Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
PLoS Pathog ; 16(2): e1008304, 2020 02.
Article in English | MEDLINE | ID: mdl-32069333

ABSTRACT

The Gram-negative Epsilonproteobacterium Campylobacter jejuni is currently the most prevalent bacterial foodborne pathogen. Like for many other human pathogens, infection studies with C. jejuni mainly employ artificial animal or cell culture models that can be limited in their ability to reflect the in-vivo environment within the human host. Here, we report the development and application of a human three-dimensional (3D) infection model based on tissue engineering to study host-pathogen interactions. Our intestinal 3D tissue model is built on a decellularized extracellular matrix scaffold, which is reseeded with human Caco-2 cells. Dynamic culture conditions enable the formation of a polarized mucosal epithelial barrier reminiscent of the 3D microarchitecture of the human small intestine. Infection with C. jejuni demonstrates that the 3D tissue model can reveal isolate-dependent colonization and barrier disruption phenotypes accompanied by perturbed localization of cell-cell junctions. Pathogenesis-related phenotypes of C. jejuni mutant strains in the 3D model deviated from those obtained with 2D-monolayers, but recapitulated phenotypes previously observed in animal models. Moreover, we demonstrate the involvement of a small regulatory RNA pair, CJnc180/190, during infections and observe different phenotypes of CJnc180/190 mutant strains in 2D vs. 3D infection models. Hereby, the CJnc190 sRNA exerts its pathogenic influence, at least in part, via repression of PtmG, which is involved in flagellin modification. Our results suggest that the Caco-2 cell-based 3D tissue model is a valuable and biologically relevant tool between in-vitro and in-vivo infection models to study virulence of C. jejuni and other gastrointestinal pathogens.


Subject(s)
Campylobacter jejuni/genetics , Host-Pathogen Interactions/physiology , Models, Biological , Caco-2 Cells , Campylobacter Infections/microbiology , Campylobacter jejuni/pathogenicity , Epithelial Cells/microbiology , Extracellular Matrix/physiology , Humans , Intestinal Mucosa/microbiology , Intestine, Small/pathology , Intestines/microbiology , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , Tissue Scaffolds , Virulence
2.
Gastroenterology ; 152(4): 867-879, 2017 03.
Article in English | MEDLINE | ID: mdl-27889570

ABSTRACT

BACKGROUND AND AIMS: Tumor necrosis factor (TNF) is a cytokine that promotes inflammation and contributes to pathogenesis of inflammatory bowel diseases. Unlike other cells and tissues, intestinal epithelial cells undergo rapid cell death upon exposure to TNF, by unclear mechanisms. We investigated the roles of inhibitor of apoptosis proteins (IAPs) in the regulation of TNF-induced cell death in the intestinal epithelium of mice and intestinal organoids. METHODS: RNA from cell lines and tissues was analyzed by quantitative polymerase chain reaction, protein levels were analyzed by immunoblot assays. BIRC2 (also called cIAP1) was expressed upon induction from lentiviral vectors in young adult mouse colon (YAMC) cells. YAMC cells, the mouse colon carcinoma cell line MC38, the mouse macrophage cell line RAW 264.7, or mouse and human organoids were incubated with second mitochondrial activator of caspases (Smac)-mimetic compound LCL161 or recombinant TNF-like weak inducer of apoptosis (TNFSF12) along with TNF, and cell death was quantified. C57BL/6 mice with disruption of Xiap, Birc2 (encodes cIAP1), Birc3 (encodes cIAP2), Tnfrsf1a, or Tnfrsf1b (Tnfrsf1a and b encode TNF receptors) were injected with TNF or saline (control); liver and intestinal tissues were collected and analyzed for apoptosis induction by cleaved caspase 3 immunohistochemistry. We also measured levels of TNF and alanine aminotransferase in serum from mice. RESULTS: YAMC cells, and mouse and human intestinal organoids, died rapidly in response to TNF. YAMC and intestinal crypts expressed lower levels of XIAP, cIAP1, cIAP2, and cFLIP than liver tissue. Smac-mimetics reduced levels of cIAP1 and XIAP in MC38 and YAMC cells, and Smac-mimetics and TNF-related weak inducer of apoptosis increased TNF-induced cell death in YAMC cells and organoids-most likely by sequestering and degrading cIAP1. Injection of TNF greatly increased levels of cell death in intestinal tissue of cIAP1-null mice, compared with wild-type C57BL/6 mice, cIAP2-null mice, or XIAP-null mice. Excessive TNF-induced cell death in the intestinal epithelium was mediated TNF receptor 1. CONCLUSIONS: In a study of mouse and human cell lines, organoids, and tissues, we found cIAP1 to be required for regulation of TNF-induced intestinal epithelial cell death and survival. These findings have important implications for the pathogenesis of TNF-mediated enteropathies and chronic inflammatory diseases of the intestine.


Subject(s)
Apoptosis , Epithelial Cells , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Baculoviral IAP Repeat-Containing 3 Protein , Cell Death/drug effects , Cell Line, Tumor , Cytokine TWEAK , Epithelial Cells/drug effects , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Liver/drug effects , Macrophages , Mice , Mice, Inbred C57BL , Mice, Knockout , Organoids , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Thiazoles/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factors/pharmacology
3.
Angew Chem Int Ed Engl ; 54(9): 2816-20, 2015 Feb 23.
Article in English | MEDLINE | ID: mdl-25640578

ABSTRACT

Biofabrication is an emerging and rapidly expanding field of research in which additive manufacturing techniques in combination with cell printing are exploited to generate hierarchical tissue-like structures. Materials that combine printability with cytocompatibility, so called bioinks, are currently the biggest bottleneck. Since recombinant spider silk proteins are non-immunogenic, cytocompatible, and exhibit physical crosslinking, their potential as a new bioink system was evaluated. Cell-loaded spider silk constructs can be printed by robotic dispensing without the need for crosslinking additives or thickeners for mechanical stabilization. Cells are able to adhere and proliferate with good viability over at least one week in such spider silk scaffolds. Introduction of a cell-binding motif to the spider silk protein further enables fine-tuned control over cell-material interactions. Spider silk hydrogels are thus a highly attractive novel bioink for biofabrication.


Subject(s)
Fibroblasts/chemistry , Fibroblasts/cytology , Silk/chemistry , Spiders/chemistry , Tissue Scaffolds/chemistry , Animals , BALB 3T3 Cells , Cell Adhesion , Cell Proliferation , Hydrogels/chemistry , Mice , Mice, Inbred BALB C
4.
J Tissue Eng ; 12: 2041731420988802, 2021.
Article in English | MEDLINE | ID: mdl-33796248

ABSTRACT

Gonorrhea, a sexually transmitted disease caused by the bacteria Neisseria gonorrhoeae, is characterized by a large number of neutrophils recruited to the site of infection. Therefore, proper modeling of the N. gonorrhoeae interaction with neutrophils is very important for investigating and understanding the mechanisms that gonococci use to evade the immune response. We have used a combination of a unique human 3D tissue model together with a dynamic culture system to study neutrophil transmigration to the site of N. gonorrhoeae infection. The triple co-culture model consisted of epithelial cells (T84 human colorectal carcinoma cells), human primary dermal fibroblasts, and human umbilical vein endothelial cells on a biological scaffold (SIS). After the infection of the tissue model with N. gonorrhoeae, we introduced primary human neutrophils to the endothelial side of the model using a perfusion-based bioreactor system. By this approach, we were able to demonstrate the activation and transmigration of neutrophils across the 3D tissue model and their recruitment to the site of infection. In summary, the triple co-culture model supplemented by neutrophils represents a promising tool for investigating N. gonorrhoeae and other bacterial infections and interactions with the innate immunity cells under conditions closely resembling the native tissue environment.

5.
mBio ; 11(1)2020 02 18.
Article in English | MEDLINE | ID: mdl-32071273

ABSTRACT

A major obstacle in infection biology is the limited ability to recapitulate human disease trajectories in traditional cell culture and animal models, which impedes the translation of basic research into clinics. Here, we introduce a three-dimensional (3D) intestinal tissue model to study human enteric infections at a level of detail that is not achieved by conventional two-dimensional monocultures. Our model comprises epithelial and endothelial layers, a primary intestinal collagen scaffold, and immune cells. Upon Salmonella infection, the model mimics human gastroenteritis, in that it restricts the pathogen to the epithelial compartment, an advantage over existing mouse models. Application of dual transcriptome sequencing to the Salmonella-infected model revealed the communication of epithelial, endothelial, monocytic, and natural killer cells among each other and with the pathogen. Our results suggest that Salmonella uses its type III secretion systems to manipulate STAT3-dependent inflammatory responses locally in the epithelium without accompanying alterations in the endothelial compartment. Our approach promises to reveal further human-specific infection strategies employed by Salmonella and other pathogens.IMPORTANCE Infection research routinely employs in vitro cell cultures or in vivo mouse models as surrogates of human hosts. Differences between murine and human immunity and the low level of complexity of traditional cell cultures, however, highlight the demand for alternative models that combine the in vivo-like properties of the human system with straightforward experimental perturbation. Here, we introduce a 3D tissue model comprising multiple cell types of the human intestinal barrier, a primary site of pathogen attack. During infection with the foodborne pathogen Salmonella enterica serovar Typhimurium, our model recapitulates human disease aspects, including pathogen restriction to the epithelial compartment, thereby deviating from the systemic infection in mice. Combination of our model with state-of-the-art genetics revealed Salmonella-mediated local manipulations of human immune responses, likely contributing to the establishment of the pathogen's infection niche. We propose the adoption of similar 3D tissue models to infection biology, to advance our understanding of molecular infection strategies employed by bacterial pathogens in their human host.


Subject(s)
Coculture Techniques/methods , Host-Pathogen Interactions/physiology , Intestines/microbiology , Salmonella Infections/microbiology , Animals , CRISPR-Cas Systems , Caco-2 Cells , Disease Models, Animal , Epithelial Cells/microbiology , Epithelium/microbiology , Gastroenteritis/microbiology , Gene Expression Regulation, Bacterial , Humans , Killer Cells, Natural , Mice , STAT3 Transcription Factor/metabolism , Salmonella Infections/immunology , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Transcriptome , Type III Secretion Systems
6.
J Crohns Colitis ; 14(10): 1473-1487, 2020 Oct 05.
Article in English | MEDLINE | ID: mdl-32342109

ABSTRACT

BACKGROUND: The mechanisms underlying loss of intestinal epithelial barrier [IEB] function in Crohn's disease [CD] are poorly understood. We tested whether human enteroids generated from isolated intestinal crypts of CD patients serve as an appropriate in vitro model to analyse changes of IEB proteins observed in patients' specimens. METHODS: Gut samples from CD patients and healthy individuals who underwent surgery were collected. Enteroids were generated from intestinal crypts and analyses of junctional proteins in comparison to full wall samples were performed. RESULTS: Histopathology confirmed the presence of CD and the extent of inflammation in intestinal full wall sections. As revealed by immunostaining and Western blot analysis, profound changes in expression patterns of tight junction, adherens junction and desmosomal proteins were observed in full wall specimens when CD was present. Unexpectedly, when enteroids were generated from specimens of CD patients with severe inflammation, alterations of most tight junction proteins and the majority of changes in desmosomal proteins but not E-cadherin were maintained under culture conditions. Importantly, these changes were maintained without any additional stimulation of cytokines. Interestingly, qRT-PCR demonstrated that mRNA levels of junctional proteins were not different when enteroids from CD patients were compared to enteroids from healthy controls. CONCLUSIONS: These data indicate that enteroids generated from patients with severe inflammation in CD maintain some characteristics of intestinal barrier protein changes on a post-transcriptional level. The enteroid in vitro model represents an appropriate tool to gain further cellular and molecular insights into the pathogenesis of barrier dysfunction in CD.


Subject(s)
Crohn Disease , Desmosomal Cadherins/metabolism , Epithelial Attachment/metabolism , Inflammation , Intestinal Mucosa , Cells, Cultured , Crohn Disease/immunology , Crohn Disease/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Models, Biological , RNA, Messenger/analysis , Tight Junctions/metabolism
7.
Biotechniques ; 67(1): 23-28, 2019 07.
Article in English | MEDLINE | ID: mdl-31218886

ABSTRACT

Organoids recapitulate the (patho)physiological processes in certain tissues and organs closer than classical cell lines. Therefore, organoid technology offers great potentials in drug development and testing, and personalized medicine. In particular, organoids can be used to study and predict drug-induced toxicity in certain tissues. However, until today few methods had been reported to analyze cell death in 3D-microtissues in a quantitative manner. Here, we describe a novel fluorometric method for the quantitative measurement of specific organoid cell death. Organoids are stained simultaneously with the cell impermeable nuclear dye propidium iodide and cell permeable Hoechst33342. While Hoechst allows in-well normalization to cell numbers, propidium iodide detects relative proportion of dead cells independent of hydrogel. Measurement and analysis time, as well as usability are drastically improved in comparison to other established methods. Parallel multiplexing of our method with established assays measuring mitochondrial activity further enhances its applicability in personalized medicine and drug discovery.


Subject(s)
Cell Death , Fluorometry/methods , Intestines/cytology , Organoids/cytology , Animals , Cell Death/drug effects , Cells, Cultured , Drug Evaluation, Preclinical/methods , Humans , Intestines/drug effects , Mice, Inbred C57BL , Microscopy, Fluorescence/methods , Organoids/drug effects
8.
Front Microbiol ; 10: 1740, 2019.
Article in English | MEDLINE | ID: mdl-31417529

ABSTRACT

Gonorrhea is the second most common sexually transmitted infection in the world and is caused by Gram-negative diplococcus Neisseria gonorrhoeae. Since N. gonorrhoeae is a human-specific pathogen, animal infection models are only of limited use. Therefore, a suitable in vitro cell culture model for studying the complete infection including adhesion, transmigration and transport to deeper tissue layers is required. In the present study, we generated three independent 3D tissue models based on porcine small intestinal submucosa (SIS) scaffold by co-culturing human dermal fibroblasts with human colorectal carcinoma, endometrial epithelial, and male uroepithelial cells. Functional analyses such as transepithelial electrical resistance (TEER) and FITC-dextran assay indicated the high barrier integrity of the created monolayer. The histological, immunohistochemical, and ultra-structural analyses showed that the 3D SIS scaffold-based models closely mimic the main characteristics of the site of gonococcal infection in human host including the epithelial monolayer, the underlying connective tissue, mucus production, tight junction, and microvilli formation. We infected the established 3D tissue models with different N. gonorrhoeae strains and derivatives presenting various phenotypes regarding adhesion and invasion. The results indicated that the disruption of tight junctions and increase in interleukin production in response to the infection is strain and cell type-dependent. In addition, the models supported bacterial survival and proved to be better suitable for studying infection over the course of several days in comparison to commonly used Transwell® models. This was primarily due to increased resilience of the SIS scaffold models to infection in terms of changes in permeability, cell destruction and bacterial transmigration. In summary, the SIS scaffold-based 3D tissue models of human mucosal tissues represent promising tools for investigating N. gonorrhoeae infections under close-to-natural conditions.

9.
J Clin Invest ; 129(7): 2824-2840, 2019 06 17.
Article in English | MEDLINE | ID: mdl-31205031

ABSTRACT

Impaired intestinal epithelial barrier (IEB) function with loss of desmosomal junctional protein desmoglein 2 (DSG2) is a hallmark in the pathogenesis of inflammatory bowel disease (IBD). While previous studies have reported that glial cell line-derived neurotrophic factor (GDNF) promotes IEB function, the mechanisms are poorly understood. We hypothesized that GDNF is involved in the loss of DSG2, resulting in impaired IEB function as seen in IBD. In the inflamed intestine of patients with IBD, there was a decrease in GDNF concentrations accompanied by a loss of DSG2, changes of the intermediate filament system, and increased phosphorylation of p38 MAPK and cytokeratins. DSG2-deficient and RET-deficient Caco2 cells revealed that GDNF specifically recruits DSG2 to the cell borders, resulting in increased DSG2-mediated intercellular adhesion via the RET receptor. Challenge of Caco2 cells and enteroids with proinflammatory cytokines as well as dextran sulfate sodium-induced (DSS-induced) colitis in C57Bl/6 mice led to impaired IEB function with reduced DSG2 mediated by p38 MAPK-dependent phosphorylation of cytokeratins. GDNF blocked all inflammation-induced changes in the IEB. GDNF attenuates inflammation-induced impairment of IEB function caused by the loss of DSG2 through p38 MAPK-dependent phosphorylation of cytokeratin. The reduced GDNF in patients with IBD indicates a disease-relevant contribution to the development of IEB dysfunction.


Subject(s)
Colon/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Inflammatory Bowel Diseases/metabolism , MAP Kinase Signaling System , Animals , Caco-2 Cells , Colon/pathology , Desmoglein 2/metabolism , Dextran Sulfate/adverse effects , Dextran Sulfate/pharmacology , Disease Models, Animal , Humans , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/pathology , Mice , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Tissue Eng Part A ; 24(3-4): 301-309, 2018 02.
Article in English | MEDLINE | ID: mdl-28783453

ABSTRACT

The loss of salivary gland function caused by radiation therapy of the head and neck is a serious condition and it affects a patient's quality of life. The current lack of effective therapies demands new options to be explored. This study tested whether human salivary gland epithelial cells (SGECs) could be successfully cultured on a decellularized porcine gut matrix (SIS-muc) in both mono- and coculture with microvascular endothelial cells (mvECs). By performing immunofluorescence imaging, transmission as well as scanning electron microscopy (SEM), quantitative polymerase chain reaction (qPCR), and an amylase enzyme assay, it was investigated as to what extent the three-dimensional (3D)-cultured cells could maintain their molecular differentiation and the production of working α-amylase (α-AMY) compared with two-dimensional (2D) culture. In both 3D mono- and coculture, SGECs were successfully cultured and formed acinar-like structures. Those findings were confirmed by SEM imaging. Immunofluorescence imaging revealed that 3D-cultured cells expressed α-AMY, Claudin-1 (CL-1), and water channel protein aquaporin-5 (AQP-5). Two-dimensional-cultured cells only were positive for α-AMY. Real time (RT)-qPCR analysis showed that α-AMY relative gene expression was higher in both 3D mono- and coculture than in 2D culture. In α-AMY enzyme assay, cocultured SGECs showed about 25 times increased enzyme activity compared with 2D-cultured cells. In conclusion, the SIS-muc combined with endothelial coculture seems a suitable culture setting for the tissue engineering of functional human salivary gland tissue.


Subject(s)
Coculture Techniques/methods , Endothelial Cells/cytology , Epithelial Cells/cytology , Salivary Glands/cytology , Amylases/metabolism , Cell Differentiation/physiology , Cells, Cultured , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Humans , Microscopy, Electron, Scanning , Tissue Engineering
11.
Biotechnol J ; 12(2)2017 Feb.
Article in English | MEDLINE | ID: mdl-27492568

ABSTRACT

Tissue Engineering (TE) bears potential to overcome the persistent shortage of donor organs in transplantation medicine. Additionally, TE products are applied as human test systems in pharmaceutical research to close the gap between animal testing and the administration of drugs to human subjects in clinical trials. However, generating a tissue requires complex culture conditions provided by bioreactors. Currently, the translation of TE technologies into clinical and industrial applications is limited due to a wide range of different tissue-specific, non-disposable bioreactor systems. To ensure a high level of standardization, a suitable cost-effectiveness, and a safe graft production, a generic modular bioreactor platform was developed. Functional modules provide robust control of culture processes, e.g. medium transport, gas exchange, heating, or trapping of floating air bubbles. Characterization revealed improved performance of the modules in comparison to traditional cell culture equipment such as incubators, or peristaltic pumps. By combining the modules, a broad range of culture conditions can be achieved. The novel bioreactor platform allows using disposable components and facilitates tissue culture in closed fluidic systems. By sustaining native carotid arteries, engineering a blood vessel, and generating intestinal tissue models according to a previously published protocol the feasibility and performance of the bioreactor platform was demonstrated.


Subject(s)
Bioreactors , Tissue Engineering/methods , Regenerative Medicine/methods
12.
Drug Deliv Transl Res ; 7(2): 217-227, 2017 04.
Article in English | MEDLINE | ID: mdl-27549106

ABSTRACT

In vitro test systems gain increasing importance in preclinical studies to increase the predictivity and reduce animal testing. Of special interest herein are barrier tissues that guard into the human body. These barriers are formed by highly specialized tissues such as the skin, the airways, and the intestine. However, to recapitulate these tissues, researchers are currently restricted by a lack of suitable supporting scaffolds. In this study, we present biological scaffolds based on decellularized porcine gut segments that offer a natural environment for cell growth and differentiation. Employing these scaffolds, human barrier models of the skin, the airways, and the intestine that mimic the natural histological architecture of the respective tissue are generated. These models show tissue specific barrier properties, such as the stratification of the skin, the mucociliary phenotype of the airways, and polarization of the intestinal epithelium. To investigate the transport characteristics of the intestinal test system, we incubated the tissue models with fluorescein (P app <1 × 106 cm/s), propranolol (P app >7 × 106 cm/s), and rhodamin123 (ratio 2.45). The here presented biological scaffolds facilitate the in vitro generation of human barrier models that might represent useful tools for drug delivery studies.


Subject(s)
Animal Testing Alternatives , Intestinal Mucosa , Intestines , Nasal Mucosa , Skin , Tissue Scaffolds , Animals , Biological Transport , Cells, Cultured , Epithelial Cells , Fibroblasts , Humans , Intestinal Mucosa/metabolism , Intestines/ultrastructure , Keratinocytes , Microscopy, Electron, Scanning , Nasal Mucosa/metabolism , Pharmaceutical Preparations/metabolism , Skin/metabolism , Swine
13.
Cell Rep ; 21(8): 2090-2103, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29166602

ABSTRACT

The host metalloprotease meprin ß is required for mucin 2 (MUC2) cleavage, which drives intestinal mucus detachment and prevents bacterial overgrowth. To gain access to the cleavage site in MUC2, meprin ß must be proteolytically shed from epithelial cells. Hence, regulation of meprin ß shedding and activation is important for physiological and pathophysiological conditions. Here, we demonstrate that meprin ß activation and shedding are mutually exclusive events. Employing ex vivo small intestinal organoid and cell culture experiments, we found that ADAM-mediated shedding is restricted to the inactive pro-form of meprin ß and is completely inhibited upon its conversion to the active form at the cell surface. This strict regulation of meprin ß activity can be overridden by pathogens, as demonstrated for the bacterial protease Arg-gingipain (RgpB). This secreted cysteine protease potently converts membrane-bound meprin ß into its active form, impairing meprin ß shedding and its function as a mucus-detaching protease.


Subject(s)
Adhesins, Bacterial/metabolism , Cysteine Endopeptidases/metabolism , Metalloendopeptidases/metabolism , Metalloproteases/metabolism , Amino Acid Sequence/genetics , Animals , Cell Membrane/metabolism , Epithelial Cells/metabolism , Female , Gingipain Cysteine Endopeptidases , HEK293 Cells , Humans , Male , Metalloendopeptidases/genetics , Mice, Transgenic , Mucin-2/genetics , Mucin-2/metabolism
14.
Tissue Eng Part C Methods ; 22(9): 873-83, 2016 09.
Article in English | MEDLINE | ID: mdl-27481569

ABSTRACT

Intestinal in vitro models are valuable tools in drug discovery and infection research. Despite several advantages, the standard cell line-based Transwell(®) models based for example on colonic epithelial Caco-2 cells, lack the cellular complexity and transport activity associated with native small intestinal tissue. An additional experimental set-back arises from the most commonly used synthetic membranes, on which the cells are routinely cultured. These can lead to an additional barrier activity during in vitro testing. To overcome these limitations, we developed an alternative primary human small intestinal tissue model. This novel approach combines previously established gut organoid technology with a natural extracellular matrix (ECM) based on porcine small intestinal scaffold (SIS). Intestinal crypts from healthy human small intestine were expanded as gut organoids and seeded as single cells on SIS in a standardized Transwell-like setting. After only 7 days on the ECM scaffold, the primary cells formed an epithelial barrier while a subpopulation differentiated into intestinal specific cell types such as mucus-producing goblet cells or hormone-secreting enteroendocrine cells. Furthermore, we tested the influence of subepithelial fibroblasts and dynamic culture conditions on epithelial barrier function. The barrier integrity was stabilized by coculture in the presence of gut-derived fibroblasts. Compared to static or dynamic culture on an orbital shaker, dynamic culture in a defined perfusion bioreactor had an additional significant impact on epithelial cell differentiation, indicated by high prismatic cell morphology and upregulation of CYP3A4 enzyme and Mdr1 transporter activity. In summary, more physiological tissue models as presented in our study might be useful tools in preclinical research and development.


Subject(s)
Cell Differentiation , Fibroblasts/cytology , Intestinal Mucosa/cytology , Intestine, Small/cytology , Models, Biological , Biological Transport , Coculture Techniques , Humans , In Vitro Techniques
SELECTION OF CITATIONS
SEARCH DETAIL