranges. Two insecticidal proteins (AfIP-1A and AfIP-1B) were evaluated for effects in the IEC assay, a seven-day insecticidal bioassay, and assessed in a high-dose 14-day acute oral toxicity study in mice. The results obtained from the human in vitro IEC assay were consistent with results obtained from an in vivo acute oral toxicity study, both demonstrating that the combination of AfIP-1A and AfIP-1B do not exhibit any identifiable harmful impacts on mammalian cells.
Collapse
Affiliation(s)
| | - Ryan F Snow
- Mucosal Immunology & Biology Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Brittany Wilson-Mifsud
- Mucosal Immunology & Biology Research Center, Massachusetts General Hospital, Boston, MA, USA
| | | | | | | | | | | | - Bryan P Hurley
- Mucosal Immunology & Biology Research Center, Massachusetts General Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Gut as the target tissue of mercury and the extraintestinal effects. Toxicology 2023; 484:153396. [PMID: 36521575 DOI: 10.1016/j.tox.2022.153396] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/09/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022]
Abstract
Mercury (Hg) is harmful to the environment and human health. The gut plays important roles as the biological, chemical, mechanical, and immune barriers in animals and human beings. It has been known that Hg can be absorbed and methylated/demethylated in the gut, on the other hand, the impacts of Hg to the gut (especially the gut microbiota) is less studied. This review paper summarizes the impacts of inorganic Hg (IHg) and methyl Hg (MeHg) on gut barriers and the extraintestinal effects (damage to other organs such as the liver and brain). Both IHg and MeHg were found to cause intestinal microbial disorders, abnormal metabolites production, tight junction damage, and immune responses in the gut. The damage to the gut also contributed to the extraintestinal effects like the hepatotoxicity by IHg and the neurotoxicity by MeHg. In all, it is proposed that the gut should be considered as an important target tissue of Hg exposure, and the regulation of gut microbiota may have the potential for the prevention and control of the toxicity of Hg.
Collapse
|
6
|
PLD2 deletion alleviates disruption of tight junctions in sepsis-induced ALI by regulating PA/STAT3 phosphorylation pathway. Int Immunopharmacol 2023; 114:109561. [PMID: 36700766 DOI: 10.1016/j.intimp.2022.109561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Increased inflammatory exudation caused by endothelium and endothelial junction damage is a typical pathological feature of acute respiratory distress syndrome/acute lung injury (ARDS/ALI). Previous studies have shown that phospholipase D2 (PLD2) can increase the inflammatory response and has a close relationship with the severity of sepsis-induced ALI and the mortality of sepsis, but its mechanism is unknown. This study explored the effect and mechanism of PLD2 deletion on the structure and function of endothelial tight junction (TJ) in lipopolysaccharide (LPS)-induced ALI. METHODS We used C57BL/6 mice (wild-type and PLD2 knockout (PLD2-/-)) and human umbilical vein endothelial cell (HUVEC) models of sepsis-ALI. The pathological changes were evaluated by hematoxylin-eosin staining. Pulmonary vascular permeability was detected using wet-dry ratio, fluorescein isothiocyanate (FITC)-dextran, FITC-albumin, and immunoglobulin M concentration of bronchoalveolar lavage fluid. FITC-dextran and trans-endothelial electrical resistance assay were used to evaluate endothelial permeability on LPS-stimulated HUVECs. The mRNA expressions of TJ proteins were detected by real-time quantitative polymerase chain reaction. Then, protein levels were detected through Western blot analysis and immunofluorescence. The content of phosphatidic acid (PA), a downstream product of PLD2, was detected using an enzyme-linked immunosorbent assay kit. RESULTS PLD2 deficiency not only alleviated lung histopathological changes and improved pulmonary vascular permeability but also increased the survival rate of ALI mice. Knockout of PLD2 or treatment with the PLD2 inhibitor can reduce the damage of endothelial TJ proteins, namely, claudin5, occludin and zonula occludens protein-1, in sepsis-ALI mice and LPS-stimulated HUVECs. The level of the PLD2 catalytic product PA increased in LPS-stimulated HUVECs, and exogenous PA can reduce the TJ protein expression and increase signal transducer and activator of transcription 3 (STAT3) phosphorylation in vitro. Inhibition of STAT3 phosphorylation attenuated PA-induced degradation of endothelial TJs. CONCLUSION PLD2 knockout or inhibition may protect against LPS-induced lung injury by regulating the PA/STAT3 phosphorylation/endothelial TJ axis.
Collapse
|
7
|
Mullins E, Bresson J, Dalmay T, Dewhurst IC, Epstein MM, Firbank LG, Guerche P, Hejatko J, Moreno FJ, Nogue F, Rostoks N, Sanchez Serrano JJ, Savoini G, Veromann E, Veronesi F, Casacuberta J, Zurbriggen MD, Fernandez A, Gomez Ruiz JA, Gennaro A, Papadopoulou N, Lanzoni A, Naegeli H. Evaluation of existing guidelines for their adequacy for the food and feed risk assessment of genetically modified plants obtained through synthetic biology. EFSA J 2022; 20:e07410. [PMID: 35873722 PMCID: PMC9297787 DOI: 10.2903/j.efsa.2022.7410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Synthetic biology (SynBio) is an interdisciplinary field at the interface of molecular engineering and biology aiming to develop new biological systems and impart new functions to living cells, tissues and organisms. EFSA has been asked by the European Commission to evaluate SynBio developments in agri-food with the aim of identifying the adequacy and sufficiency of existing guidelines for risk assessment and determine if updated guidance is needed. In this context, the GMO Panel has previously adopted an Opinion evaluating the SynBio developments in agri-food/feed and the adequacy and sufficiency of existing guidelines for the molecular characterisation and environmental risk assessment of genetically modified plants (GMPs) obtained through SynBio and reaching the market in the next decade. Complementing the above, in this Opinion, the GMO Panel evaluated the adequacy and sufficiency of existing guidelines for the food and feed risk assessment of GMPs obtained through SynBio. Using selected hypothetical case studies, the GMO Panel did not identify novel potential hazards and risks that could be posed by food and feed from GMPs obtained through current and near future SynBio approaches; considers that the existing guidelines are adequate and sufficient in some Synbio applications; in other cases, existing guidelines may be just adequate and hence need updating; areas needing updating include those related to the safety assessment of new proteins and the comparative analysis. The GMO Panel recommends that future guidance documents provide indications on how to integrate the knowledge available from the SynBio design and modelling in the food and feed risk assessment and encourages due consideration to be given to food and feed safety aspects throughout the SynBio design process as a way to facilitate the risk assessment of SynBio GMPs and reduce the amount of data required.
Collapse
|
8
|
Mullins E, Bresson J, Dalmay T, Dewhurst IC, Epstein MM, George Firbank L, Guerche P, Hejatko J, Naegeli H, Nogué F, Rostoks N, Sánchez Serrano JJ, Savoini G, Veromann E, Veronesi F, Fernandez Dumont A, Moreno FJ. Scientific Opinion on development needs for the allergenicity and protein safety assessment of food and feed products derived from biotechnology. EFSA J 2022; 20:e07044. [PMID: 35106091 PMCID: PMC8787593 DOI: 10.2903/j.efsa.2022.7044] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
This Scientific Opinion addresses the formulation of specific development needs, including research requirements for allergenicity assessment and protein safety, in general, which is urgently needed in a world that demands more sustainable food systems. Current allergenicity risk assessment strategies are based on the principles and guidelines of the Codex Alimentarius for the safety assessment of foods derived from 'modern' biotechnology initially published in 2003. The core approach for the safety assessment is based on a 'weight-of-evidence' approach because no single piece of information or experimental method provides sufficient evidence to predict allergenicity. Although the Codex Alimentarius and EFSA guidance documents successfully addressed allergenicity assessments of single/stacked event GM applications, experience gained and new developments in the field call for a modernisation of some key elements of the risk assessment. These should include the consideration of clinical relevance, route of exposure and potential threshold values of food allergens, the update of in silico tools used with more targeted databases and better integration and standardisation of test materials and in vitro/in vivo protocols. Furthermore, more complex future products will likely challenge the overall practical implementation of current guidelines, which were mainly targeted to assess a few newly expressed proteins. Therefore, it is timely to review and clarify the main purpose of the allergenicity risk assessment and the vital role it plays in protecting consumers' health. A roadmap to (re)define the allergenicity safety objectives and risk assessment needs will be required to inform a series of key questions for risk assessors and risk managers such as 'what is the purpose of the allergenicity risk assessment?' or 'what level of confidence is necessary for the predictions?'.
Collapse
|
9
|
Nickerson KP, Llanos-Chea A, Ingano L, Serena G, Miranda-Ribera A, Perlman M, Lima R, Sztein MB, Fasano A, Senger S, Faherty CS. A Versatile Human Intestinal Organoid-Derived Epithelial Monolayer Model for the Study of Enteric Pathogens. Microbiol Spectr 2021; 9:e0000321. [PMID: 34106568 PMCID: PMC8552518 DOI: 10.1128/spectrum.00003-21] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 01/09/2023] Open
Abstract
Gastrointestinal infections cause significant morbidity and mortality worldwide. The complexity of human biology and limited insights into host-specific infection mechanisms are key barriers to current therapeutic development. Here, we demonstrate that two-dimensional epithelial monolayers derived from human intestinal organoids, combined with in vivo-like bacterial culturing conditions, provide significant advancements for the study of enteropathogens. Monolayers from the terminal ileum, cecum, and ascending colon recapitulated the composition of the gastrointestinal epithelium, in which several techniques were used to detect the presence of enterocytes, mucus-producing goblet cells, and other cell types following differentiation. Importantly, the addition of receptor activator of nuclear factor kappa-B ligand (RANKL) increased the presence of M cells, critical antigen-sampling cells often exploited by enteric pathogens. For infections, bacteria were grown under in vivo-like conditions known to induce virulence. Overall, interesting patterns of tissue tropism and clinical manifestations were observed. Shigella flexneri adhered efficiently to the cecum and colon; however, invasion in the colon was best following RANKL treatment. Both Salmonella enterica serovars Typhi and Typhimurium displayed different infection patterns, with S. Typhimurium causing more destruction of the terminal ileum and S. Typhi infecting the cecum more efficiently than the ileum, particularly with regard to adherence. Finally, various pathovars of Escherichia coli validated the model by confirming only adherence was observed with these strains. This work demonstrates that the combination of human-derived tissue with targeted bacterial growth conditions enables powerful analyses of human-specific infections that could lead to important insights into pathogenesis and accelerate future vaccine development. IMPORTANCE While traditional laboratory techniques and animal models have provided valuable knowledge in discerning virulence mechanisms of enteric pathogens, the complexity of the human gastrointestinal tract has hindered our understanding of physiologically relevant, human-specific interactions; and thus, has significantly delayed successful vaccine development. The human intestinal organoid-derived epithelial monolayer (HIODEM) model closely recapitulates the diverse cell populations of the intestine, allowing for the study of human-specific infections. Differentiation conditions permit the expansion of various cell populations, including M cells that are vital to immune recognition and the establishment of infection by some bacteria. We provide details of reproducible culture methods and infection conditions for the analyses of Shigella, Salmonella, and pathogenic Escherichia coli in which tissue tropism and pathogen-specific infection patterns were detected. This system will be vital for future studies that explore infection conditions, health status, or epigenetic differences and will serve as a novel screening platform for therapeutic development.
Collapse
Affiliation(s)
- Kourtney P. Nickerson
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Alejandro Llanos-Chea
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Laura Ingano
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Gloria Serena
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Alba Miranda-Ribera
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Meryl Perlman
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Rosiane Lima
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Marcelo B. Sztein
- Center for Vaccine Development and Global Health, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Alessio Fasano
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Stefania Senger
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Christina S. Faherty
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Naegeli H, Bresson J, Dalmay T, Dewhurst IC, Epstein MM, Firbank LG, Guerche P, Hejatko J, Moreno FJ, Mullins E, Nogué F, Rostoks N, Sánchez Serrano JJ, Savoini G, Veromann E, Veronesi F, Dumont AF. Statement on in vitro protein digestibility tests in allergenicity and protein safety assessment of genetically modified plants. EFSA J 2021; 19:e06350. [PMID: 33473251 PMCID: PMC7801955 DOI: 10.2903/j.efsa.2021.6350] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This statement supplements and updates the GMO Panel guidance document on allergenicity of genetically modified (GM) plants published in 2017. In that guidance document, the GMO Panel considered that additional investigations on in vitro protein digestibility were needed before providing any additional recommendations in the form of guidance to applicants. Thus, an interim phase was proposed to assess the utility of an enhanced in vitro digestion test, as compared to the classical pepsin resistance test. Historically, resistance to degradation by pepsin using the classical pepsin resistance test has been considered as additional information, in a weight-of-evidence approach, for the assessment of allergenicity and toxicity of newly expressed proteins in GM plants. However, more recent evidence does not support this test as a good predictor of allergenic potential for hazard. Furthermore, there is a need for more reliable systems to predict the fate of the proteins in the gastrointestinal tract and how they interact with the relevant human cells. Nevertheless, the classical pepsin resistance test can still provide some information on the physicochemical properties of novel proteins relating to their stability under acidic conditions. But other methods can be used to obtain data on protein's structural and/or functional integrity. It is acknowledged that the classical pepsin resistance test is embedded into international guidelines, e.g. Codex Alimentarius and Regulation (EU) No 503/2013. For future development, a deeper understanding of protein digestion in the gastrointestinal tract could enable the framing of more robust strategies for the safety assessment of proteins. Given the high complexity of the digestion and absorption process of dietary proteins, it is needed to clarify and identify the aspects that could be relevant to assess potential risks of allergenicity and toxicity of proteins. To this end, a series of research questions to be addressed are also formulated in this statement.
Collapse
|
11
|
Huang WK, Xie C, Young RL, Zhao JB, Ebendorff-Heidepriem H, Jones KL, Rayner CK, Wu TZ. Development of innovative tools for investigation of nutrient-gut interaction. World J Gastroenterol 2020; 26:3562-3576. [PMID: 32742126 PMCID: PMC7366065 DOI: 10.3748/wjg.v26.i25.3562] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 05/29/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023] Open
Abstract
The gastrointestinal tract is the key interface between the ingesta and the human body. There is wide recognition that the gastrointestinal response to nutrients or bioactive compounds, particularly the secretion of numerous hormones, is critical to the regulation of appetite, body weight and blood glucose. This concept has led to an increasing focus on “gut-based” strategies for the management of metabolic disorders, including type 2 diabetes and obesity. Understanding the underlying mechanisms and downstream effects of nutrient-gut interactions is fundamental to effective translation of this knowledge to clinical practice. To this end, an array of research tools and platforms have been developed to better understand the mechanisms of gut hormone secretion from enteroendocrine cells. This review discusses the evolution of in vitro and in vivo models and the integration of innovative techniques that will ultimately enable the development of novel therapies for metabolic diseases.
Collapse
Affiliation(s)
- Wei-Kun Huang
- Adelaide Medical School, Centre of Research Excellence in Translating Nutritional Science to Good Health, the University of Adelaide, Adelaide, SA 5005, Australia
- Institute for Photonics and Advanced Sensing, School of Physical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
- The ARC Centre of Excellence for Nanoscale BioPhotonics, Adelaide, SA 5005, Australia
| | - Cong Xie
- Adelaide Medical School, Centre of Research Excellence in Translating Nutritional Science to Good Health, the University of Adelaide, Adelaide, SA 5005, Australia
| | - Richard L Young
- Adelaide Medical School, Centre of Research Excellence in Translating Nutritional Science to Good Health, the University of Adelaide, Adelaide, SA 5005, Australia
- Diabetes, Nutrition and Gut Health, Lifelong Health, South Australia Health and Medical Research Institute, Adelaide, SA 5005, Australia
| | - Jiang-Bo Zhao
- Institute for Photonics and Advanced Sensing, School of Physical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
- The ARC Centre of Excellence for Nanoscale BioPhotonics, Adelaide, SA 5005, Australia
| | - Heike Ebendorff-Heidepriem
- Institute for Photonics and Advanced Sensing, School of Physical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
- The ARC Centre of Excellence for Nanoscale BioPhotonics, Adelaide, SA 5005, Australia
| | - Karen L Jones
- Adelaide Medical School, Centre of Research Excellence in Translating Nutritional Science to Good Health, the University of Adelaide, Adelaide, SA 5005, Australia
| | - Christopher K Rayner
- Adelaide Medical School, Centre of Research Excellence in Translating Nutritional Science to Good Health, the University of Adelaide, Adelaide, SA 5005, Australia
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Tong-Zhi Wu
- Adelaide Medical School, Centre of Research Excellence in Translating Nutritional Science to Good Health, the University of Adelaide, Adelaide, SA 5005, Australia
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing 210009, Jiangsu Province, China
| |
Collapse
|
12
|
Lanter BB, Eaton AD, Roper JM, Zimmermann C, Delaney B, Hurley BP. Single versus repeated exposure to human polarized intestinal epithelial monolayers for in vitro protein hazard characterization. Food Chem Toxicol 2019; 132:110666. [PMID: 31288052 DOI: 10.1016/j.fct.2019.110666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/03/2019] [Accepted: 07/04/2019] [Indexed: 11/25/2022]
Abstract
Recent studies suggest human-derived intestinal epithelial cell (IEC) lines cultured as polarized monolayers on permeable Transwell® filters are effective at differentiating between hazardous and non-hazardous proteins following a single exposure. In this study, IEC polarized monolayers were subjected to hazardous or non-hazardous proteins in nine exposures over 30 days and compared to a single exposure of the same protein. The objective was to evaluate whether repeated exposures to a protein differently alter barrier integrity or compromise cell viability compared to single exposures. Proteins tested included Clostridium difficile toxin A, Streptolysin O, Wheat Germ Agglutinin, Phaseolus vulgaris Hemagglutinin-E, bovine serum albumin, porcine serum albumin, and fibronectin. Evidence of diminished barrier integrity and/or cell viability following exposure to hazardous proteins was more pronounced in magnitude when IECs were subjected to multiple rather than single exposures. In some cases, an effect on IEC monolayers was observed only with repeated exposures. In general, IEC responses to non-hazardous proteins following either single or repeated exposures were minimal. Results from these studies support the utility of using cultured human IEC polarized monolayers to differentiate between hazardous and non-hazardous proteins and suggest that repeated exposures may reveal a greater magnitude of response when compared to single exposures.
Collapse
Affiliation(s)
- B B Lanter
- Department of Pediatrics, Mucosal Immunology & Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, United States
| | - A D Eaton
- Department of Pediatrics, Mucosal Immunology & Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, United States
| | - J M Roper
- Corteva Agriscience, Newark, DE, 19711, United States
| | - C Zimmermann
- Corteva Agriscience, Johnston, IA, 50131, United States.
| | - B Delaney
- Corteva Agriscience, Johnston, IA, 50131, United States
| | - B P Hurley
- Department of Pediatrics, Mucosal Immunology & Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, United States.
| |
Collapse
|
13
|
Dutton JS, Hinman SS, Kim R, Wang Y, Allbritton NL. Primary Cell-Derived Intestinal Models: Recapitulating Physiology. Trends Biotechnol 2018; 37:744-760. [PMID: 30591184 DOI: 10.1016/j.tibtech.2018.12.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 12/02/2018] [Accepted: 12/03/2018] [Indexed: 02/07/2023]
Abstract
The development of physiologically relevant intestinal models fueled by breakthroughs in primary cell-culture methods has enabled successful recapitulation of key features of intestinal physiology. These advances, paired with engineering methods, for example incorporating chemical gradients or physical forces across the tissues, have yielded ever more sophisticated systems that enhance our understanding of the impact of the host microbiome on human physiology as well as on the genesis of intestinal diseases such as inflammatory bowel disease and colon cancer. In this review we highlight recent advances in the development and usage of primary cell-derived intestinal models incorporating monolayers, organoids, microengineered platforms, and macrostructured systems, and discuss the expected directions of the field.
Collapse
Affiliation(s)
- Johanna S Dutton
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, NC, USA
| | - Samuel S Hinman
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Raehyun Kim
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, NC, USA
| | - Yuli Wang
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Nancy L Allbritton
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, NC, USA; Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
14
|
Jiang X, Gu S, Liu D, Zhao L, Xia S, He X, Chen H, Ge J. Lactobacillus brevis 23017 Relieves Mercury Toxicity in the Colon by Modulation of Oxidative Stress and Inflammation Through the Interplay of MAPK and NF-κB Signaling Cascades. Front Microbiol 2018; 9:2425. [PMID: 30369917 PMCID: PMC6194351 DOI: 10.3389/fmicb.2018.02425] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/21/2018] [Indexed: 02/06/2023] Open
Abstract
Aims: Lactobacillus strains have protective effects against heavy metals while relieving oxidative stress and modulating the immune response. Mechanisms that ameliorate heavy metal toxicity and the relationship between probiotics and gut barrier protection in the process of heavy metal pathogenesis was poorly understood. Methods and Results: In this study, Lactobacillus brevis 23017 (LAB, L. brevis 23017), a selected probiotics strain with strong mercury binding capacities, was applied to evaluate the efficiency against mercury toxicity in a mouse model. Histopathological results along with HE stains show that L. brevis 23017 protects the integrity of the small intestinal villus, which slows weight loss in response to Hg exposure. The qRT-PCR results demonstrate that L. brevis 23017 maintains a normal mucosal barrier via modulation of tight junction proteins. Importantly, the present study demonstrates that L. brevis 23017 effectively ameliorates injury of the small intestine by reducing intestinal inflammation and alleviating oxidative stress in animal models. Moreover, L. brevis 23017 blocks oxidative stress and inflammation through MAPK and NF-κB pathways, as shown by western blot. Conclusions: Together, these results reveal that L. brevis 23017 may have applications in the prevention and treatment of oral Hg exposure with fermented functional foods by protecting gut health in daily life.
Collapse
Affiliation(s)
- Xinpeng Jiang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Shanshan Gu
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Di Liu
- Key Laboratory of Combining Farming and Animal Husbandry, Ministry of Agriculture, Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, Harbin, China
| | - Lili Zhao
- Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, State Key Laboratory of Veterinary Biotechnology, Chinese Academy of Agricultural Sciences, Harbin Veterinary Research Institute, Harbin, China
| | - Shuang Xia
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xinmiao He
- Key Laboratory of Combining Farming and Animal Husbandry, Ministry of Agriculture, Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, Harbin, China
| | - Hongyan Chen
- Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, State Key Laboratory of Veterinary Biotechnology, Chinese Academy of Agricultural Sciences, Harbin Veterinary Research Institute, Harbin, China
| | - Junwei Ge
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
15
|
Zimmermann C, Eaton A, Lanter B, Roper J, Hurley B, Delaney B. Extended exposure duration of cultured intestinal epithelial cell monolayers in characterizing hazardous and non-hazardous proteins. Food Chem Toxicol 2018; 115:451-459. [DOI: 10.1016/j.fct.2018.03.047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/27/2018] [Accepted: 03/30/2018] [Indexed: 01/08/2023]
|
16
|
Ayehunie S, Landry T, Stevens Z, Armento A, Hayden P, Klausner M. Human Primary Cell-Based Organotypic Microtissues for Modeling Small Intestinal Drug Absorption. Pharm Res 2018; 35:72. [PMID: 29476278 PMCID: PMC6599640 DOI: 10.1007/s11095-018-2362-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 02/02/2018] [Indexed: 12/11/2022]
Abstract
PURPOSE The study evaluates the use of new in vitro primary human cell-based organotypic small intestinal (SMI) microtissues for predicting intestinal drug absorption and drug-drug interaction. METHODS The SMI microtissues were reconstructed using human intestinal fibroblasts and enterocytes cultured on a permeable support. To evaluate the suitability of the intestinal microtissues to model drug absorption, the permeability coefficients across the microtissues were determined for a panel of 11 benchmark drugs with known human absorption and Caco-2 permeability data. Drug-drug interactions were examined using efflux transporter substrates and inhibitors. RESULTS The 3D-intestinal microtissues recapitulate the structural features and physiological barrier properties of the human small intestine. The microtissues also expressed drug transporters and metabolizing enzymes found on the intestinal wall. Functionally, the SMI microtissues were able to discriminate between low and high permeability drugs and correlated better with human absorption data (r2 = 0.91) compared to Caco-2 cells (r2 = 0.71). Finally, the functionality of efflux transporters was confirmed using efflux substrates and inhibitors which resulted in efflux ratios of >2.0 fold and by a decrease in efflux ratios following the addition of inhibitors. CONCLUSION The SMI microtissues appear to be a useful pre-clinical tool for predicting drug bioavailability of orally administered drugs.
Collapse
Affiliation(s)
- Seyoum Ayehunie
- MatTek Corporation, 200 Homer Avenue, Ashland, Massachusetts, USA.
| | - Tim Landry
- MatTek Corporation, 200 Homer Avenue, Ashland, Massachusetts, USA
| | - Zachary Stevens
- MatTek Corporation, 200 Homer Avenue, Ashland, Massachusetts, USA
| | - Alex Armento
- MatTek Corporation, 200 Homer Avenue, Ashland, Massachusetts, USA
| | - Patrick Hayden
- MatTek Corporation, 200 Homer Avenue, Ashland, Massachusetts, USA
| | | |
Collapse
|
17
|
Delaney B. In vitro studies with human intestinal epithelial cell line monolayers for protein hazard characterization. Food Chem Toxicol 2017; 110:425-433. [DOI: 10.1016/j.fct.2017.09.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/12/2017] [Accepted: 09/14/2017] [Indexed: 10/18/2022]
|
18
|
Incorporation of in vitro digestive enzymes in an intestinal epithelial cell line model for protein hazard identification. Toxicol In Vitro 2017; 44:85-93. [DOI: 10.1016/j.tiv.2017.06.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/04/2017] [Accepted: 06/17/2017] [Indexed: 11/18/2022]
|