bims-orenst Biomed News
on Organs-on-chips and engineered stem cell models
Issue of 2021‒08‒29
ten papers selected by
Joram Mooiweer
University of Groningen


  1. Micromachines (Basel). 2021 Aug 16. pii: 967. [Epub ahead of print]12(8):
      Progress in understanding kidney disease mechanisms and the development of targeted therapeutics have been limited by the lack of functional in vitro models that can closely recapitulate human physiological responses. Organ Chip (or organ-on-a-chip) microfluidic devices provide unique opportunities to overcome some of these challenges given their ability to model the structure and function of tissues and organs in vitro. Previously established organ chip models typically consist of heterogenous cell populations sourced from multiple donors, limiting their applications in patient-specific disease modeling and personalized medicine. In this study, we engineered a personalized glomerulus chip system reconstituted from human induced pluripotent stem (iPS) cell-derived vascular endothelial cells (ECs) and podocytes from a single patient. Our stem cell-derived kidney glomerulus chip successfully mimics the structure and some essential functions of the glomerular filtration barrier. We further modeled glomerular injury in our tissue chips by administering a clinically relevant dose of the chemotherapy drug Adriamycin. The drug disrupts the structural integrity of the endothelium and the podocyte tissue layers, leading to significant albuminuria as observed in patients with glomerulopathies. We anticipate that the personalized glomerulus chip model established in this report could help advance future studies of kidney disease mechanisms and the discovery of personalized therapies. Given the remarkable ability of human iPS cells to differentiate into almost any cell type, this work also provides a blueprint for the establishment of more personalized organ chip and 'body-on-a-chip' models in the future.
    Keywords:  disease models; endothelial cells; glomerulus chip; human induced pluripotent stem cells; kidney glomerulus; microfluidics; organ-on-a-chip; personalized medicine; podocytes; stem cell technologies
    DOI:  https://doi.org/10.3390/mi12080967
  2. Exp Ther Med. 2021 Oct;22(4): 1070
      Bone microvascular endothelial cells (BMECs) constitute the central part of the femoral head's intramural microenvironment network and have an essential role in the development of steroid-induced osteonecrosis of the femoral head. Recently, the rapid development of microfluidic technology has led to innovations in the fields of chemistry, medicine and life sciences. It is now possible to use microfluidics organ-on-a-chip techniques to assess osteonecrosis. In the present study, BMECs were cultured on a microfluidic organ-on-a-chip platform to explore the pathogenesis of femoral-head necrosis. The aim of the present study was to explore the effects of different interventions on BMECs and study the pathogenesis of steroid-induced osteonecrosis through a microfluidic organ-on-a-chip platform. Methods including SU-8 lithography were used to produce a microfluidic organ-on-a-chip and human umbilical vein endothelial cells (HUVECs) were used to test whether it was possible to culture cells on the chip. Subsequently, a set of methods were applied for the isolation, purification, culture and identification of BMECs. Hydroxyapatite (HA) was used for co-culture, dexamethasone was used at different concentrations as an intervention in the cells and icariin was used for protection. BMECs were isolated and cultured from the femoral head obtained following total hip arthroplasty and were then inoculated into the microfluidic organ-on-a-chip for further treatment. In part I of the experiment, HUVECs and BMECs both successfully survived on the chip and a comparison of the growth and morphology was performed. HA and BMECs were then co-cultured for comparison with the control group. The cell growth was observed by confocal microscopy after 24 h. In part II, the effects of different concentrations of glucocorticoid (0.4 or 0.6 mg/ml dexamethasone) and the protection of icariin were evaluated. The morphology of BMECs and the cleaved caspase-3/7 content were observed by immunofluorescence staining and confocal microscopy after 24 h. In the microfluidic organ-on-a-chip, the response of the cells was able to be accurately observed. In part I, at the same concentration of injected cells, BMECs exhibited improved viability compared with HUVECs (P<0.05). In addition, it was indicated that HA was not only able to promote the germination and growth of BMECs but also improve the survival of the cells (P<0.05). In part II, it was identified that dexamethasone was able to induce BMECs to produce cleaved caspase 3/7; the caspase 3/7 content was significantly higher than that in the blank control group (P<0.05) and a dose correlation was observed. Icariin was able to inhibit this process and protect the microvascular structure of BMECs. The content of cleaved caspase 3/7 in the icariin-protected group was significantly lower than that in the group without icariin (P<0.05). It was concluded that BMECs are more likely to survive than HUVECs and HA promoted the growth of BMECs on the microfluidic organ-on-a-chip platform. Glucocorticoid caused damage to BMECs through the production of cleaved caspase 3/7, which was observed through the microfluidic organ-on-a-chip platform, and icariin protected BMECs from damage.
    Keywords:  apoptosis; bone microvascular endothelial cells; glucocorticoid; microfluidic chip; osteonecrosis
    DOI:  https://doi.org/10.3892/etm.2021.10504
  3. Micromachines (Basel). 2021 Aug 19. pii: 983. [Epub ahead of print]12(8):
      Kidney microphysiological systems (MPS) serve as potentially valuable preclinical instruments in probing mechanisms of renal clearance and osmoregulation. Current kidney MPS models target regions of the nephron, such as the glomerulus and proximal tubule (PCT), but fail to incorporate multiple filtration and absorption interfaces. Here, we describe a novel, partially open glomerulus and PCT microdevice that integrates filtration and absorption in a single MPS. The system equalizes pressure on each side of the PCT that operates with one side "closed" by recirculating into the bloodstream, and the other "opened" by exiting as primary filtrate. This design precisely controls the internal fluid dynamics and prevents loss of all fluid to the open side. Through this feature, an in vitro human glomerulus and proximal tubule MPS was constructed to filter human serum albumin and reabsorb glucose for seven days of operation. For proof-of-concept experiments, three human-derived cell types-conditionally immortalized human podocytes (CIHP-1), human umbilical vein endothelial cells (HUVECs), and human proximal tubule cells (HK-2)-were adapted into a common serum-free medium prior to being seeded into the three-component MPS (T-junction splitter, glomerular housing unit, and parallel proximal tubule barrier model). This system was optimized geometrically (tubing length, tubing internal diameter, and inlet flow rate) using in silico computational modeling. The prototype tri-culture MPS successfully filtered blood serum protein and generated albumin filtration in a physiologically realistic manner, while the device cultured only with proximal tubule cells did not. This glomerulus and proximal convoluted tubule MPS is a potential prototype for the human kidney used in both human-relevant testing and examining pharmacokinetic interactions.
    Keywords:  cytoskeleton; cytotoxicity; kidney; microfluidics; pharmacokinetics
    DOI:  https://doi.org/10.3390/mi12080983
  4. Sensors (Basel). 2021 Aug 17. pii: 5529. [Epub ahead of print]21(16):
      Microfluidic lab-on-chip devices are widely being developed for chemical and biological studies. One of the most commonly used types of these chips is perfusion microwells for culturing multicellular spheroids. The main challenge in such systems is the formation of substantial necrotic and quiescent zones within the cultured spheroids. Herein, we propose a novel acoustofluidic integrated platform to tackle this bottleneck problem. It will be shown numerically that such an approach is a potential candidate to be implemented to enhance cell viability and shrinks necrotic and quiescent zones without the need to increase the flow rate, leading to a significant reduction in costly reagents' consumption in conventional spheroid-on-a-chip platforms. Proof-of-concept, designing procedures and numerical simulation are discussed in detail. Additionally, the effects of acoustic and hydrodynamic parameters on the cultured cells are investigated. The results show that by increasing acoustic boundary displacement amplitude (d0), the spheroid's proliferating zone enlarges greatly. Moreover, it is shown that by implementing d0&nbsp; = 0.5 nm, the required flow rate to maintain the necrotic zone below 13% will be decreased 12 times compared to non-acoustic chips.
    Keywords:  acoustic microfluidics; lab-on-chip; necrotic; quiescent zones; spheroid-on-chip
    DOI:  https://doi.org/10.3390/s21165529
  5. Commun Biol. 2021 Aug 24. 4(1): 1001
      Microphysiological in vitro systems are platforms for preclinical evaluation of drug effects and significant advances have been made in recent years. However, existing microfluidic devices are not yet able to deliver compounds to cell models in a way that reproduces the real physiological drug exposure. Here, we introduce a novel tumour-on-chip microfluidic system that mimics the pharmacokinetic profile of compounds on 3D tumour spheroids to evaluate their response to the treatments. We used this platform to test the response of SW620 colorectal cancer spheroids to irinotecan (SN38) alone and in combination with the ATM inhibitor AZD0156, using concentrations mimicking mouse plasma exposure profiles of both agents. We explored spheroid volume and viability as a measure of cancer cells response and changes in mechanistically relevant pharmacodynamic biomarkers (γH2AX, cleaved-caspase 3 and Ki67). We demonstrate here that our microfluidic tumour-on-chip platform can successfully predict the efficacy from in vivo studies and therefore represents an innovative tool to guide drug dose and schedules for optimal efficacy and pharmacodynamic assessment, while reducing the need for animal studies.
    DOI:  https://doi.org/10.1038/s42003-021-02526-y
  6. Cancers (Basel). 2021 Aug 21. pii: 4208. [Epub ahead of print]13(16):
      Predicting patient responses to anticancer drugs is a major challenge both at the drug development stage and during cancer treatment. Tumor explant culture platforms (TECPs) preserve the native tissue architecture and are well-suited for drug response assays. However, tissue longevity in these models is relatively low. Several methodologies have been developed to address this issue, although no study has compared their efficacy in a controlled fashion. We investigated the effect of two variables in TECPs, specifically, the tissue size and culture vessel on tissue survival using micro-dissected tumor tissue (MDT) and tissue slices which were cultured in microfluidic chips and plastic well plates. Tumor models were produced from ovarian and prostate cancer cell line xenografts and were matched in terms of the specimen, total volume of tissue, and respective volume of medium in each culture system. We examined morphology, viability, and hypoxia in the various tumor models. Our observations suggest that the viability and proliferative capacity of MDTs were not affected during the time course of the experiments. In contrast, tissue slices had reduced proliferation and showed increased cell death and hypoxia under both culture conditions. Tissue slices cultured in microfluidic devices had a lower degree of hypoxia compared to those in 96-well plates. Globally, our results show that tissue slices have lower survival rates compared to MDTs due to inherent diffusion limitations, and that microfluidic devices may decrease hypoxia in tumor models.
    Keywords:  cancer treatment; drug screening assays; ex vivo model; hypoxia; tumor explant culture platform
    DOI:  https://doi.org/10.3390/cancers13164208
  7. Front Immunol. 2021 ;12 674727
      Electric fields are generated in vivo in a variety of physiologic and pathologic settings, including wound healing and immune response to injuries to epithelial barriers (e.g. lung pneumocytes). Immune cells are known to migrate towards both chemical (chemotaxis), physical (mechanotaxis) and electric stimuli (electrotaxis). Electrotaxis is the guided migration of cells along electric fields, and has previously been reported in T-cells and cancer cells. However, there remains a need for engineering tools with high spatial and temporal resolution to quantify EF guided migration. Here we report the development of an electrotaxis-on-chip (ETOC) platform that enables the quantification of dHL-60 cell, a model neutrophil-like cell line, migration toward both electrical and chemoattractant gradients. Neutrophils are the most abundant white blood cells and set the stage for the magnitude of the immune response. Therefore, developing engineering tools to direct neutrophil migration patterns has applications in both infectious disease and inflammatory disorders. The ETOC developed in this study has embedded electrodes and four migration zones connected to a central cell-loading chamber with migration channels [10 µm X 10 µm]. This device enables both parallel and competing chemoattractant and electric fields. We use our novel ETOC platform to investigate dHL-60 cell migration in three biologically relevant conditions: 1) in a DC electric field; 2) parallel chemical gradient and electric fields; and 3) perpendicular chemical gradient and electric field. In this study we used differentiated leukemia cancer cells (dHL60 cells), an accepted model for human peripheral blood neutrophils. We first quantified effects of electric field intensities (0.4V/cm-1V/cm) on dHL-60 cell electrotaxis. Our results show optimal migration at 0.6 V/cm. In the second scenario, we tested whether it was possible to increase dHL-60 cell migration to a bacterial signal [N-formylated peptides (fMLP)] by adding a parallel electric field. Our results show that there was significant increase (6-fold increase) in dHL60 migration toward fMLP and cathode of DC electric field (0.6V/cm, n=4, p-value<0.005) vs. fMLP alone. Finally, we evaluated whether we could decrease or re-direct dHL-60 cell migration away from an inflammatory signal [leukotriene B4 (LTB4)]. The perpendicular electric field significantly decreased migration (2.9-fold decrease) of dHL60s toward LTB4 vs. LTB4 alone. Our microfluidic device enabled us to quantify single-cell electrotaxis velocity (7.9 µm/min ± 3.6). The magnitude and direction of the electric field can be more precisely and quickly changed than most other guidance cues such as chemical cues in clinical investigation. A better understanding of EF guided cell migration will enable the development of new EF-based treatments to precisely direct immune cell migration for wound care, infection, and other inflammatory disorders.
    Keywords:  electrotaxis; immunomodulation; microfluidics; migration; neutrophil; wound healing
    DOI:  https://doi.org/10.3389/fimmu.2021.674727
  8. Cancers (Basel). 2021 Aug 05. pii: 3930. [Epub ahead of print]13(16):
      Recent advances in immunotherapies and molecularly targeted therapies have led to an increased interest in exploring the field of in vitro tumor mimetic platforms. An increasing need to understand the mechanisms of anti-cancer therapies has led to the development of natural tumor tissue-like in vitro platforms capable of simulating the tumor microenvironment. The incorporation of vascular structures into the in vitro platforms could be a crucial factor for functional investigation of most anti-cancer therapies, including immunotherapies, which are closely related to the circulatory system. Decellularized lung extracellular matrix (ldECM), comprised of ECM components and pro-angiogenic factors, can initiate vascularization and is ideal for mimicking the natural microenvironment. In this study, we used a ldECM-based hydrogel to develop a 3D vascularized lung cancer-on-a-chip (VLCC). We specifically encapsulated tri-cellular spheroids made from A549 cells, HUVECs, and human lung fibroblasts, for simulating solid type lung cancer. Additionally, two channels were incorporated in the hydrogel construct to mimic perfusable vessel structures that resemble arterioles or venules. Our study highlights how a more effective dose-dependent action of the anti-cancer drug Doxorubicin was observed using a VLCC over 2D screening. This observation confirmed the potential of the VLCC as a 3D in vitro drug screening tool.
    Keywords:  angiogenesis; cancer-on-a-chip; decellularized extracellular matrix; drug screening; tumor microenvironment; vascularization
    DOI:  https://doi.org/10.3390/cancers13163930
  9. Sci Rep. 2021 Aug 23. 11(1): 17028
      In order to circumvent the limited access and donor variability of human primary alveolar cells, directed differentiation of human pluripotent stem cells (hiPSCs) into alveolar-like cells, provides a promising tool for respiratory disease modeling and drug discovery assays. In this work, a unique, miniaturized 96-Transwell microplate system is described where hiPSC-derived alveolar-like cells were cultured at an air-liquid interface (ALI). To this end, hiPSCs were differentiated into lung epithelial progenitor cells (LPCs) and subsequently matured into a functional alveolar type 2 (AT2)-like epithelium with monolayer-like morphology. AT2-like cells cultured at the physiological ALI conditions displayed characteristics of AT2 cells with classical alveolar surfactant protein expressions and lamellar-body like structures. The integrity of the epithelial barriers between the AT2-like cells was confirmed by applying a custom-made device for 96-parallelized transepithelial electric resistance (TEER) measurements. In order to generate an IPF disease-like phenotype in vitro, the functional AT2-like cells were stimulated with cytokines and growth factors present in the alveolar tissue of IPF patients. The cytokines stimulated the secretion of pro-fibrotic biomarker proteins both on the mRNA (messenger ribonucleic acid) and protein level. Thus, the hiPSC-derived and cellular model system enables the recapitulation of certain IPF hallmarks, while paving the route towards a miniaturized medium throughput approach of pharmaceutical drug discovery.
    DOI:  https://doi.org/10.1038/s41598-021-96565-4
  10. Int J Mol Sci. 2021 Aug 18. pii: 8887. [Epub ahead of print]22(16):
      Immunotherapy of bladder cancer is known to have favorable effects, although it is difficult to determine which patients will show a good response because of the different tumor microenvironments (TME). Here, we developed a bladder cancer-on-a-chip (BCOC) to mimic the TME using three-dimensional (3D) bioprinting and microfluidic technology. We fabricated a T24 and a 5637-cell line-based BCOC that also incorporated MRC-5, HUVEC, and THP-1 cells. We evaluated the effects of TME and assessed the immunologic reactions in response to different concentrations of Bacillus Calmette-Guérin (BCG) via live/dead assay and THP-1 monocytic migration, and concentrations of growth factors and cytokines. The results show that cell viability was maintained at 15% filling density in circle-shaped cell constructs at 20 μL/min microfluidic flow rate. A 3D co-culture increased the proliferation of BCOCs. We found that the appropriate time to evaluate the viability of BCOC, concentration of cytokines, and migration of monocytes was 6 h, 24 h, and three days after BGC treatment. Lastly, the immunotherapeutic effects of BCOC increased according to BCG dosage. To predict effects of immunotherapeutic agent in bladder cancer, we constructed a 3D bioprinted BCOC model. The BCOC was validated with BCG, which has been proven to be effective in the immunotherapy of bladder cancer.
    Keywords:  3D bioprinting; BCG vaccine; intravesical administration; urinary bladder neoplasms
    DOI:  https://doi.org/10.3390/ijms22168887