bims-orenst Biomed News
on Organs-on-chips and engineered stem cell models
Issue of 2021‒06‒20
ten papers selected by
Joram Mooiweer
University of Groningen


  1. Nat Biomed Eng. 2021 Jun 14.
      The neurovascular unit, which consists of vascular cells surrounded by astrocytic end-feet and neurons, controls cerebral blood flow and the permeability of the blood-brain barrier (BBB) to maintain homeostasis in the neuronal milieu. Studying how some pathogens and drugs can penetrate the human BBB and disrupt neuronal homeostasis requires in vitro microphysiological models of the neurovascular unit. Here we show that the neurotropism of Cryptococcus neoformans-the most common pathogen causing fungal meningitis-and its ability to penetrate the BBB can be modelled by the co-culture of human neural stem cells, brain microvascular endothelial cells and brain vascular pericytes in a human-neurovascular-unit-on-a-chip maintained by a stepwise gravity-driven unidirectional flow and recapitulating the structural and functional features of the BBB. We found that the pathogen forms clusters of cells that penetrate the BBB without altering tight junctions, suggesting a transcytosis-mediated mechanism. The neurovascular-unit-on-a-chip may facilitate the study of the mechanisms of brain infection by pathogens, and the development of drugs for a range of brain diseases.
    DOI:  https://doi.org/10.1038/s41551-021-00743-8
  2. Biomaterials. 2021 Jun 06. pii: S0142-9612(21)00313-6. [Epub ahead of print]275 120957
      Successful translation of in vivo experimental data to human patients is an unmet need and a bottleneck in the development of effective therapeutics. Organ-on-Chip technology aims to address this need by leveraging recent significant advancements in microfabrication and biomaterials, which enable modeling of organs and their functionality. These microengineered chips offer researchers the possibility to recreate critical elements of native tissue architecture such as in vivo relevant tissue-tissue interface, air-liquid interface, and mechanical forces, including mechanical stretch and fluidic shear stress, which are crucial to recapitulate tissue level functions. Here, we present the development of a new, comprehensive 3D cell-culture system, where we combined our proprietary Organ-Chip technology with the advantages offered by three-dimensional organotypic culture. Leveraging microfabrication techniques, we engineered a flexible chip that consists of a chamber containing an organotypic epithelium, surrounded by two vacuum channels that can be actuated to stretch the hydrogel throughout its thickness. Furthermore, the ceiling of this chamber is a removable lid with a built-in microchannel that can be perfused with liquid or air and removed as needed for direct access to the tissue. The bottom part of this chamber is made from a porous flexible membrane which allows diffusive mass transport to and from the microfluidic channel positioned below the membrane. This additional microfluidic channel can be coated with endothelial cells to emulate a blood vessel and recapitulate endothelial interactions. Our results show that the Open-Top Chip design successfully addresses common challenges associated with the Organs-on-Chip technology, including the capability to incorporate a tissue-specific extracellular matrix gel seeded with primary stromal cells, to reproduce the architectural complexity of tissues by micropatterning the gel, and to extract the gel for H&E staining. We also provide proof-of-concept data on the feasibility of using the system with primary human skin and alveolar epithelial cells.
    Keywords:  3D-organotypic cell based models; Hydrogel micropatterning; Microfluidic; Organs-on-Chips; Stretching; Stroma
    DOI:  https://doi.org/10.1016/j.biomaterials.2021.120957
  3. Microphysiol Syst. 2020 Jun;4
      Background: Decreasing the amount of liquid inside microphysiological systems (MPS) can help uncover the presence of toxic drug metabolites. However, maintaining near-physiological volume ratios among blood surrogate and multiple organ mimics is technically challenging. Here, we developed a body cube and tested its ability to support four human tissues (kidney, GI tract, liver, and bone marrow) scaled down from in vivo functional volumes by a factor of 73,000 with 80 μL of cell culture medium (corresponding to ~1/73000th of in vivo blood volume).Methods: GI tract cells (Caco-2), liver cells (HepG2/C3A), bone marrow cells (Meg-01), and kidney cells (HK-2) were co-cultured inside the body cube with 80 μL of common, recirculating cell culture medium for 72 h. The system was challenged with acetaminophen and troglitazone, and concentrations of aspartate aminotransferase (AST), albumin, and urea were monitored over time.
    Results: Cell viability analysis showed that 95.5%±3.2% of liver cells, 89.8%±4.7% of bone marrow cells, 82.8%±8.1% of GI tract cells, and 80.1%±11.5% of kidney cells were viable in co-culture for 72 h. Both acetaminophen and troglitazone significantly lowered cell viability in the liver chamber as indicated by viability analysis and a temporary increase of AST in the cell culture medium. Both drugs also lowered urea production in the liver by up to 45%.
    Conclusions: Cell viability data and the production of urea and albumin indicate that the co-culture of GI tract, liver, bone marrow, and kidney tissues with near-physiological volume ratios of tissues to blood surrogate is possible for up to 72 h. The body-cube was capable of reproducing liver toxicity to HepG2/C3A liver cells via acetaminophen and troglitazone. The developed design provides a viable format for acute toxicity testing with near-physiological blood surrogate to tissue volume ratios.
    Keywords:  MPS; Multi-organ microphysiological system; body cube; body-on-a-chip; microfluidic cell culture
    DOI:  https://doi.org/10.21037/mps-19-8
  4. Biofabrication. 2021 Jun 17.
      The synovium of OA patients can be characterized by an abnormal accumulation of macrophages originating from extravasated monocytes. Since targeting monocyte extravasation may represent a promising therapeutic strategy, our aim was to develop an organotypic microfluidic model recapitulating this process. Synovium and cartilage were modeled by hydrogel-embedded OA synovial fibroblasts and chondrocytes separated by a synovial fluid channel. The synovium compartment included a perfusable endothelialized channel dedicated to monocyte injection. Monocyte extravasation in response to chemokines and OA synovial fluid was quantified. The efficacy of chemokine receptor antagonists, RS-504393 (CCR2 antagonist) and Cenicriviroc (CCR2/CCR5 antagonist) in inhibiting extravasation was tested preincubating monocytes with the antagonists before injection. After designing and fabricating the chip, culture conditions were optimized to achieve an organotypic model including synovial fibroblasts, articular chondrocytes, and a continuous endothelial monolayer expressing ICAM-1 and VCAM-1. A significantly higher number of monocytes extravasated in response to the chemokine mix (p<0.01) and OA synovial fluid (p<0.01), compared to a control condition. In both cases, endothelium pre-activation enhanced monocyte extravasation. The simultaneous blocking of CCR2 and CCR5 proved to be more effective (p<0.001) in inhibiting monocyte extravasation in response to OA synovial fluid than blocking of CCR2 only (p<0.01). The study of extravasation in the model provided direct evidence that OA synovial fluid induces monocytes to cross the endothelium and invade the synovial compartment. The model can be exploited either to test molecules antagonizing this process or to investigate the effect of extravasated monocytes on synovium and cartilage cells.
    Keywords:  Extravasation; Inflammation; Microfluidics; Monocyte; Organ-on-a-chip; Osteoarthritis
    DOI:  https://doi.org/10.1088/1758-5090/ac0c5e
  5. Front Immunol. 2021 ;12 678030
      The actin-related protein (ARP) 2/3 complex, essential for organizing and nucleating branched actin filaments, is required for several cellular immune processes, including cell migration and granule exocytosis. Recently, genetic defects in ARPC1B, a subunit of this complex, were reported. Mutations in ARPC1B result in defective ARP2/3-dependent actin filament branching, leading to a combined immunodeficiency with severe inflammation. In vitro, neutrophils of these patients showed defects in actin polymerization and chemotaxis, whereas adhesion was not altered under static conditions. Here we show that under physiological flow conditions human ARPC1B-deficient neutrophils were able to transmigrate through TNF-α-pre-activated endothelial cells with a decreased efficiency and, once transmigrated, showed definite impairment in subendothelial crawling. Furthermore, severe locomotion and migration defects were observed in a 3D collagen matrix and a perfusable vessel-on-a-chip model. These data illustrate that neutrophils employ ARP2/3-independent steps of adhesion strengthening for transmigration but rely on ARP2/3-dependent modes of migration in a more complex multidimensional environment.
    Keywords:  ARP2/3 complex; ARPC1B deficiency; inborn error of immunity; neutrophil; neutrophil transmigration; primary immunodeficiency; vessel-on-a-chip
    DOI:  https://doi.org/10.3389/fimmu.2021.678030
  6. Front Bioeng Biotechnol. 2021 ;9 624553
      Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a major renal pathology provoked by the deletion of PKD1 or PKD2 genes leading to local renal tubule dilation followed by the formation of numerous cysts, ending up with renal failure in adulthood. In vivo, renal tubules are tightly packed, so that dilating tubules and expanding cysts may have mechanical influence on adjacent tubules. To decipher the role of this coupling between adjacent tubules, we developed a kidney-on-chip reproducing parallel networks of tightly packed tubes. This original microdevice is composed of cylindrical hollow tubes of physiological dimensions, parallel and closely packed with 100-200 μm spacing, embedded in a collagen I matrix. These multitubular systems were properly colonized by different types of renal cells with long-term survival, up to 2 months. While no significant tube dilation over time was observed with Madin-Darby Canine Kidney (MDCK) cells, wild-type mouse proximal tubule (PCT) cells, or with PCT Pkd1 +/- cells (with only one functional Pkd1 allele), we observed a typical 1.5-fold increase in tube diameter with isogenic PCT Pkd1 -/- cells, an ADPKD cellular model. This tube dilation was associated with an increased cell proliferation, as well as a decrease in F-actin stress fibers density along the tube axis. With this kidney-on-chip model, we also observed that for larger tube spacing, PCT Pkd1 -/- tube deformations were not spatially correlated with adjacent tubes whereas for shorter spacing, tube deformations were increased between adjacent tubes. Our device reveals the interplay between tightly packed renal tubes, constituting a pioneering tool well-adapted to further study kidney pathophysiology.
    Keywords:  ADPKD; hydrogel; kidney-on-chip; microfabrication; tube deformation
    DOI:  https://doi.org/10.3389/fbioe.2021.624553
  7. Biomaterials. 2021 Jun 01. pii: S0142-9612(21)00260-X. [Epub ahead of print]275 120904
      Nonalcoholic fatty liver disease (NAFLD) is a significant liver disease without approved therapy, lacking human NAFLD models to aid drug development. Existing models are either under-performing or too complex to allow robust drug screening. Here we have developed a 100-well drug testing platform with improved HepaRG organoids formed with uniform size distribution, and differentiated in situ in a perfusion microfluidic device, SteatoChip, to recapitulate major NAFLD features. Compared with the pre-differentiated spheroids, the in situ differentiated HepaRG organoids with perfusion experience well-controlled chemical and mechanical microenvironment, and 3D cellular niche, to exhibit enhanced hepatic differentiation (albumin+ cells ratio: 66.2% in situ perfusion vs 46.1% pre-differentiation), enriched and uniform hepatocyte distribution in organoids, higher level of hepatocyte functions (5.2 folds in albumin secretion and 7.6 folds in urea synthesis), enhanced cell polarity and bile canaliculi structures. When induced with free fatty acid (FFA), cells exhibit significantly higher level of lipid accumulation (6.6 folds for in situ perfusion vs 4.4 folds for pre-differentiation), altered glucose regulation and reduced Akt phosphorylation in the organoids. SteatoChip detects reduction of steatosis when cells are incubated with three different anti-steatosis compounds, 78.5% by metformin hydrochloride, 71.3% by pioglitazone hydrochloride and 66.6% by obeticholic acid, versus the control FFA-free media (38% reduction). The precision microenvironment control in SteatoChip enables improved formation, differentiation, and function of HepaRG organoids to serve as a scalable and sensitive drug testing platform, to potentially accelerate the NAFLD drug development.
    Keywords:  Differentiation; HepaRG; Microfluidic chip; Microphysiological; NAFLD; Organoid; Steatosis
    DOI:  https://doi.org/10.1016/j.biomaterials.2021.120904
  8. Biotechnol Bioeng. 2021 Jun 18.
      Mucociliary clearance is a crucial mechanism that supports the elimination of inhaled particles, bacteria, pollution and hazardous agents from the human airways, and it also limits the diffusion of aerosolized drugs into the airway epithelium. In spite of its relevance, few in vitro models sufficiently address the cumulative effect of the steric and interactive barrier function of mucus on the one hand, and the dynamic mucus transport imposed by ciliary mucus propulsion on the other hand. Here, ad hoc mucus models of physiological and pathological mucus are combined with magnetic artificial cilia to model mucociliary transport in both physiological and pathological states. The modular concept adopted in this study, enables the development of mucociliary clearance models with high versatility, since these can be easily modified to reproduce phenomena characteristic of healthy and diseased human airways, while allowing to determine the effect of each parameter and/or structure separately on the overall mucociliary transport. These modular airway models can be available off-the-shelf because they are exclusively made of readily available materials, thus ensuring reproducibility across different laboratories. This article is protected by copyright. All rights reserved.
    Keywords:  cilia; dynamic systems; microphysiological models; mucociliary clearance; mucus
    DOI:  https://doi.org/10.1002/bit.27866
  9. Adv Biol (Weinh). 2021 Jun 19. e2100609
      Safety assessment of the effects of developmental toxicants on pregnant women is challenging, and systemic effects in embryo-maternal interactions are largely unknown. However, most developmental toxicity studies rely on animal trials, while in vitro platforms that recapitulate the maternal-placental-embryonic axis are missing. Here, the development of a dedicated microfluidic device for co-cultivation of a placental barrier and 3D embryoid bodies to enable systemic toxicity testing at the embryo-maternal interface is reported. The microfluidic platform features simple handling and recuperation of both tissue models, which facilitates post-hoc in-depth analysis at the tissue and single-cell level. Gravity-driven flow enables inter-tissue communication through the liquid phase as well as simple and robust operation and renders the platform parallelizable. As a proof of concept and to demonstrate platform use for systemic embryotoxicity testing in vitro, maternal exposure to plastic microparticles is emulated, and microparticle effects on the embryo-placental co-culture are investigated.
    Keywords:  3D microtissues; developmental toxicity; embryoid bodies; hanging drops; microfluidics; placenta
    DOI:  https://doi.org/10.1002/adbi.202100609
  10. Front Oncol. 2021 ;11 676135
      Metastatic breast cancer remains a largely incurable and fatal disease with liver involvement bearing the worst prognosis. The danger is compounded by a subset of disseminated tumor cells that may lie dormant for years to decades before re-emerging as clinically detectable metastases. Pathophysiological signals can drive these tumor cells to emerge. Prior studies indicated CXCR3 ligands as being the predominant signals synergistically and significantly unregulated during inflammation in the gut-liver axis. Of the CXCR3 ligands, IP-10 (CXCL10) was the most abundant, correlated significantly with shortened survival of human breast cancer patients with metastatic disease and was highest in those with triple negative (TNBC) disease. Using a complex ex vivo all-human liver microphysiological (MPS) model of dormant-emergent metastatic progression, CXCR3 ligands were found to be elevated in actively growing populations of metastatic TNBC breast cancer cells whereas they remained similar to the tumor-free hepatic niche in those with dormant breast cancer cells. Subsequent stimulation of dormant breast cancer cells in the ex vivo metastatic liver MPS model with IP-10 triggered their emergence in a dose-dependent manner. Emergence was indicated to occur indirectly possibly via activation of the resident liver cells in the surrounding metastatic microenvironment, as stimulation of breast cancer cells with exogenous IP-10 did not significantly change their migratory, invasive or proliferative behavior. The findings reveal that IP-10 is capable of triggering the emergence of dormant breast cancer cells within the liver metastatic niche and identifies the IP-10/CXCR3 as a candidate targetable pathway for rational approaches aimed at maintaining dormancy.
    Keywords:   microphysiological system; CXCL10; IP-10; breast cancer dormancy; metastasis; organ-on-a-chip; tumor dormancy; tumor emergence
    DOI:  https://doi.org/10.3389/fonc.2021.676135