bims-orenst Biomed News
on Organs-on-chips and engineered stem cell models
Issue of 2021‒12‒12
ten papers selected by
Joram Mooiweer
University of Groningen


  1. Front Bioeng Biotechnol. 2021 ;9 781566
      Microfluidic tumour spheroid-on-a-chip platforms enable control of spheroid size and their microenvironment and offer the capability of high-throughput drug screening, but drug supply to spheroids is a complex process that depends on a combination of mechanical, biochemical, and biophysical factors. To account for these coupled effects, many microfluidic device designs and operating conditions must be considered and optimized in a time- and labour-intensive trial-and-error process. Computational modelling facilitates a systematic exploration of a large design parameter space via in silico simulations, but the majority of in silico models apply only a small set of conditions or parametric levels. Novel approaches to computational modelling are needed to explore large parameter spaces and accelerate the optimization of spheroid-on-a-chip and other organ-on-a-chip designs. Here, we report an efficient computational approach for simulating fluid flow and transport of drugs in a high-throughput arrayed cancer spheroid-on-a-chip platform. Our strategy combines four key factors: i) governing physical equations; ii) parametric sweeping; iii) parallel computing; and iv) extensive dataset analysis, thereby enabling a complete "full-factorial" exploration of the design parameter space in combinatorial fashion. The simulations were conducted in a time-efficient manner without requiring massive computational time. As a case study, we simulated >15,000 microfluidic device designs and flow conditions for a representative multicellular spheroids-on-a-chip arrayed device, thus acquiring a single dataset consisting of ∼10 billion datapoints in ∼95 GBs. To validate our computational model, we performed physical experiments in a representative spheroid-on-a-chip device that showed excellent agreement between experimental and simulated data. This study offers a computational strategy to accelerate the optimization of microfluidic device designs and provide insight on the flow and drug transport in spheroid-on-a-chip and other biomicrofluidic platforms.
    Keywords:  cancer spheroids; drug delivery; full-factorial experiments; hierarchical clustering; microfluidic design space exploration; organ-on-a-chip
    DOI:  https://doi.org/10.3389/fbioe.2021.781566
  2. Lab Chip. 2021 Dec 08.
      Transepithelial/transendothelial electrical resistance (TEER) is a label-free assay that is commonly used to assess tissue barrier integrity. TEER measurement systems have been embedded in organ-on-a-chip devices to provide live readouts of barrier functionality. Yet, these systems commonly provide the impedance values which correspond to the highest level of permeability throughout the chip and cannot provide localized information on specific regions of interest. This work introduces a system that provides this essential information: a spatial-TEER (S-TEER) organ-on-a-chip platform, which incorporates moving (scanning) electrodes that can measure electrical resistance at any desired location along the chip. We demonstrate the system's capacity to obtain localized measurements of permeability in selected regions of a cell sample. We show how, in a layer with non-uniform levels of cell coverage, permeability is higher in areas with lower cell density-suggesting that the system can be used to monitor local cellular growth in vitro. To demonstrate the applicability of the chip in studies of barrier function, we characterize tissue response to TNF-α and to EGTA, agents known to harm tissue barrier integrity.
    DOI:  https://doi.org/10.1039/d1lc00789k
  3. Lab Chip. 2021 Dec 08.
      The majority of intestinal in vitro screening models use cell lines that do not reflect the complexity of the human intestinal tract and hence often fail to accurately predict intestinal drug absorption. Tissue explants have intact intestinal architecture and cell type diversity, but show short viability in static conditions. Here, we present a medium throughput microphysiological system, Intestinal Explant Barrier Chip (IEBC), that creates a dynamic microfluidic microenvironment and prolongs tissue viability. Using a snap fit mechanism, we successfully incorporated human and porcine colon tissue explants and studied tissue functionality, integrity and viability for 24 hours. With a proper distinction of transcellular over paracellular transport (ratio >2), tissue functionality was good at early and late timepoints. Low leakage of FITC-dextran and preserved intracellular lactate dehydrogenase levels indicate maintained tissue integrity and viability, respectively. From a selection of low to high permeability drugs, 6 out of 7 properly ranked according to their fraction absorbed. In conclusion, the IEBC is a novel screening platform benefitting from the complexity of tissue explants and the flow in microfluidic chips.
    DOI:  https://doi.org/10.1039/d1lc00669j
  4. Cancer Res. 2021 Dec 06. pii: canres.0799.2021. [Epub ahead of print]
      Optimal treatment of cancer requires diagnostic methods to facilitate therapy choice and prevent ineffective treatments. Direct assessment of therapy response in viable tumor specimens could fill this diagnostic gap. Therefore, we designed a microfluidic platform for assessment of patient treatment response using tumor tissue slices under precisely controlled growth conditions. The optimized Cancer-on-Chip (CoC) platform maintained viability and sustained proliferation of breast and prostate tumor slices for 7 days. No major changes in tissue morphology or gene expression patterns were observed within this time frame, suggesting that the CoC system provides a reliable and effective way to probe intrinsic chemotherapeutic sensitivity of tumors. The customized CoC platform accurately predicted cisplatin and apalutamide treatment response in breast and prostate tumor xenograft models, respectively. The culture period for breast cancer could be extended up to 14 days without major changes in tissue morphology and viability. These culture characteristics enable assessment of treatment outcomes and open possibilities for detailed mechanistic studies.
    DOI:  https://doi.org/10.1158/0008-5472.CAN-21-0799
  5. Front Cell Dev Biol. 2021 ;9 745897
      Myasthenia gravis (MG) is a chronic and progressive neuromuscular disease where autoantibodies target essential proteins such as the nicotinic acetylcholine receptor (nAChR) at the neuromuscular junction (NMJ) causing muscle fatigue and weakness. Autoantibodies directed against nAChRs are proposed to work by three main pathological mechanisms of receptor disruption: blocking, receptor internalization, and downregulation. Current in vivo models using experimental autoimmune animal models fail to recapitulate the disease pathology and are limited in clinical translatability due to disproportionate disease severity and high animal death rates. The development of a highly sensitive antibody assay that mimics human disease pathology is desirable for clinical advancement and therapeutic development. To address this lack of relevant models, an NMJ platform derived from human iPSC differentiated motoneurons and primary skeletal muscle was used to investigate the ability of an anti-nAChR antibody to induce clinically relevant MG pathology in the serum-free, spatially organized, functionally mature NMJ platform. Treatment of the NMJ model with the anti-nAChR antibody revealed decreasing NMJ stability as measured by the number of NMJs before and after the synchrony stimulation protocol. This decrease in NMJ stability was dose-dependent over a concentration range of 0.01-20 μg/mL. Immunocytochemical (ICC) analysis was used to distinguish between pathological mechanisms of antibody-mediated receptor disruption including blocking, receptor internalization and downregulation. Antibody treatment also activated the complement cascade as indicated by complement protein 3 deposition near the nAChRs. Additionally, complement cascade activation significantly altered other readouts of NMJ function including the NMJ fidelity parameter as measured by the number of muscle contractions missed in response to increasing motoneuron stimulation frequencies. This synchrony readout mimics the clinical phenotype of neurological blocking that results in failure of muscle contractions despite motoneuron stimulations. Taken together, these data indicate the establishment of a relevant disease model of MG that mimics reduction of functional nAChRs at the NMJ, decreased NMJ stability, complement activation and blocking of neuromuscular transmission. This system is the first functional human in vitro model of MG to be used to simulate three potential disease mechanisms as well as to establish a preclinical platform for evaluation of disease modifying treatments (etiology).
    Keywords:  acetylcholine receptor; autoantibodies; disease pathology; human-on-a-chip; induced pluripotent stem cells (iPSCs); microphysiological systems; myasthenia gravis; neuromuscular junction (NMJ)
    DOI:  https://doi.org/10.3389/fcell.2021.745897
  6. Int J Mol Sci. 2021 Nov 26. pii: 12788. [Epub ahead of print]22(23):
      The skin is subject to both intrinsic aging caused by metabolic processes in the body and extrinsic aging caused by exposure to environmental factors. Intrinsic aging is an important obstacle to in vitro experimentation as its long-term progression is difficult to replicate. Here, we accelerated aging of a full-thickness skin equivalent by applying periodic mechanical stimulation, replicating the circadian rhythm for 28 days. This aging skin model was developed by culturing a full-thickness, three-dimensional skin equivalent with human fibroblasts and keratinocytes to produce flexible skin-on-a-chip. Accelerated aging associated with periodic compressive stress was evidenced by reductions in the epidermal layer thickness, contraction rate, and secretion of Myb. Increases in β-galactosidase gene expression and secretion of reactive oxygen species and transforming growth factor-β1 were also observed. This in vitro aging skin model is expected to greatly accelerate drug development for skin diseases and cosmetics that cannot be tested on animals.
    Keywords:  circadian rhythm; flexible skin-on-a-chip; periodic compressive stress
    DOI:  https://doi.org/10.3390/ijms222312788
  7. Cancers (Basel). 2021 Nov 26. pii: 5955. [Epub ahead of print]13(23):
      Activated pancreatic stellate cells (aPSCs) and M2 macrophages modulate tumor progression and therapeutic efficacy in pancreatic ductal adenocarcinoma (PDAC) via epithelial-mesenchymal transition (EMT). Here, our aim was to analyze the anti-invasion effects of anti-cancer agents where EMT-inducing cancer-stroma interaction occurs under three-dimensional (3D) culture conditions. We used microfluidic channel chips to co-culture pancreatic tumor spheroids (TSs) with aPSCs and THP-1-derived M2 macrophages (M2 THP-1 cells) embedded in type I collagen. Under stromal cell co-culture conditions, PANC-1 TSs displayed elevated expression of EMT-related proteins and increased invasion and migration. When PANC-1 TSs were exposed to gemcitabine, 5-fluorouracil, oxaliplatin, or paclitaxel, 30-50% cells were found unaffected, with no significant changes in the dose-response profiles under stromal cell co-culture conditions. This indicated intrinsic resistance to these drugs and no further induction of drug resistance by stromal cells. Paclitaxel had a significant anti-invasion effect; in contrast, oxaliplatin did not show such effect despite its specific cytotoxicity in M2 THP-1 cells. Overall, our findings demonstrate that the TS-stroma co-culture model of PDAC is useful for activity profiling of anti-cancer agents against cancer and stromal cells, and analyzing the relationship between anti-stromal activity and anti-invasion effects.
    Keywords:  3D culture; M2 macrophage; anti-cancer drug; pancreatic cancer; pancreatic stellate cell; tumor microenvironment; tumor spheroid
    DOI:  https://doi.org/10.3390/cancers13235955
  8. Cancers (Basel). 2021 Nov 30. pii: 6044. [Epub ahead of print]13(23):
      Breast cancer frequently metastasizes to lymphatics and the presence of breast cancer cells in regional lymph nodes is an important prognostic factor. Delineating the mechanisms by which breast cancer cells disseminate and spatiotemporal aspects of interactions between breast cancer cells and lymphatics is needed to design new therapies to prevent lymphatic metastases. As triple-negative breast cancer (TNBC) has a high incidence of lymphatic metastasis, we used a three-dimensional (3D) coculture model of human TNBC cells and human microvascular lymphatic endothelial cells (LECs) to analyze TNBC:LEC interactions. Non-invasive analyses such as live-cell imaging in real-time and collection of conditioned media for secretomic analysis were facilitated by our novel microfluidic chambers. The volumes of 3D structures formed in TNBC:LEC cocultures are greater than that of 3D structures formed by either LEC or TNBC monocultures. Over 4 days of culture there is an increase in multicellular invasive outgrowths from TNBC spheroids and an association of TNBC spheroids with LEC networks. The increase in invasive phenotype also occurred when TNBC spheroids were cultured in LEC-conditioned media and in wells linked to ones containing LEC networks. Our results suggest that modeling spatiotemporal interactions between TNBC and LECs may reveal paracrine signaling that could be targeted to reduce lymphatic metastasis.
    Keywords:  3D cocultures; lymphatic endothelial cells (LECs); lymphatic metastasis; microfluidic chambers; triple-negative breast cancer (TNBC)
    DOI:  https://doi.org/10.3390/cancers13236044
  9. Front Bioeng Biotechnol. 2021 ;9 764212
      Three-dimensional (3D) co-culture models have closer physiological cell composition and behavior than traditional 2D culture. They exhibit pharmacological effects like in vivo responses, and therefore serve as a high-throughput drug screening model to evaluate drug efficacy and safety in vitro. In this study, we created a 3D co-culture environment to mimic pathological characteristics of rheumatoid arthritis (RA) pannus tissue. 3D scaffold was constructed by bioprinting technology with synovial fibroblasts (MH7A), vascular endothelial cells (EA.hy 926) and gelatin/alginate hydrogels. Cell viability was observed during 7-day culture and the proliferation rate of co-culture cells showed a stable increase stage. Cell-cell interactions were evaluated in the 3D printed scaffold and we found that spheroid size increased with time. TNF-α stimulated MH7A and EA.hy 926 in 3D pannus model showed higher vascular endothelial growth factor (VEGF) and angiopoietin (ANG) protein expression over time. For drug validation, methotrexate (MTX) was used to examine inhibition effects of angiogenesis in 3D pannus co-culture model. In conclusion, this 3D co-culture pannus model with biological characteristics may help the development of anti-RA drug research.
    Keywords:  3D bioprinting; drug screening; pannus tissue model; rheumatoid arthritis; tissue engineering
    DOI:  https://doi.org/10.3389/fbioe.2021.764212
  10. ACS Biomater Sci Eng. 2021 Dec 07.
      Injured or diseased airway epithelium due to repeated environmental insults or genetic mutations can lead to a functional decline of the lung and incurable lung diseases. Bioengineered airway tissue constructs can facilitate in vitro investigation of human lung diseases and accelerate the development of effective therapeutics. Here, we report robust tissue manipulation modalities that allow: (i) selective removal of the endogenous epithelium of in vitro cultured airway tissues and (ii) spatially uniform distribution and prolonged cultivation of exogenous cells that are implanted topically onto the denuded airway lumen. Results obtained highlight that our approach to airway tissue manipulation can facilitate controlled removal of the airway epithelium and subsequent homogeneous distribution of newly implanted cells. This study can contribute to the creation of innovative tissue engineering methodologies that can facilitate the treatment of lung diseases, such as cystic fibrosis, primary ciliary dyskinesia, and chronic obstructive pulmonary disease.
    Keywords:  bioreactor; cell replacement; hydrogel; imaging; lung disease; tissue-on-a-chip
    DOI:  https://doi.org/10.1021/acsbiomaterials.1c01031