Duodenum Intestine-Chip for Toxicity Assessment

Overview

Gastrointestinal toxicity is one of the most common clinical side effects caused by therapeutics and other xenobiotics. Accurately predicting the risk of intestinal toxicity during preclinical drug development could help improve quality of drug treatments and reduce unwanted side effects in the clinic.

In this application note, we review how our Duodenum Intestine-Chip was applied to assess the toxicity of indomethacin—a known GI toxicant.

Key Highlights:

  • Co-culture model incorporates organoid-derived primary epithelial cells and primary human small intestinal microvascular endothelial cells (siHIMECs).
  • The Duodenum Intestine-Chip emulates intestinal tissue and recreates in vivo-like physiology for use in assessment of drug safety.
  • Indomethacin safety assessed across multiple endpoints, including I-FABP accumulation, LDH release, morphology, and barrier function.
  • Future potential applications include assessment of drug absorption and drug-drug interactions, as well as donor-to-donor variability patient-derived cells.

Utilizing the Colon Intestine-Chip to Investigate Mechanisms of Gastrointestinal Disease

Organoids provide long-term viability, spatial organization, and cellular diversity representative of human intestinal epithelium. However, they don’t accurately mimic the architecture and the molecular signature of the intestinal epithelium. This limitation has been an impediment to allowing them to be harnessed for investigating mechanisms that drive leaky-gut syndrome in humans.

Emulate combines advancements in organoid and Organs-on-a-Chip technologies to develop a microphysiological Organ-on-Chip model designed to mimic properties of human intestinal epithelium to enable insights into barrier integrity. With the Colon Intestine-Chip, researchers can investigate novel mechanisms driving leaky gut syndrome and enable their translation from bench to patient bedside.

In this webinar, experts from Emulate and The Johns Hopkins University School of Medicine will share data from studies highlighting how the Colon Intestine-Chip can be used to investigate GI-related diseases.

Key points you will learn: 

  • How the enhanced microenvironment of Organ-Chips—including flow, stretch, and endothelial co-culture—promotes cell functionality, in vivo-like gene expression, and increased enterocyte resistance  
  • Novel insights into the effect of interleukin-22 (IL-22) on barrier function 
  • How the inclusion of mechanical forces brought new aspects of host-pathogen effects to light  
  • How Emulate created a reproducible cytokine-mediated barrier disruption model using interferon gamma (IFNγ)

Human colorectal cancer-on-chip model to study the microenvironmental influence on early metastatic spread

Organ Model: Intestine (Caco2 & organoids)

Application: Cancer

Abstract: Colorectal cancer (CRC) progression is a complex process that is not well understood. We describe an in vitro organ-on-chip model that emulates in vivo tissue structure and the tumor microenvironment (TME) to better understand intravasation, an early step in metastasis. The CRC-on-chip incorporates fluid flow and peristalsis-like cyclic stretching and consists of endothelial and epithelial compartments, separated by a porous membrane. On-chip imaging and effluent analyses are used to interrogate CRC progression and the resulting cellular heterogeneity. Mass spectrometry-based metabolite profiles are indicative of a CRC disease state. Tumor cells intravasate from the epithelial channel to the endothelial channel, revealing differences in invasion between aggressive and non-aggressive tumor cells. Tuning the TME by peristalsis-like mechanical forces, the epithelial:endothelial interface, and the addition of fibroblasts influences the invasive capabilities of tumor cells. The CRC-on-chip is a tunable human-relevant model system and a valuable tool to study early invasive events in cancer.

Characterization of an engineered live bacterial therapeutic for the treatment of phenylketonuria in a human gut-on-a-chip

Organ Model: Intestine (Caco2)

Application: Microbiome

Abstract: Engineered bacteria (synthetic biotics) represent a new class of therapeutics that leverage the tools of synthetic biology. Translational testing strategies are required to predict synthetic biotic function in the human body. Gut-on-a-chip microfluidics technology presents an opportunity to characterize strain function within a simulated human gastrointestinal tract. Here, we apply a human gut-chip model and a synthetic biotic designed for the treatment of phenylketonuria to demonstrate dose-dependent production of a strain-specific biomarker, to describe human tissue responses to the engineered strain, and to show reduced blood phenylalanine accumulation after administration of the engineered strain. Lastly, we show how in vitro gut-chip models can be used to construct mechanistic models of strain activity and recapitulate the behavior of the engineered strain in a non-human primate model. These data demonstrate that gut-chip models, together with mechanistic models, provide a framework to predict the function of candidate strains in vivo.

Colon Intestine-Chip Webinar

Webinar Abstract

In this breakout session from our March 2021 virtual event, ‘Human Biology: An Exploration of Organs-on-Chips,’ we give an overview of our Colon Intestine-Chip and how it can be applied to study cytokine-mediated inflammation, anti-inflammatory therapeutic efficacy, and (coming soon) immune cell recruitment.

Watch the Full Virtual Event

Click here to watch the full event, and learn how Organs-on-Chips technology is transforming therapeutic research. The event features informative breakout sessions on neurology, inflammation, infectious diseases, and the future of Organs-on-Chips.


Harnessing Colon Chip Technology to Identify Commensal Bacteria That Promote Host Tolerance to Infection

Organ Model: Intestine (Colon mouse)

Applications: Microbiome, Infectious Disease

Abstract: Commensal bacteria within the gut microbiome contribute to development of host tolerance to infection, however, identifying specific microbes responsible for this response is difficult. Here we describe methods for developing microfluidic organ-on-a-chip models of small and large intestine lined with epithelial cells isolated from duodenal, jejunal, ileal, or colon organoids derived from wild type or transgenic mice. To focus on host-microbiome interactions, we carried out studies with the mouse Colon Chip and demonstrated that it can support co-culture with living gut microbiome and enable assessment of effects on epithelial adhesion, tight junctions, barrier function, mucus production, and cytokine release. Moreover, infection of the Colon Chips with the pathogenic bacterium, Salmonella typhimurium, resulted in epithelial detachment, decreased tight junction staining, and increased release of chemokines (CXCL1, CXCL2, and CCL20) that closely mimicked changes previously seen in mice. Symbiosis between microbiome bacteria and the intestinal epithelium was also recapitulated by populating Colon Chips with complex living mouse or human microbiome. By taking advantage of differences in the composition between complex microbiome samples cultured on each chip using 16s sequencing, we were able to identify Enterococcus faecium as a positive contributor to host tolerance, confirming past findings obtained in mouse experiments. Thus, mouse Intestine Chips may represent new experimental in vitro platforms for identifying particular bacterial strains that modulate host response to pathogens, as well as for investigating the cellular and molecular basis of host-microbe interactions.

Microfluidic device facilitates in vitro modeling of human neonatal necrotizing enterocolitis-on-a-chip

Organ Model: Intestine (Ileum)

Application: Inflammation

Abstract: Necrotizing enterocolitis (NEC) is a deadly gastrointestinal disease of premature infants that is associated with an exaggerated inflammatory response, dysbiosis of the gut microbiome, decreased epithelial cell proliferation, and gut barrier disruption. We describe an in vitro model of the human neonatal small intestinal epithelium (Neonatal-Intestine-on-a-Chip) that mimics key features of intestinal physiology. This model utilizes intestinal enteroids grown from surgically harvested intestinal tissue from premature infants and cocultured with human intestinal microvascular endothelial cells within a microfluidic device. We used our Neonatal-Intestine-on-a-Chip to recapitulate NEC pathophysiology by adding infant-derived microbiota. This model, named NEC-on-a-Chip, simulates the predominant features of NEC, including significant upregulation of proinflammatory cytokines, decreased intestinal epithelial cell markers, reduced epithelial proliferation, and disrupted epithelial barrier integrity. NEC-on-a-Chip provides an improved preclinical model of NEC that facilitates comprehensive analysis of the pathophysiology of NEC using precious clinical samples. This model is an advance toward a personalized medicine approach to test new therapeutics for this devastating disease.

Effects of Human Milk Oligosaccharides on the Adult Gut Microbiota and Barrier Function

Abstract

Human milk oligosaccharides (HMOs) shape the gut microbiota in infants by selectively stimulating the growth of bifidobacteria. Here, we investigated the impact of HMOs on adult gut microbiota and gut barrier function using the Simulator of the Human Intestinal Microbial Ecosystem (SHIME®), Caco2 cell lines, and human intestinal gut organoid-on-chips. We showed that fermentation of 2’-O-fucosyllactose (2’FL), lacto-N-neotetraose (LNnT), and combinations thereof (MIX) led to an increase of bifidobacteria, accompanied by an increase of short chain fatty acid (SCFA), in particular butyrate with 2’FL. A significant reduction in paracellular permeability of FITC-dextran probe was observed using Caco2 cell monolayers with fermented 2’FL and MIX, which was accompanied by an increase in claudin-8 gene expression as shown by qPCR, and a reduction in IL-6 as determined by multiplex ELISA. Using gut-on-chips generated from human organoids derived from proximal, transverse, and distal colon biopsies (Colon Intestine-Chips), we showed that claudin-5 was significantly upregulated across all three gut-on-chips following treatment with fermented 2’FL under microfluidic conditions. Taken together, these data show that, in addition to their bifidogenic activity, HMOs have the capacity to modulate immune function and the gut barrier, supporting the potential of HMOs to provide health benefits in adults.

Mechanical Stimuli Affect Escherichia coli Heat-Stable Enterotoxin-Cyclic GMP Signaling in a Human Enteroid Intestine-Chip Model

Organ Model: Intestine (Jejunum)

Application: Infectious Disease

Abstract: Modeling host-pathogen interactions with human intestinal epithelia using enteroid monolayers on permeable supports (such as Transwells) represents an alternative to animal studies or use of colon cancer-derived cell lines. However, the static monolayer model does not expose epithelial cells to mechanical forces normally present in the intestine, including luminal flow and serosal blood flow (shear force) or peristaltic forces. To determine the contribution of mechanical forces in the functional response of human small intestine to a virulence factor of a pathogenic intestinal bacterium, human jejunal enteroids were cultured as monolayers in microengineered fluidic-based Organ-Chips (Intestine-Chips) exposed to enterotoxigenic Escherichia coli heat-stable enterotoxin A (ST) and evaluated under conditions of static fluid, apical and basolateral flow, and flow plus repetitive stretch. Application of flow increased epithelial cell height and apical and basolateral secretion of cyclic GMP (cGMP) under baseline, unstimulated conditions. Addition of ST under flow conditions increased apical and basolateral secretion of cGMP relative to the level under static conditions but did not enhance intracellular cGMP accumulation. Cyclic stretch did not have any significant effect beyond that contributed by flow. This study demonstrates that fluid flow application initiates changes in intestinal epithelial cell characteristics relative to those of static culture conditions under both baseline conditions and with exposure to ST enterotoxin and suggests that further investigations of the application of these mechanical forces will provide insights into physiology and pathophysiology that more closely resemble intact intestine than study under static conditions.

Duodenum Intestine-Chip for Preclinical Drug Assessment in a Human Relevant Model

Abstract

Induction of intestinal drug metabolizing enzymes can complicate the development of new drugs, owing to the potential to cause drug-drug interactions (DDIs) leading to changes in pharmacokinetics, safety and efficacy. The development of a human-relevant model of the adult intestine that accurately predicts CYP450 induction could help address this challenge as species differences preclude extrapolation from animals. Here, we combined organoids and Organ-Chip technology to create a human Duodenum Intestine-Chip that emulates intestinal tissue architecture and functions, that are relevant for the study of drug transport, metabolism, and DDI. Duodenum Intestine-Chip demonstrates the polarized cell architecture, intestinal barrier function, presence of specialized cell subpopulations, and in vivo relevant expression, localization, and function of major intestinal drug transporters. Notably, in comparison to Caco-2, it displays improved CYP3A4 expression and induction capability. This model could enable improved in vitro to in vivo extrapolation for better predictions of human pharmacokinetics and risk of DDIs.