Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Future Sci OA ; 5(8): FSO413, 2019 Sep 10.
Article in English | MEDLINE | ID: mdl-31534781

ABSTRACT

Microphysiological systems play a pivotal role in progressing toward a global paradigm shift in drug development. Here, we designed a four-organ-chip interconnecting miniaturized human intestine, liver, brain and kidney equivalents. All four organ models were predifferentiated from induced pluripotent stem cells from the same healthy donor and integrated into the microphysiological system. The coculture of the four autologous tissue models in one common medium deprived of tissue specific growth factors was successful over 14-days. Although there were no added growth factors present in the coculture medium, the intestine, liver and neuronal model maintained defined marker expression. Only the renal model was overgrown by coexisting cells and did not further differentiate. This model platform will pave the way for autologous coculture cross-talk assays, disease induction and subsequent drug testing.

2.
Sci Rep ; 8(1): 15010, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30301942

ABSTRACT

Antibody therapies targeting the epithelial growth factor receptor (EGFR) are being increasingly applied in cancer therapy. However, increased tumour containment correlates proportionally with the severity of well-known adverse events in skin. The prediction of the latter is not currently possible in conventional in vitro systems and limited in existing laboratory animal models. Here we established a repeated dose "safficacy" test assay for the simultaneous generation of safety and efficacy data. Therefore, a commercially available multi-organ chip platform connecting two organ culture compartments was adapted for the microfluidic co-culture of human H292 lung cancer microtissues and human full-thickness skin equivalents. Repeated dose treatment of the anti-EGFR-antibody cetuximab showed an increased pro-apoptotic related gene expression in the tumour microtissues. Simultaneously, proliferative keratinocytes in the basal layer of the skin microtissues were eliminated, demonstrating crucial inhibitory effects on the physiological skin cell turnover. Furthermore, antibody exposure modulated the release of CXCL8 and CXCL10, reflecting the pattern changes seen in antibody-treated patients. The combination of a metastatic tumour environment with a miniaturized healthy organotypic human skin equivalent make this "safficacy" assay an ideal tool for evaluation of the therapeutic index of EGFR inhibitors and other promising oncology candidates.


Subject(s)
Antibodies, Monoclonal/adverse effects , Antineoplastic Agents, Immunological/adverse effects , ErbB Receptors/antagonists & inhibitors , Microfluidics , Neoplasms/etiology , Skin/drug effects , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Cell Culture Techniques , Cell Line, Tumor , Cetuximab/adverse effects , Cetuximab/pharmacology , Coculture Techniques , Equipment Design , Humans , Microfluidics/instrumentation , Microfluidics/methods , Neoplasms/pathology
3.
Future Sci OA ; 3(2): FSO185, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28670475

ABSTRACT

The development of drugs is a process obstructed with manifold security and efficacy concerns. Although animal models are still widely used to meet the diligence required, they are regarded as outdated tools with limited predictability. Novel microphysiological systems intend to create systemic models of human biology. Their ability to host 3D organoid constructs in a controlled microenvironment with mechanical and electrophysiological stimuli enables them to create and maintain homeostasis. These platforms are, thus, envisioned to be superior tools for testing and developing substances such as drugs, cosmetics and chemicals. We will present reasons why microphysiological systems are required for the emerging demands, highlight current technological and regulatory obstacles, and depict possible solutions from state-of-the-art platforms from major contributors.

4.
Article in English | MEDLINE | ID: mdl-28459049

ABSTRACT

Vascular network engineering is essential for nutrient delivery to tissue-engineered constructs and, consequently, their survival. In addition, the functionality of tissues also depends on tissue drainage and immune cell accessibility, which are the main functions of the lymphatic system. Engineering both the blood and lymphatic microvasculature would advance the survival and functionality of tissue-engineered constructs. The aim of this study was to isolate pure populations of lymphatic endothelial cells (LEC) and blood vascular endothelial cells (BEC) from human dermal microvascular endothelial cells and to study their network formation in our previously described coculture model with adipose-derived stromal cells (ASC) in fibrin scaffolds. We could follow the network development over a period of 4 weeks by fluorescently labeling the cells. We show that LEC and BEC form separate networks, which are morphologically distinguishable and sustainable over several weeks. In addition, lymphatic network development was dependent on vascular endothelial growth factor (VEGF)-C, resulting in denser networks with increasing VEGF-C concentration. Finally, we confirm the necessity of cell-cell contact between endothelial cells and ASC for the formation of both blood and lymphatic microvascular networks. This model represents a valuable platform for in vitro drug testing and for the future in vivo studies on lymphatic and blood microvascularization.

5.
J Biotechnol ; 216: 1-10, 2015 Dec 20.
Article in English | MEDLINE | ID: mdl-26435219

ABSTRACT

Current microfluidic chip-based tissue culture systems lack a capillary endothelial vessel system, which would enable perfusion with blood. We utilise spatial cell cultures to populate a perfused multi-organ-chip platform-a microfluidic device recently introduced for substance testing. Complete biological vascularization of such culture systems is vital to properly emulate physiological tissue behaviour. In this study, we incorporated a fibrin scaffold into the two-organ-chip design. Herein, adipose-derived stromal cells (ASCs) directed human umbilical vein endothelial cells (HUVECs) to organise into tube-like structures. The ASCs induced tube formation of HUVECs in static and dynamic conditions. The replacement of full medium enriched with growth factors and foetal calf serum with basal medium resulted in viable cells with similar gene expression profiles. We regard this as a prerequisite for studies with organ constructs that have a need for a different medium formulation. Furthermore, we here address stability issues of the fibrin gel and fibrin composition for optimal microvessel formation.


Subject(s)
Capillaries/physiology , Microfluidics/methods , Tissue Engineering/methods , Adipose Tissue/cytology , Cell Culture Techniques , Coculture Techniques , Culture Media/pharmacology , Fibrin/pharmacology , Gels/pharmacology , Green Fluorescent Proteins/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Perfusion , Polymerase Chain Reaction , Rheology/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Time Factors , Tissue Scaffolds/chemistry , Transcriptome/genetics
6.
Lab Chip ; 15(12): 2688-99, 2015 Jun 21.
Article in English | MEDLINE | ID: mdl-25996126

ABSTRACT

Systemic absorption and metabolism of drugs in the small intestine, metabolism by the liver as well as excretion by the kidney are key determinants of efficacy and safety for therapeutic candidates. However, these systemic responses of applied substances lack in most in vitro assays. In this study, a microphysiological system maintaining the functionality of four organs over 28 days in co-culture has been established at a minute but standardized microsystem scale. Preformed human intestine and skin models have been integrated into the four-organ-chip on standard cell culture inserts at a size 100,000-fold smaller than their human counterpart organs. A 3D-based spheroid, equivalent to ten liver lobules, mimics liver function. Finally, a barrier segregating the media flow through the organs from fluids excreted by the kidney has been generated by a polymeric membrane covered by a monolayer of human proximal tubule epithelial cells. A peristaltic on-chip micropump ensures pulsatile media flow interconnecting the four tissue culture compartments through microfluidic channels. A second microfluidic circuit ensures drainage of the fluid excreted through the kidney epithelial cell layer. This four-organ-chip system assures near to physiological fluid-to-tissue ratios. In-depth metabolic and gene analysis revealed the establishment of reproducible homeostasis among the co-cultures within two to four days, sustainable over at least 28 days independent of the individual human cell line or tissue donor background used for each organ equivalent. Lastly, 3D imaging two-photon microscopy visualised details of spatiotemporal segregation of the two microfluidic flows by proximal tubule epithelia. To our knowledge, this study is the first approach to establish a system for in vitro microfluidic ADME profiling and repeated dose systemic toxicity testing of drug candidates over 28 days.


Subject(s)
Coculture Techniques/instrumentation , Kidney/cytology , Liver/cytology , Microfluidic Analytical Techniques/instrumentation , Models, Biological , Skin/cytology , Cell Line , Cells, Cultured , Coculture Techniques/methods , Equipment Design , Gene Expression Profiling , Humans , Kidney/metabolism , Liver/metabolism , Skin/metabolism , Toxicity Tests
7.
Eur J Pharm Biopharm ; 95(Pt A): 77-87, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25857839

ABSTRACT

Systemic repeated dose safety assessment and systemic efficacy evaluation of substances are currently carried out on laboratory animals and in humans due to the lack of predictive alternatives. Relevant international regulations, such as OECD and ICH guidelines, demand long-term testing and oral, dermal, inhalation, and systemic exposure routes for such evaluations. So-called "human-on-a-chip" concepts are aiming to replace respective animals and humans in substance evaluation with miniaturized functional human organisms. The major technical hurdle toward success in this field is the life-like combination of human barrier organ models, such as intestine, lung or skin, with parenchymal organ equivalents, such as liver, at the smallest biologically acceptable scale. Here, we report on a reproducible homeostatic long-term co-culture of human liver equivalents with either a reconstructed human intestinal barrier model or a human skin biopsy applying a microphysiological system. We used a multi-organ chip (MOC) platform, which provides pulsatile fluid flow within physiological ranges at low media-to-tissue ratios. The MOC supports submerse cultivation of an intact intestinal barrier model and an air-liquid interface for the skin model during their co-culture with the liver equivalents respectively at (1)/100.000 the scale of their human counterparts in vivo. To increase the degree of organismal emulation, microfluidic channels of the liver-skin co-culture could be successfully covered with human endothelial cells, thus mimicking human vasculature, for the first time. Finally, exposure routes emulating oral and systemic administration in humans have been qualified by applying a repeated dose administration of a model substance - troglitazone - to the chip-based co-cultures.


Subject(s)
Chromans/administration & dosage , Ileum/metabolism , Lab-On-A-Chip Devices , Liver/metabolism , Skin/metabolism , Thiazolidinediones/administration & dosage , Cell Line, Tumor , Coculture Techniques/methods , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Humans , Ileum/drug effects , Liver/drug effects , Skin/drug effects , Troglitazone , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...