Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Angiogenesis ; 27(1): 37-49, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37493987

ABSTRACT

Modern drug development increasingly requires comprehensive models that can be utilized in the earliest stages of compound and target discovery. Here we report a phenotypic screening exercise in a high-throughput Organ-on-a-Chip setup. We assessed the inhibitory effect of 1537 protein kinase inhibitors in an angiogenesis assay. Over 4000 micro-vessels were grown under perfusion flow in microfluidic chips, exposed to a cocktail of pro-angiogenic factors and subsequently exposed to the respective kinase inhibitors. Efficacy of compounds was evaluated by reduced angiogenic sprouting, whereas reduced integrity of the main micro-vessel was taken as a measure for toxicity. The screen yielded 53 hits with high anti-angiogenicity and low toxicity, of which 44 were previously unassociated with angiogenic pathways. This study demonstrates that Organ-on-a-Chip models can be screened in high numbers to identify novel compounds and targets. This will ultimately reduce bias in early-stage drug development and increases probability to identify first in class compounds and targets for today's intractable diseases.


Subject(s)
Angiogenesis , Antineoplastic Agents , Humans , Microphysiological Systems , Antineoplastic Agents/therapeutic use , Neovascularization, Pathologic/drug therapy , Protein Kinase Inhibitors/pharmacology
2.
Angiogenesis ; 25(4): 455-470, 2022 11.
Article in English | MEDLINE | ID: mdl-35704148

ABSTRACT

With recent progress in modeling liver organogenesis and regeneration, the lack of vasculature is becoming the bottleneck in progressing our ability to model human hepatic tissues in vitro. Here, we introduce a platform for routine grafting of liver and other tissues on an in vitro grown microvascular bed. The platform consists of 64 microfluidic chips patterned underneath a 384-well microtiter plate. Each chip allows the formation of a microvascular bed between two main lateral vessels by inducing angiogenesis. Chips consist of an open-top microfluidic chamber, which enables addition of a target tissue by manual or robotic pipetting. Upon grafting a liver microtissue, the microvascular bed undergoes anastomosis, resulting in a stable, perfusable vascular network. Interactions with vasculature were found in spheroids and organoids upon 7 days of co-culture with space of Disse-like architecture in between hepatocytes and endothelium. Veno-occlusive disease was induced by azathioprine exposure, leading to impeded perfusion of the vascularized spheroid. The platform holds the potential to replace animals with an in vitro alternative for routine grafting of spheroids, organoids, or (patient-derived) explants.


Subject(s)
Microfluidics , Organoids , Animals , Azathioprine , Coculture Techniques , Humans , Liver , Microfluidics/methods
3.
Int J Mol Sci ; 22(10)2021 May 19.
Article in English | MEDLINE | ID: mdl-34069732

ABSTRACT

Lowe syndrome and Dent II disease are X-linked monogenetic diseases characterised by a renal reabsorption defect in the proximal tubules and caused by mutations in the OCRL gene, which codes for an inositol-5-phosphatase. The life expectancy of patients suffering from Lowe syndrome is largely reduced because of the development of chronic kidney disease and related complications. There is a need for physiological human in vitro models for Lowe syndrome/Dent II disease to study the underpinning disease mechanisms and to identify and characterise potential drugs and drug targets. Here, we describe a proximal tubule organ on chip model combining a 3D tubule architecture with fluid flow shear stress that phenocopies hallmarks of Lowe syndrome/Dent II disease. We demonstrate the high suitability of our in vitro model for drug target validation. Furthermore, using this model, we demonstrate that proximal tubule cells lacking OCRL expression upregulate markers typical for epithelial-mesenchymal transition (EMT), including the transcription factor SNAI2/Slug, and show increased collagen expression and deposition, which potentially contributes to interstitial fibrosis and disease progression as observed in Lowe syndrome and Dent II disease.


Subject(s)
Genetic Diseases, X-Linked/metabolism , Kidney Tubules, Proximal/metabolism , Nephrolithiasis/metabolism , Oculocerebrorenal Syndrome/metabolism , Humans , Lab-On-A-Chip Devices , Models, Biological , Mutation , Phenotype , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism
4.
Int J Mol Sci ; 20(18)2019 Sep 19.
Article in English | MEDLINE | ID: mdl-31546820

ABSTRACT

Pancreatic Ductal Adenocarcinoma (PDAC) is one of the most lethal cancers due to a high chemoresistance and poor vascularization, which results in an ineffective systemic therapy. PDAC is characterized by a high intratumoral pressure, which is not captured by current 2D and 3D in vitro models. Here, we demonstrated a 3D microfluidic interstitial flow model to mimic the intratumoral pressure in PDAC. We found that subjecting the S2-028 PDAC cell line to interstitial flow inhibits the proliferation, while maintaining a high viability. We observed increased gemcitabine chemoresistance, with an almost nine-fold higher EC50 as compared to a monolayer culture (31 nM versus 277 nM), and an alleviated expression and function of the multidrug resistance protein (MRP) family. In conclusion, we developed a 3D cell culture modality for studying intratissue pressure and flow that exhibits more predictive capabilities than conventional 2D cell culture and is less time-consuming, and more scalable and accessible than animal models. This increase in microphysiological relevance might support improved efficiency in the drug development pipeline.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Intestine, Small/metabolism , Microfluidic Analytical Techniques , Models, Biological , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Deoxycytidine/pharmacology , Humans , Intestine, Small/pathology , Lab-On-A-Chip Devices , Pancreatic Neoplasms/pathology , Gemcitabine
5.
Int J Mol Sci ; 20(22)2019 Nov 12.
Article in English | MEDLINE | ID: mdl-31726729

ABSTRACT

A common bottleneck in any drug development process is finding sufficiently accurate models that capture key aspects of disease development and progression. Conventional drug screening models often rely on simple 2D culture systems that fail to recapitulate the complexity of the organ situation. In this study, we show the application of a robust high throughput 3D gut-on-a-chip model for investigating hallmarks of inflammatory bowel disease (IBD). Using the OrganoPlate platform, we subjected enterocyte-like cells to an immune-relevant inflammatory trigger in order to recapitulate key events of IBD and to further investigate the suitability of this model for compound discovery and target validation activities. The induction of inflammatory conditions caused a loss of barrier function of the intestinal epithelium and its activation by increased cytokine production, two events observed in IBD physiopathology. More importantly, anti-inflammatory compound exposure prevented the loss of barrier function and the increased cytokine release. Furthermore, knockdown of key inflammatory regulators RELA and MYD88 through on-chip adenoviral shRNA transduction alleviated IBD phenotype by decreasing cytokine production. In summary, we demonstrate the routine use of a gut-on-a-chip platform for disease-specific aspects modeling. The approach can be used for larger scale disease modeling, target validation and drug discovery purposes.


Subject(s)
Drug Discovery , Inflammatory Bowel Diseases , Microchip Analytical Procedures , Models, Biological , Caco-2 Cells , Drug Evaluation, Preclinical , Gene Knockout Techniques , Humans , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Lab-On-A-Chip Devices , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
6.
BMC Cancer ; 17(1): 709, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29096610

ABSTRACT

BACKGROUND: Breast cancer is the most common invasive cancer among women. Currently, there are only a few models used for therapy selection, and they are often poor predictors of therapeutic response or take months to set up and assay. In this report, we introduce a microfluidic OrganoPlate® platform for extracellular matrix (ECM) embedded tumor culture under perfusion as an initial study designed to investigate the feasibility of adapting this technology for therapy selection. METHODS: The triple negative breast cancer cell lines MDA-MB-453, MDA-MB-231 and HCC1937 were selected based on their different BRCA1 and P53 status, and were seeded in the platform. We evaluate seeding densities, ECM composition (Matrigel®, BME2rgf, collagen I) and biomechanical (perfusion vs static) conditions. We then exposed the cells to a series of anti-cancer drugs (paclitaxel, olaparib, cisplatin) and compared their responses to those in 2D cultures. Finally, we generated cisplatin dose responses in 3D cultures of breast cancer cells derived from 2 PDX models. RESULTS: The microfluidic platform allows the simultaneous culture of 96 perfused micro tissues, using limited amounts of material, enabling drug screening of patient-derived material. 3D cell culture viability is improved by constant perfusion of the medium. Furthermore, the drug response of these triple negative breast cancer cells was attenuated by culture in 3D and differed from that observed in 2D substrates. CONCLUSIONS: We have investigated the use of a high-throughput organ-on-a-chip platform to select therapies. Our results have raised the possibility to use this technology in personalized medicine to support selection of appropriate drugs and to predict response to therapy in a real time fashion.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Culture Techniques/methods , Extracellular Matrix/metabolism , Microfluidics/methods , BRCA1 Protein/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Collagen , Drug Combinations , Female , Humans , Laminin , Mutation , Outcome Assessment, Health Care/methods , Paclitaxel/pharmacology , Phthalazines/pharmacology , Piperazines/pharmacology , Prognosis , Proteoglycans , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
STAR Protoc ; 5(1): 102879, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38358879

ABSTRACT

Organ-on-a-chip technology incorporating stem cell techniques represents a promising strategy to improve modeling of human organs. Here, we present a protocol for generating a standardized 3D placenta-on-a-chip model using trophoblast derived from human induced pluripotent stem cells (hiPSCs). We describe steps for seeding hiPSCs into multi-chip OrganoPlate devices and on-chip differentiation into trophoblasts against an extracellular matrix under perfused conditions. We then detail procedures for conducting a functional barrier integrity assay, immunostaining, and collecting protein or RNA for molecular analysis. For complete details on the use and execution of this protocol, please refer to Lermant et al. (2023).1.


Subject(s)
Induced Pluripotent Stem Cells , Pregnancy , Female , Humans , Placenta , Trophoblasts , Cell Differentiation , Lab-On-A-Chip Devices
8.
iScience ; 26(7): 107240, 2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37534160

ABSTRACT

Although recently developed placenta-on-chip systems opened promising perspectives for placental barrier modeling, they still lack physiologically relevant trophoblasts and are poorly amenable to high-throughput studies. We aimed to implement human-induced pluripotent stem cells (hiPSC)-derived trophoblasts into a multi-well microfluidic device to develop a physiologically relevant and scalable placental barrier model. When cultured in a perfused micro-channel against a collagen-based matrix, hiPSC-derived trophoblasts self-arranged into a 3D structure showing invasive behavior, fusogenic and endocrine activities, structural integrity, and expressing placental transporters. RNA-seq analysis revealed that the microfluidic 3D environment boosted expression of genes related to early placental structural development, mainly involved in mechanosensing and cell surface receptor signaling. These results demonstrated the feasibility of generating a differentiated primitive syncytium from hiPSC in a microfluidic platform. Besides expanding hiPSC-derived trophoblast scope of applications, this study constitutes an important resource to improve placental barrier models and boost research and therapeutics evaluation in pregnancy.

9.
STAR Protoc ; 4(1): 102051, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36861838

ABSTRACT

Traditionally, to quantify permeability of a biological barrier, the initial slope is used, based on the assumption of sink condition (concentration of the donor is constant, and the receiver increases less than 10%). With on-a-chip barrier models, this assumption fails in cell-free or leaky conditions, which requires the use of the exact solution. To encounter a time delay from performing the assay and acquiring the data, we present a protocol with the exact equation modified to incorporate a time offset.


Subject(s)
Biological Assay , Lab-On-A-Chip Devices , Permeability
10.
Acta Biomater ; 164: 363-376, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37116636

ABSTRACT

Pathologies associated with uteroplacental hypoxia, such as preeclampsia are among the leading causes of maternal and perinatal morbidity in the world. Its fundamental mechanisms are yet poorly understood due to a lack of good experimental models. Here we report an in vitro model of the placental barrier, based on co-culture of trophoblasts and endothelial cells against a collagen extracellular matrix in a microfluidic platform. The model yields a functional syncytium with barrier properties, polarization, secretion of relevant extracellular membrane components, thinning of the materno-fetal space, hormone secretion, and transporter function. The model is exposed to low oxygen conditions and perfusion flow is modulated to induce a pathological environment. This results in reduced barrier function, hormone secretion, and microvilli as well as an increased nuclei count, characteristics of preeclamptic placentas. The model is implemented in a titer plate-based microfluidic platform fully amenable to high-throughput screening. We thus believe this model could aid mechanistic understanding of preeclampsia and other placental pathologies associated with hypoxia/ischemia, as well as support future development of effective therapies through target and compound screening campaigns. STATEMENT OF SIGNIFICANCE: The human placenta is a unique organ sustaining fetal growth but is also the source of severe pathologies, such as preeclampsia. Though leading cause of perinatal mortality in the world, preeclampsia remains untreatable due to a lack of relevant in vitro placenta models. To better understand the pathology, we have developed 3D placental barrier models in a microfluidic device. The platform allows parallel culture of 40 perfused physiological miniaturized placental barriers, comprising a differentiated syncytium and endothelium that have been validated for transporter functions. Exposure to a hypoxic and ischemic environment enabled the mimicking of preeclamptic characteristics in high-throughput, which we believe could lead to a better understanding of the pathology as well as support future effective therapies development.


Subject(s)
Placenta , Pre-Eclampsia , Pregnancy , Female , Humans , Endothelial Cells , Hypoxia , Ischemia , Lab-On-A-Chip Devices , Hormones
11.
Biochem Biophys Res Commun ; 422(1): 169-73, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22575449

ABSTRACT

The small viral protein apoptin is capable of inducing apoptosis selectively in human tumor cells. In normal cells apoptin localizes in the cytoplasm where it forms aggregates, becomes epitope-shielded and eventually degraded. By inhibiting the proteasome activity with the chemical inhibitors bortezomib and Ada-Ahx(3)L(3)VS apoptin levels can be stabilized in normal cells similar to the tumor suppressor p53 protein. In contrast, proteasome inhibition in tumor cells did not affect the apoptin stability while it still stabilized p53 levels. Apparently, apoptin is degraded by proteasomal activity in normal human cells, a process that no longer takes place in tumor cells. This loss of proteasomal susceptibility appears to be specific for apoptin.


Subject(s)
Capsid Proteins/metabolism , Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Bortezomib , Capsid Proteins/genetics , Capsid Proteins/pharmacology , Cell Line , Cell Line, Tumor , Fibroblasts/metabolism , Humans , Mutation , Phosphorylation , Protein Stability , Proteolysis , Pyrazines/pharmacology , Tumor Suppressor Protein p53/metabolism
12.
Anal Biochem ; 421(1): 68-74, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22080040

ABSTRACT

Apoptin, a protein derived from chicken anemia virus (CAV), induces apoptosis selectively in human tumor cells as compared with normal cells. This activity depends on phosphorylation and relocation of apoptin to the nucleus of cancer cells. Here, we describe an in vitro kinase assay that allows the biochemical characterization of apoptin kinase activity in tumor cells. The kinase phosphorylates apoptin in a strictly ATP-dependent fashion and in a broad salt range. The kinase activity is present constitutively in both cytoplasm and nucleus of various human tumor cells. Q-column chromatography showed that both cytoplasmic and nuclear fractions have identical fractionation characteristics, suggesting that the same kinase is present in both cellular compartments. Kinase activity derived from positive Q-column fractions bound to amylose-maltose-binding protein (MBP)-apoptin and could be eluted with ATP only in the presence of the cofactor Mg(2+). Apparently, unphosphorylated apoptin interacts with the kinase and is released only after phosphorylation has occurred, proving that our assay recognizes the genuine apoptin kinase. This is further corroborated by the finding that apoptin is phosphorylated in vitro at positions Thr108 and Thr107, in concert with earlier in vivo observations. Our assay excludes cyclin-dependent kinase 2 (CDK2) and protein kinase C beta (PKC-ß), previously nominated by two separate studies as being the genuine apoptin kinase.


Subject(s)
Capsid Proteins/metabolism , Capsid Proteins/pharmacology , Neoplasms/metabolism , Protein Kinases/analysis , Active Transport, Cell Nucleus , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Apoptosis/drug effects , Capsid Proteins/chemistry , Capsid Proteins/genetics , Cell Line, Tumor , Cell Nucleus/enzymology , Cyclin-Dependent Kinase 2/metabolism , Cytoplasm/enzymology , HeLa Cells , Humans , In Vitro Techniques , Jurkat Cells , Kinetics , Mass Spectrometry , Neoplasms/drug therapy , Neoplasms/pathology , Phosphorylation , Protein Kinase C/metabolism , Protein Kinases/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Threonine/chemistry
13.
Methods Mol Biol ; 2373: 87-105, 2022.
Article in English | MEDLINE | ID: mdl-34520008

ABSTRACT

The study of epithelial barrier properties in the human body is of paramount interest to a range of disciplines, including disease modeling, drug transport studies, toxicology, developmental biology, and regenerative biology. Current day in vitro studies largely rely on growing epithelial cells in a static environment on membrane cell culture inserts. With the advancement of microfluidic and organ-on-a-chip techniques it became possible to culture 3D intestinal tubules directly against an extracellular matrix (ECM) under flow and without the need for artificial membranes. Here we describe detailed protocols for culturing epithelial tubules in a high-throughput format, assessing their permeability and marker expression. The platform harbors 40 independent microfluidic chips in a microtiter plate format. The resulting 40 epithelial tubules are analyzed in parallel using a high-content microscopy. Protocols described here allow for adoption and routine application of microfluidic techniques by nonspecialized end-users.


Subject(s)
Intestinal Mucosa , Lab-On-A-Chip Devices , Cell Culture Techniques , Epithelial Cells , Humans , Microfluidics
14.
Redox Biol ; 57: 102488, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36201911

ABSTRACT

Reactive oxygen species (ROS) have different properties and biological functions. They contribute to cell signaling and, in excessive amounts, to oxidative stress (OS). Although ROS is pivotal in a wide number of physiological systems and pathophysiological processes, direct quantification in vivo is quite challenging and mainly limited to in vitro studies. Even though advanced in vitro cell culture techniques, like on-a-chip culture, have overcome the lack of crucial in vivo-like physiological aspects in 2D culture, the majority of in vitro ROS quantification studies are generally performed in 2D. Here we report the development, application, and validation of a multiplexed assay to quantify ROS and cell viability in organ-on-a-chip models. The assay utilizes three dyes to stain live cells for ROS, dead cells, and DNA. Confocal images were analyzed to quantify ROS probes and determine the number of nuclei and dead cells. We found that, in contrast to what has been reported with 2D cell culture, on-a-chip models are more prone to scavenge ROS rather than accumulate them. The assay is sensitive enough to distinguish between different phenotypes of endothelial cells (ECs) based on the level of OS to detect higher level in tumor than normal cells. Our results indicate that the use of physiologically relevant models and this assay could help unravelling the mechanisms behind OS and ROS accumulation. A further step could be taken in data analysis by implementing AI in the pipeline to also analyze images for morphological changes to have an even broader view of OS mechanism.

15.
Kidney360 ; 3(2): 217-231, 2022 02 24.
Article in English | MEDLINE | ID: mdl-35373131

ABSTRACT

Background: Renal ischemia/reperfusion injury (rIRI) is one of the major causes of AKI. Although animal models are suitable for investigating systemic symptoms of AKI, they are limited in translatability. Human in vitro models are crucial in giving mechanistic insights into rIRI; however, they miss out on crucial aspects such as reperfusion injury and the multitissue aspect of AKI. Methods: We advanced the current renal proximal tubule-on-a-chip model to a coculture model with a perfused endothelial vessel separated by an extracellular matrix. The coculture was characterized for its three-dimensional structure, protein expression, and response to nephrotoxins. Then, rIRI was captured through control of oxygen levels, nutrient availability, and perfusion flow settings. Injury was quantified through morphologic assessment, caspase-3/7 activation, and cell viability. Results: The combination of low oxygen, reduced glucose, and interrupted flow was potent to disturb the proximal tubules. This effect was strongly amplified upon reperfusion. Endothelial vessels were less sensitive to the ischemia-reperfusion parameters. Adenosine treatment showed a protective effect on the disruption of the epithelium and on the caspase-3/7 activation. Conclusions: A human in vitro rIRI model was developed using a coculture of a proximal tubule and blood vessel on-a-chip, which was used to characterize the renoprotective effect of adenosine. The robustness of the model and assays in combination with the throughput of the platform make it ideal to advance pathophysiological research and enable the development of novel therapeutic modalities.


Subject(s)
Acute Kidney Injury , Lab-On-A-Chip Devices , Acute Kidney Injury/prevention & control , Animals , Humans , Ischemia/complications , Kidney Tubules/metabolism , Reperfusion/adverse effects
16.
Sci Rep ; 12(1): 16930, 2022 10 08.
Article in English | MEDLINE | ID: mdl-36209279

ABSTRACT

In early systemic sclerosis (Scleroderma, SSc), the vasculature is impaired. Although the exact etiology of endothelial cell damage in SSc remains unclear, it is hypothesized that endothelial to mesenchymal transition (EndoMT) plays a key role. To perform physiologically relevant angiogenic studies, we set out to develop an angiogenesis-on-a-chip platform that is suitable for assessing disease parameters that are relevant to SSc and other vasculopathies. In the model, we substituted Fetal Bovine Serum (FBS) with Human Serum without impairing the stability of the culture. We showed that 3D microvessels and angiogenic factor-induced sprouts exposed to key pro-inflammatory and pro-fibrotic cytokines (TNFα and TGFß) undergo structural alterations consisting of destructive vasculopathy (loss of small vessels). We also showed that these detrimental effects can be prevented by compound-mediated inhibition of TGFß-ALK5 signaling or addition of a TNFα neutralizing antibody to the 3D cultures. This demonstrates that our in vitro model is suitable for compound testing and identification of new drugs that can protect from microvascular destabilization or regression in disease-mimicking conditions. To support this, we demonstrated that sera obtained from SSc patients can exert an anti-angiogenic effect on the 3D vessel model, opening the doors to screening for potential SSc drugs, enabling direct patient translatability and personalization of drug treatment.


Subject(s)
Scleroderma, Systemic , Tumor Necrosis Factor-alpha , Angiogenesis Inducing Agents , Antibodies, Neutralizing , Humans , Lab-On-A-Chip Devices , Microvessels , Neovascularization, Pathologic , Serum Albumin, Bovine , Transforming Growth Factor beta
17.
Fluids Barriers CNS ; 18(1): 59, 2021 Dec 14.
Article in English | MEDLINE | ID: mdl-34906183

ABSTRACT

BACKGROUND: In ischemic stroke, the function of the cerebral vasculature is impaired. This vascular structure is formed by the so-called neurovascular unit (NVU). A better understanding of the mechanisms involved in NVU dysfunction and recovery may lead to new insights for the development of highly sought therapeutic approaches. To date, there remains an unmet need for complex human in vitro models of the NVU to study ischemic events seen in the human brain. METHODS: We here describe the development of a human NVU on-a-chip model using a platform that allows culture of 40 chips in parallel. The model comprises a perfused vessel of primary human brain endothelial cells in co-culture with induced pluripotent stem cell derived astrocytes and neurons. Ischemic stroke was mimicked using a threefold approach that combines chemical hypoxia, hypoglycemia, and halted perfusion. RESULTS: Immunofluorescent staining confirmed expression of endothelial adherens and tight junction proteins, as well as astrocytic and neuronal markers. In addition, the model expresses relevant brain endothelial transporters and shows spontaneous neuronal firing. The NVU on-a-chip model demonstrates tight barrier function, evidenced by retention of small molecule sodium fluorescein in its lumen. Exposure to the toxic compound staurosporine disrupted the endothelial barrier, causing reduced transepithelial electrical resistance and increased permeability to sodium fluorescein. Under stroke mimicking conditions, brain endothelial cells showed strongly reduced barrier function (35-fold higher apparent permeability) and 7.3-fold decreased mitochondrial potential. Furthermore, levels of adenosine triphosphate were significantly reduced on both the blood- and the brain side of the model (4.8-fold and 11.7-fold reduction, respectively). CONCLUSIONS: The NVU on-a-chip model presented here can be used for fundamental studies of NVU function in stroke and other neurological diseases and for investigation of potential restorative therapies to fight neurological disorders. Due to the platform's relatively high throughput and compatibility with automation, the model holds potential for drug compound screening.


Subject(s)
Astrocytes , Endothelial Cells , Induced Pluripotent Stem Cells , Ischemic Stroke , Lab-On-A-Chip Devices , Models, Biological , Neurons , Neurovascular Coupling , Humans
18.
Nat Protoc ; 16(4): 2023-2050, 2021 04.
Article in English | MEDLINE | ID: mdl-33674788

ABSTRACT

Advanced in vitro kidney models are of great importance to the study of renal physiology and disease. Kidney tubuloids can be established from primary cells derived from adult kidney tissue or urine. Tubuloids are three-dimensional multicellular structures that recapitulate tubular function and have been used to study infectious, malignant, metabolic, and genetic diseases. For tubuloids to more closely represent the in vivo kidney, they can be integrated into an organ-on-a-chip system that has a more physiological tubular architecture and allows flow and interaction with vasculature or epithelial and mesenchymal cells from other organs. Here, we describe a detailed protocol for establishing tubuloid cultures from tissue and urine (1-3 weeks), as well as for generating and characterizing tubuloid cell-derived three-dimensional tubular structures in a perfused microfluidic multi-chip platform (7 d). The combination of the two systems yields a powerful in vitro tool that better recapitulates the complexity of the kidney tubule with donor-specific properties.


Subject(s)
Kidney Tubules/growth & development , Lab-On-A-Chip Devices , Organoids/growth & development , Perfusion , Tissue Culture Techniques/methods , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Cell Fractionation , Child , Child, Preschool , Electric Impedance , Female , Fluorescent Dyes/chemistry , Humans , Infant , Male , Membrane Transport Proteins/metabolism , Microfluidics , Middle Aged , Rats , Young Adult
19.
J Pharm Sci ; 110(4): 1601-1614, 2021 04.
Article in English | MEDLINE | ID: mdl-33545187

ABSTRACT

Proximal tubule epithelial cells (PTEC) are susceptible to drug-induced kidney injury (DIKI). Cell-based, two-dimensional (2D) in vitro PTEC models are often poor predictors of DIKI, probably due to the lack of physiological architecture and flow. Here, we assessed a high throughput, 3D microfluidic platform (Nephroscreen) for the detection of DIKI in pharmaceutical development. This system was established with four model nephrotoxic drugs (cisplatin, tenofovir, tobramycin and cyclosporin A) and tested with eight pharmaceutical compounds. Measured parameters included cell viability, release of lactate dehydrogenase (LDH) and N-acetyl-ß-d-glucosaminidase (NAG), barrier integrity, release of specific miRNAs, and gene expression of toxicity markers. Drug-transporter interactions for P-gp and MRP2/4 were also determined. The most predictive read outs for DIKI were a combination of cell viability, LDH and miRNA release. In conclusion, Nephroscreen detected DIKI in a robust manner, is compatible with automated pipetting, proved to be amenable to long-term experiments, and was easily transferred between laboratories. This proof-of-concept-study demonstrated the usability and reproducibility of Nephroscreen for the detection of DIKI and drug-transporter interactions. Nephroscreen it represents a valuable tool towards replacing animal testing and supporting the 3Rs (Reduce, Refine and Replace animal experimentation).


Subject(s)
Kidney Tubules, Proximal , Lab-On-A-Chip Devices , Animals , Drug Interactions , Humans , Kidney , Reproducibility of Results
20.
SLAS Technol ; 25(6): 585-597, 2020 12.
Article in English | MEDLINE | ID: mdl-32576063

ABSTRACT

Development of efficient drugs and therapies for the treatment of inflammatory conditions in the intestine is often hampered by the lack of reliable, robust, and high-throughput in vitro and in vivo models. Current models generally fail to recapitulate key aspects of the intestine, resulting in low translatability to the human situation. Here, an immunocompetent 3D perfused intestine-on-a-chip platform was developed and characterized for studying intestinal inflammation. Forty independent polarized 3D perfused epithelial tubular structures were grown from cells of mixed epithelial origin, including enterocytes (Caco-2) and goblet cells (HT29-MTX-E12). Immune cells THP-1 and MUTZ-3, which can be activated, were added to the system and assessed for cytokine release. Intestinal inflammation was mimicked through exposure to tumor necrosis factor-α (TNFα) and interleukin (IL)-1ß. The effects were quantified by measuring transepithelial electrical resistance (TEER) and proinflammatory cytokine secretion on the apical and basal sides. Cytokines induced an inflammatory state in the culture, as demonstrated by the impaired barrier function and increased IL-8 secretion. Exposure to the known anti-inflammatory drug TPCA-1 prevented the inflammatory state. The model provides biological modularity for key aspects of intestinal inflammation, making use of well-established cell lines. This allows robust assays that can be tailored in complexity to serve all preclinical stages in the drug discovery and development process.


Subject(s)
Intestinal Mucosa , Lab-On-A-Chip Devices , Caco-2 Cells , Humans , Intestines
SELECTION OF CITATIONS
SEARCH DETAIL