Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Stem Cell ; 31(7): 989-1002.e9, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38754430

ABSTRACT

Physiologically relevant human models that recapitulate the challenges of solid tumors and the tumor microenvironment (TME) are highly desired in the chimeric antigen receptor (CAR)-T cell field. We developed a breast cancer-on-chip model with an integrated endothelial barrier that enables the transmigration of perfused immune cells, their infiltration into the tumor, and concomitant monitoring of cytokine release during perfused culture over a period of up to 8 days. Here, we exemplified its use for investigating CAR-T cell efficacy and the ability to control the immune reaction with a pharmacological on/off switch. Additionally, we integrated primary breast cancer organoids to study patient-specific CAR-T cell efficacy. The modular architecture of our tumor-on-chip paves the way for studying the role of other cell types in the TME and thus provides the potential for broad application in bench-to-bedside translation as well as acceleration of the preclinical development of CAR-T cell products.


Subject(s)
Breast Neoplasms , Receptors, Chimeric Antigen , Humans , Breast Neoplasms/pathology , Breast Neoplasms/immunology , Female , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Immunotherapy, Adoptive/methods , Tumor Microenvironment , T-Lymphocytes/immunology , Lab-On-A-Chip Devices , Cell Line, Tumor , Organoids/pathology
2.
Adv Sci (Weinh) ; 9(18): e2104451, 2022 06.
Article in English | MEDLINE | ID: mdl-35466539

ABSTRACT

Obesity and associated diseases, such as diabetes, have reached epidemic proportions globally. In this era of "diabesity", white adipose tissue (WAT) has become a target of high interest for therapeutic strategies. To gain insights into mechanisms of adipose (patho-)physiology, researchers traditionally relied on animal models. Leveraging Organ-on-Chip technology, a microphysiological in vitro model of human WAT is introduced: a tailored microfluidic platform featuring vasculature-like perfusion that integrates 3D tissues comprising all major WAT-associated cellular components (mature adipocytes, organotypic endothelial barriers, stromovascular cells including adipose tissue macrophages) in an autologous manner and recapitulates pivotal WAT functions, such as energy storage and mobilization as well as endocrine and immunomodulatory activities. A precisely controllable bottom-up approach enables the generation of a multitude of replicates per donor circumventing inter-donor variability issues and paving the way for personalized medicine. Moreover, it allows to adjust the model's degree of complexity via a flexible mix-and-match approach. This WAT-on-Chip system constitutes the first human-based, autologous, and immunocompetent in vitro adipose tissue model that recapitulates almost full tissue heterogeneity and can become a powerful tool for human-relevant research in the field of metabolism and its associated diseases as well as for compound testing and personalized- and precision medicine applications.


Subject(s)
Adipose Tissue, White , Adipose Tissue , Adipocytes, White/metabolism , Adipose Tissue/metabolism , Adipose Tissue, White/metabolism , Animals , Humans , Microfluidics , Obesity/metabolism
3.
Methods Mol Biol ; 2373: 297-313, 2022.
Article in English | MEDLINE | ID: mdl-34520020

ABSTRACT

Research on white adipose tissue (WAT), which constitutes one-fifth to one-half of the total body mass of a human's body, has gained more and more interest and attention in the era of "diabesity". In vitro research on mature human WAT is hampered by many challenges and, hence, a majority of WAT-related research is conducted using animal models as well as clinical observations and genome-wide association studies (GWAS), both featuring limitations in terms of translatability and potential for experimental interventions, respectively. Here, we describe methods to isolate primary mature human adipocytes from biopsies and to fabricate tailored organ-on-chip platforms for the long-term culture of WAT constructs.


Subject(s)
Adipocytes , Adipose Tissue , Adipose Tissue, White , Animals , Genome-Wide Association Study , Humans , Technology
4.
Stem Cell Reports ; 16(9): 2242-2256, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525384

ABSTRACT

Gene therapies using adeno-associated viruses (AAVs) are among the most promising strategies to treat or even cure hereditary and acquired retinal diseases. However, the development of new efficient AAV vectors is slow and costly, largely because of the lack of suitable non-clinical models. By faithfully recreating structure and function of human tissues, human induced pluripotent stem cell (iPSC)-derived retinal organoids could become an essential part of the test cascade addressing translational aspects. Organ-on-chip (OoC) technology further provides the capability to recapitulate microphysiological tissue environments as well as a precise control over structural and temporal parameters. By employing our recently developed retina on chip that merges organoid and OoC technology, we analyzed the efficacy, kinetics, and cell tropism of seven first- and second-generation AAV vectors. The presented data demonstrate the potential of iPSC-based OoC models as the next generation of screening platforms for future gene therapeutic studies.


Subject(s)
Dependovirus/genetics , Genetic Vectors/genetics , Induced Pluripotent Stem Cells/cytology , Lab-On-A-Chip Devices , Organoids/metabolism , Retina/metabolism , Transduction, Genetic , Biomarkers , Cell Culture Techniques , Cell Culture Techniques, Three Dimensional , Cell Differentiation , Fluorescent Antibody Technique , Gene Expression , Genes, Reporter , Genetic Therapy , Humans , Organoids/cytology , Retina/cytology , Transgenes
5.
Sci Rep ; 10(1): 6666, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32313039

ABSTRACT

Obesity and its numerous adverse health consequences have taken on global, pandemic proportions. White adipose tissue (WAT) - a key contributor in many metabolic diseases - contributes about one fourth of a healthy human's body mass. Despite its significance, many WAT-related pathophysiogical mechanisms in humans are still not understood, largely due to the reliance on non-human animal models. In recent years, Organ-on-a-chip (OoC) platforms have developed into promising alternatives for animal models; these systems integrate engineered human tissues into physiological microenvironment supplied by a vasculature-like microfluidic perfusion. Here, we report the development of a novel OoC that integrates functional mature human white adipocytes. The WAT-on-a-chip is a multilayer device that features tissue chambers tailored specifically for the maintenance of 3D tissues based on human primary adipocytes, with supporting nourishment provided through perfused media channels. The platform's capability to maintain long-term viability and functionality of white adipocytes was confirmed by real-time monitoring of fatty acid uptake, by quantification of metabolite release into the effluent media as well as by an intact responsiveness to a therapeutic compound. The novel system provides a promising tool for wide-ranging applications in mechanistic research of WAT-related biology, in studying of pathophysiological mechanisms in obesity and diabetes, and in R&D of pharmaceutical industry.


Subject(s)
Adipocytes, White/drug effects , Adipose Tissue, White/drug effects , Culture Media/pharmacology , Fatty Acids/metabolism , Lab-On-A-Chip Devices , Adipocytes, White/cytology , Adipocytes, White/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Biological Transport/drug effects , Culture Media/chemistry , Dimethylpolysiloxanes/chemistry , Humans , Isoproterenol/pharmacology , L-Lactate Dehydrogenase/metabolism , Microtechnology/methods , Models, Biological , Obesity/drug therapy , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Primary Cell Culture
SELECTION OF CITATIONS
SEARCH DETAIL
...