Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Lab Chip ; 24(7): 2080-2093, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38441218

ABSTRACT

Pancreatic in vitro research is of major importance to advance mechanistic understanding and development of treatment options for diseases such as diabetes mellitus. We present a thermoplastic-based microphysiological system aiming to model the complex microphysiological structure and function of the endocrine pancreas with concurrent real-time read-out capabilities. The specifically tailored platform enables self-guided trapping of single islets at defined locations: ß-cells are assembled to pseudo-islets and injected into the tissue chamber using hydrostatic pressure-driven flow. The pseudo-islets can further be embedded in an ECM-like hydrogel mimicking the native microenvironment of pancreatic islets in vivo. Non-invasive real-time monitoring of the oxygen levels on-chip is realized by the integration of luminescence-based optical sensors to the platform. To monitor insulin secretion kinetics in response to glucose stimulation in a time-resolved manner, an automated cycling of different glucose conditions is implemented. The model's response to glucose stimulation can be monitored via offline analysis of insulin secretion and via specific changes in oxygen consumption due to higher metabolic activity of pseudo-islets at high glucose levels. To demonstrate applicability for drug testing, the effects of antidiabetic medications are assessed and changes in dynamic insulin secretion are observed in line with the respective mechanism of action. Finally, by integrating human pancreatic islet microtissues, we highlight the flexibility of the platform and demonstrate the preservation of long-term functionality of human endocrine pancreatic tissue.


Subject(s)
Insulin , Islets of Langerhans , Humans , Insulin/metabolism , Pancreas , Glucose/analysis , Insulin Secretion
2.
ACS Biomater Sci Eng ; 8(11): 4643-4647, 2022 11 14.
Article in English | MEDLINE | ID: mdl-35760397

ABSTRACT

Over the past decade, organ-on-chip research has been one of the most prolific areas of the entire field of tissue engineering. The development of organ-on-chip models requires an integrated interdisciplinary approach merging technologies and concepts from several different disciplines, including microfabrication, microfluidics, biomaterials, stem cell science, pharma-/toxicology, and medicine. In this perspective, we follow the journey of an organ-on-chip through its many different stages, from (i) the initial idea/specific scientific question to (ii) the design/concept phase, (iii) the engineering (fabrication and materials, sensor/actuator integration) and (iv) biology considerations (cell sources, biomaterials/scaffold), (v) the cell injection and tissue assembly process, (vi) the assay development, and (vii) the functional validation, all the way to (viii) the final applications. By summarizing some of the key learnings and findings from a developer's perspective and identifying suitable introductory reviews, this perspective strives to provide a conceptual, stepwise guide for the holistic development of an organ-on-chip model.


Subject(s)
Lab-On-A-Chip Devices , Microfluidics , Tissue Engineering , Biocompatible Materials , Stem Cells
3.
Commun Biol ; 5(1): 52, 2022 01 13.
Article in English | MEDLINE | ID: mdl-35027657

ABSTRACT

Disorders of the eye leading to visual impairment are a major issue that affects millions of people. On the other side ocular toxicities were described for e.g. molecularly targeted therapies in oncology and may hamper their development. Current ocular model systems feature a number of limitations affecting human-relevance and availability. To find new options for pharmacological treatment and assess mechanisms of toxicity, hence, novel complex model systems that are human-relevant and readily available are urgently required. Here, we report the development of a human immunocompetent Choroid-on-Chip (CoC), a human cell-based in vitro model of the choroid layer of the eye integrating melanocytes and microvascular endothelial cells, covered by a layer of retinal pigmented epithelial cells. Immunocompetence is achieved by perfusion of peripheral immune cells. We demonstrate controlled immune cell recruitment into the stromal compartments through a vascular monolayer and in vivo-like cytokine release profiles. To investigate applicability for both efficacy testing of immunosuppressive compounds as well as safety profiling of immunoactivating antibodies, we exposed the CoCs to cyclosporine and tested CD3 bispecific antibodies.


Subject(s)
Biological Products/pharmacology , Choroid/drug effects , Endothelial Cells/drug effects , Microchip Analytical Procedures , Antibodies, Bispecific/drug effects , Antibodies, Bispecific/metabolism , Humans , Melanocytes/drug effects , Melanocytes/metabolism
4.
Cell Rep ; 36(3): 109393, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34289365

ABSTRACT

Alcohol-associated liver disease (ALD) is a global health issue and leads to progressive liver injury, comorbidities, and increased mortality. Human-relevant preclinical models of ALD are urgently needed. Here, we leverage a triculture human Liver-Chip with biomimetic hepatic sinusoids and bile canaliculi to model ALD employing human-relevant blood alcohol concentrations (BACs) and multimodal profiling of clinically relevant endpoints. Our Liver-Chip recapitulates established ALD markers in response to 48 h of exposure to ethanol, including lipid accumulation and oxidative stress, in a concentration-dependent manner and supports the study of secondary insults, such as high blood endotoxin levels. We show that remodeling of the bile canalicular network can provide an in vitro quantitative readout of alcoholic liver toxicity. In summary, we report the development of a human ALD Liver-Chip as a powerful platform for modeling alcohol-induced liver injury with the potential for direct translation to clinical research and evaluation of patient-specific responses.


Subject(s)
Lab-On-A-Chip Devices , Liver Diseases, Alcoholic/pathology , Liver/pathology , Models, Biological , Ethanol , Gene Expression Profiling , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver Diseases, Alcoholic/genetics , Polyploidy
5.
Micromachines (Basel) ; 12(5)2021 May 18.
Article in English | MEDLINE | ID: mdl-34070209

ABSTRACT

The emergence and spread of microfluidics over the last decades relied almost exclusively on the elastomer polydimethylsiloxane (PDMS). The main reason for the success of PDMS in the field of microfluidic research is its suitability for rapid prototyping and simple bonding methods. PDMS allows for precise microstructuring by replica molding and bonding to different substrates through various established strategies. However, large-scale production and commercialization efforts are hindered by the low scalability of PDMS-based chip fabrication and high material costs. Furthermore, fundamental limitations of PDMS, such as small molecule absorption and high water evaporation, have resulted in a shift toward PDMS-free systems. Thermoplastic elastomers (TPE) are a promising alternative, combining properties from both thermoplastic materials and elastomers. Here, we present a rapid and scalable fabrication method for microfluidic systems based on a polycarbonate (PC) and TPE hybrid material. Microstructured PC/TPE-hybrid modules are generated by hot embossing precise features into the TPE while simultaneously fusing the flexible TPE to a rigid thermoplastic layer through thermal fusion bonding. Compared to TPE alone, the resulting, more rigid composite material improves device handling while maintaining the key advantages of TPE. In a fast and simple process, the PC/TPE-hybrid can be bonded to several types of thermoplastics as well as glass substrates. The resulting bond strength withstands at least 7.5 bar of applied pressure, even after seven days of exposure to a high-temperature and humid environment, which makes the PC/TPE-hybrid suitable for most microfluidic applications. Furthermore, we demonstrate that the PC/TPE-hybrid features low absorption of small molecules while being biocompatible, making it a suitable material for microfluidic biotechnological applications.

6.
Matrix Biol ; 85-86: 205-220, 2020 01.
Article in English | MEDLINE | ID: mdl-31238092

ABSTRACT

The increasing prevalence of diabetes, its heterogeneity, and the limited number of treatment options drive the need for physiologically relevant assay platforms with human genetic background that have the potential to improve mechanistic understanding and e\xpedite diabetes-related research and treatment. In this study, we developed an endocrine pancreas-on-a-chip model based on a tailored microfluidic platform, which enables self-guided trapping of single human pseudo-islets. Continuous, low-shear perfusion provides a physiologically relevant microenvironment especially important for modeling and monitoring of the endocrine function as well as sufficient supply with nutrients and oxygen. Human pseudo-islets, generated from the conditionally immortalized EndoC-ßH3 cell line, were successfully injected by hydrostatic pressure-driven flow without altered viability. To track insulin secretion kinetics in response to glucose stimulation in a time-resolved manner, dynamic sampling of the supernatant as well as non-invasive real-time monitoring using Raman microspectroscopy was established on-chip. Dynamic sampling indicated a biphasic glucose-stimulated insulin response. Raman microspectroscopy allowed to trace glucose responsiveness in situ and to visualize different molecular structures such as lipids, mitochondria and nuclei. In-depth spectral analyses demonstrated a glucose stimulation-dependent, increased mitochondrial activity, and a switch in lipid composition of insulin secreting vesicles, supporting the high performance of our pancreas-on-a-chip model.


Subject(s)
Glucose/pharmacology , Insulin/metabolism , Islets of Langerhans/cytology , Cell Line , Cellular Microenvironment , Humans , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Lab-On-A-Chip Devices , Organ Culture Techniques , Spectrum Analysis, Raman
SELECTION OF CITATIONS
SEARCH DETAIL
...