Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Int J Mol Sci ; 21(11)2020 May 28.
Article in English | MEDLINE | ID: mdl-32481600

ABSTRACT

Hepatocytes are key players in the innate immune response to liver pathogens but are challenging to study because of inaccessibility and a short half-life. Recent advances in in vitro differentiation of hepatocyte-like cells (HLCs) facilitated studies of hepatocyte-pathogen interactions. Here, we aimed to define the anti-viral innate immune potential of human HLCs with a focus on toll-like receptor (TLR)-expression and the presence of a metabolic switch. We analysed cytoplasmic pattern recognition receptor (PRR)- and endosomal TLR-expression and activity and adaptation of HLCs to an inflammatory environment. We found that transcript levels of retinoic acid inducible gene I (RIG-I), melanoma differentiation antigen 5 (MDA5), and TLR3 became downregulated during differentiation, indicating the acquisition of a more tolerogenic phenotype, as expected in healthy hepatocytes. HLCs responded to activation of RIG-I by producing interferons (IFNs) and IFN-stimulated genes. Despite low-level expression of TLR3, receptor expression was upregulated in an inflammatory environment. TLR3 signalling induced expression of proinflammatory cytokines at the gene level, indicating that several PRRs need to interact for successful innate immune activation. The inflammatory responsiveness of HLCs was accompanied by the downregulation of cytochrome P450 3A and 1A2 activity and decreased serum protein production, showing that the metabolic switch seen in primary hepatocytes during anti-viral responses is also present in HLCs.


Subject(s)
Hepatocytes/immunology , Immunity, Innate , Receptors, Pattern Recognition/metabolism , Receptors, Virus/immunology , Antiviral Agents/pharmacology , Cell Differentiation , Cytoplasm/metabolism , Embryonic Stem Cells/metabolism , Endosomes/metabolism , Hepatocytes/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Inflammation , Janus Kinases/metabolism , Ligands , Microscopy, Fluorescence , Receptors, Virus/metabolism , Signal Transduction/immunology , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
2.
Arch Toxicol ; 91(11): 3645, 2017 11.
Article in English | MEDLINE | ID: mdl-28980015

ABSTRACT

During manuscript proofing, the following sentence was not deleted in the section "Results" at the end of the paragraph: "Both male and female hepatocytes responded in a similar fashion to cotinine, whereas male hepatocyte function was more sensitive to chrysene, fluorene and naphthalene than female hepatocytes".

3.
Arch Toxicol ; 91(11): 3633-3643, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28510779

ABSTRACT

The liver is a dynamic organ which is both multifunctional and highly regenerative. A major role of the liver is to process both endo and xenobiotics. Cigarettes are an example of a legal and widely used drug which can cause major health problems for adults and constitute a particular risk to the foetus, if the mother smokes during pregnancy. Cigarette smoke contains a complex mixture of thousands of different xenobiotics, including nicotine and polycyclic aromatic hydrocarbons. These affect foetal development in a sex-specific manner, inducing sex-dependant molecular responses in different organs. To date, the effect of maternal smoking on the foetal liver has been studied in vitro using cell lines, primary tissue and animal models. While these models have proven to be useful, poor cell phenotype, tissue scarcity, batch-to-batch variation and species differences have led to difficulties in data extrapolation toward human development. Therefore, in this study we have employed hepatoblasts, derived from pluripotent stem cells, to model the effects of xenobiotics from cigarette smoke on human hepatocyte development. Highly pure hepatocyte populations (>90%) were produced in vitro and exposed to factors present in cigarette smoke. Analysis of ATP levels revealed that, independent of the sex, the majority of smoking derivatives tested individually did not deplete ATP levels below 50%. However, following exposure to a cocktail of smoking derivatives, ATP production fell below 50% in a sex-dependent manner. This was paralleled by a loss metabolic activity and secretory ability in both female and male hepatocytes. Interestingly, cell depletion was less pronounced in female hepatocytes, whereas caspase activation was ~twofold greater, indicating sex differences in cell death upon exposure to the smoking derivatives tested.


Subject(s)
Hepatocytes/cytology , Hepatocytes/drug effects , Pluripotent Stem Cells/drug effects , Smoking/adverse effects , Adenosine Triphosphate/metabolism , Cell Differentiation , Cells, Cultured , Cotinine/toxicity , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Female , Humans , Male , Pluripotent Stem Cells/cytology , Polycyclic Aromatic Hydrocarbons/toxicity , Sex Factors , alpha-Fetoproteins/metabolism
4.
J Hepatol ; 63(4): 934-42, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26022688

ABSTRACT

BACKGROUND & AIMS: The differentiation of stem cells to hepatocyte-like cells (HLC) offers the perspective of unlimited supply of human hepatocytes. However, the degree of differentiation of HLC remains controversial. To obtain an unbiased characterization, we performed a transcriptomic study with HLC derived from human embryonic and induced stem cells (ESC, hiPSC) from three different laboratories. METHODS: Genome-wide gene expression profiles of ESC and HLC were compared to freshly isolated and up to 14days cultivated primary human hepatocytes. Gene networks representing successful and failed hepatocyte differentiation, and the transcription factors involved in their regulation were identified. RESULTS: Gene regulatory network analysis demonstrated that HLC represent a mixed cell type with features of liver, intestine, fibroblast and stem cells. The "unwanted" intestinal features were associated with KLF5 and CDX2 transcriptional networks. Cluster analysis identified highly correlated groups of genes associated with mature liver functions (n=1057) and downregulated proliferation associated genes (n=1562) that approach levels of primary hepatocytes. However, three further clusters containing 447, 101, and 505 genes failed to reach levels of hepatocytes. Key TF of two of these clusters include SOX11, FOXQ1, and YBX3. The third unsuccessful cluster, controlled by HNF1, CAR, FXR, and PXR, strongly overlaps with genes repressed in cultivated hepatocytes compared to freshly isolated hepatocytes, suggesting that current in vitro conditions lack stimuli required to maintain gene expression in hepatocytes, which consequently also explains a corresponding deficiency of HLC. CONCLUSIONS: The present gene regulatory network approach identifies key transcription factors which require modulation to improve HLC differentiation.


Subject(s)
Embryonic Stem Cells/cytology , Hepatocytes/cytology , Induced Pluripotent Stem Cells/cytology , Liver/metabolism , RNA/genetics , Transcription Factors/genetics , Transcriptome , Cell Differentiation , Cells, Cultured , Embryonic Stem Cells/metabolism , Gene Regulatory Networks , Hepatocytes/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Liver/cytology , Transcription Factors/biosynthesis
7.
STAR Protoc ; 2(2): 100502, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33997816

ABSTRACT

This protocol describes how to produce human liver spheres from pluripotent stem cell-derived hepatic progenitors, endothelial cells, and hepatic stellate cells. Liver spheres form by self-assembly in microwells, generating up to 73 spheres per well of a 96-well plate. This process was automated using liquid handling and pipetting systems, permitting cost-effective scale-up and reducing sphere variability. In its current form, this system provides a powerful tool to generate human liver tissue for disease modeling and drug screening. For complete details on the use and execution of this protocol, please refer to Lucendo-Villarin et al. (2020) (https://doi.org/10.1088/1758-5090/abbdb2).


Subject(s)
Automation, Laboratory , Cell Culture Techniques , Cell Differentiation , Liver , Pluripotent Stem Cells , Spheroids, Cellular , Humans , Liver/cytology , Liver/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism
8.
STAR Protoc ; 2(2): 100493, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33997813

ABSTRACT

This protocol describes the production of hepatocyte-like cells (HLCs) from human pluripotent stem cells and how to induce hepatic steatosis, a condition characterized by intracellular lipid accumulation. Following differentiation to an HLC phenotype, intracellular lipid accumulation is induced with a steatosis induction cocktail, allowing the user to examine the cellular processes that underpin hepatic steatosis. Furthermore, the renewable nature of our system, on a defined genetic background, permits in-depth mechanistic analysis, which may facilitate therapeutic target identification in the future. For complete details on the use and execution of this protocol, please refer to Sinton et al. (2021).


Subject(s)
Cell Differentiation , Fatty Liver/metabolism , Hepatocytes/metabolism , Models, Biological , Pluripotent Stem Cells/metabolism , Fatty Liver/pathology , Hepatocytes/pathology , Humans , Pluripotent Stem Cells/pathology
9.
PLoS One ; 16(2): e0244070, 2021.
Article in English | MEDLINE | ID: mdl-33556073

ABSTRACT

A major bottleneck in the study of human liver physiology is the provision of stable liver tissue in sufficient quantity. As a result, current approaches to modelling human drug efficacy and toxicity rely heavily on immortalized human and animal cell lines. These models are informative but do possess significant drawbacks. To address the issues presented by those models, researchers have turned to pluripotent stem cells (PSCs). PSCs can be generated from defined genetic backgrounds, are scalable, and capable of differentiation to all the cell types found in the human body, representing an attractive source of somatic cells for in vitro and in vivo endeavours. Although unlimited numbers of somatic cell types can be generated in vitro, their maturation still remains problematic. In order to develop high fidelity PSC-derived liver tissue, it is necessary to better understand the cell microenvironment in vitro including key elements of liver physiology. In vivo a major driver of zonated liver function is the oxygen gradient that exists from periportal to pericentral regions. In this paper, we demonstrate how cell culture conditions for PSC-derived liver sphere systems can be optimised to recapitulate physiologically relevant oxygen gradients by using mathematical modelling. The mathematical model incorporates some often-understated features and mechanisms of traditional spheroid systems such as cell-specific oxygen uptake, media volume, spheroid size, and well dimensions that can lead to a spatially heterogeneous distribution of oxygen. This mathematical modelling approach allows for the calibration and identification of culture conditions required to generate physiologically realistic function within the microtissue through recapitulation of the in vivo microenvironment.


Subject(s)
Hepatocytes/metabolism , Liver/metabolism , Oxygen/metabolism , Pluripotent Stem Cells/metabolism , Hepatocytes/cytology , Humans , Liver/cytology , Models, Theoretical , Pluripotent Stem Cells/cytology
10.
Cells ; 11(1)2021 12 22.
Article in English | MEDLINE | ID: mdl-35011586

ABSTRACT

Regenerative medicine aims to replace damaged tissues by stimulating endogenous tissue repair or by transplanting autologous or allogeneic cells. Due to their capacity to produce unlimited numbers of cells of a given cell type, pluripotent stem cells, whether of embryonic origin or induced via the reprogramming of somatic cells, are of considerable therapeutic interest in the regenerative medicine field. However, regardless of the cell type, host immune responses present a barrier to success. The aim of this study was to investigate in vitro the immunological properties of human pluripotent stem cell (PSC)-derived hepatocyte-like cells (HLCs). These cells expressed MHC class I molecules while they lacked MHC class II and co-stimulatory molecules, such as CD80 and CD86. Following stimulation with IFN-γ, HLCs upregulated CD40, PD-L1 and MHC class I molecules. When co-cultured with allogeneic T cells, HLCs did not induce T cell proliferation; furthermore, when T cells were stimulated via αCD3/CD28 beads, HLCs inhibited their proliferation via IDO1 and tryptophan deprivation. These results demonstrate that PSC-derived HLCs possess immunoregulatory functions, at least in vitro.


Subject(s)
Hepatocytes/cytology , Induced Pluripotent Stem Cells/cytology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Tryptophan/deficiency , Allogeneic Cells/cytology , Cell Proliferation , Humans , Immunologic Factors/metabolism , Immunophenotyping , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology
11.
iScience ; 24(1): 101931, 2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33409477

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is currently the most prevalent form of liver disease worldwide. This term encompasses a spectrum of pathologies, from benign hepatic steatosis to non-alcoholic steatohepatitis, which have, to date, been challenging to model in the laboratory setting. Here, we present a human pluripotent stem cell (hPSC)-derived model of hepatic steatosis, which overcomes inherent challenges of current models and provides insights into the metabolic rewiring associated with steatosis. Following induction of macrovesicular steatosis in hepatocyte-like cells using lactate, pyruvate, and octanoate (LPO), respirometry and transcriptomic analyses revealed compromised electron transport chain activity. 13C isotopic tracing studies revealed enhanced TCA cycle anaplerosis, with concomitant development of a compensatory purine nucleotide cycle shunt leading to excess generation of fumarate. This model of hepatic steatosis is reproducible, scalable, and overcomes the challenges of studying mitochondrial metabolism in currently available models.

12.
iScience ; 24(6): 102552, 2021 Jun 25.
Article in English | MEDLINE | ID: mdl-34151225

ABSTRACT

Liver disease is a major cause of premature death. Oxidative stress in the liver represents a key disease driver. Compounds, such as dimethyl fumarate (DMF), can activate the antioxidant response and are used clinically to treat disease. In this study, we tested the protective properties of DMF before or after paracetamol exposure. Following DMF administration, Nrf2 nuclear translocation was tracked at the single-cell level and target gene transactivation confirmed. Next, the protective properties of DMF were examined following paracetamol exposure. Transcriptomic and biochemical analysis revealed that DMF rescue was underpinned by reduced Nf-kB and TGF-ß signaling and cell senescence. Following on from these studies, we employed a Zebrafish model to study paracetamol exposure in vivo. We combined a genetically modified Zebrafish model, expressing green fluorescent protein exclusively in the liver, with automated microscopy. Pre-treatment with DMF, prior to paracetamol exposure, led to reduced liver damage in Zebrafish demonstrating protective properties.

13.
J Vis Exp ; (159)2020 05 10.
Article in English | MEDLINE | ID: mdl-32449710

ABSTRACT

Liver disease is an escalating global health issue. While liver transplantation is an effective mode of therapy, patient mortality has increased due to shortages in donor organ availability. Organ scarcity also affects the routine supply of human hepatocytes for basic research and the clinic. Therefore, the development of renewable sources of human liver progenitor cells is desirable and is the goal of this study. To be able to effectively generate and deploy human liver progenitors on a large scale, a reproducible hepatic progenitor differentiation system was developed. This protocol aids experimental reproducibility between users in a range of cell cultureware formats and permits differentiations using both, human embryonic and induced pluripotent stem cell lines. These are important advantages over current differentiation systems that will enhance the basic research and may pave the way towards clinical product development.


Subject(s)
Cell Differentiation , Liver/cytology , Pluripotent Stem Cells/cytology , Cell Count , Cell Differentiation/drug effects , Cells, Cultured , Endoderm/cytology , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Image Processing, Computer-Assisted , Laminin/pharmacology , Pluripotent Stem Cells/drug effects , Reproducibility of Results
14.
Expert Opin Drug Metab Toxicol ; 15(1): 77-83, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30572740

ABSTRACT

Background: The liver plays a central role in human drug metabolism. To model drug metabolism, the major cell type of the liver, the hepatocyte, is commonly used. Hepatocytes can be derived from human and animal sources, including pluripotent stem cells. Cell-based models have shown promise in modeling human drug exposure. The assays used in those studies are normally 'snap-shot' in nature, and do not provide the complete picture of human drug exposure. Research design and methods: In this study, we employ stem cell-derived hepatocytes and impedance sensing to model human drug toxicity. This impedance-based stem cell assay reports hepatotoxicity in real time after treatment with compounds provided by industry. Results: Using electric cell-substrate impedance Sensing (ECIS), we were able to accurately measure drug toxicity post-drug exposure in real time and more quickly than gold standard biochemical assays. Conclusions: ECIS is robust and non-destructive methodology capable of monitoring human drug exposure with superior performance to current gold standard 'snapshot' assays. We believe that the methodology presented within this article could prove valuable in the quest to better predict off-target effects of drugs in humans.


Subject(s)
Chemical and Drug Induced Liver Injury/diagnosis , Electric Impedance , Hepatocytes/drug effects , Pluripotent Stem Cells/cytology , Cell Differentiation , Drug-Related Side Effects and Adverse Reactions/diagnosis , Hepatocytes/cytology , Humans , Time Factors , Toxicity Tests/methods
15.
iScience ; 16: 206-217, 2019 Jun 28.
Article in English | MEDLINE | ID: mdl-31185456

ABSTRACT

During mammalian development, liver differentiation is driven by signals that converge on multiple transcription factor networks. The hepatocyte nuclear factor signaling network is known to be essential for hepatocyte specification and maintenance. In this study, we have generated deletion and point mutants of hepatocyte nuclear factor-4alpha (HNF4α) to precisely evaluate the function of protein domains during hepatocyte specification from human pluripotent stem cells. We demonstrate that nuclear HNF4α is essential for hepatic progenitor specification, and the introduction of point mutations in HNF4α's Small Ubiquitin-like Modifier (SUMO) consensus motif leads to disrupted hepatocyte differentiation. Taking a multiomics approach, we identified key deficiencies in cell biology, which included dysfunctional metabolism, substrate adhesion, tricarboxylic acid cycle flux, microRNA transport, and mRNA processing. In summary, the combination of genome editing and multiomics analyses has provided valuable insight into the diverse functions of HNF4α during pluripotent stem cell entry into the hepatic lineage and during hepatocellular differentiation.

16.
Article in English | MEDLINE | ID: mdl-29786559

ABSTRACT

Stem cell technology in regenerative medicine has the potential to provide an unlimited supply of cells for drug testing, medical transplantation and academic research. In order to engineer a realistic tissue model using stem cells as an alternative to human tissue, it is essential to create artificial stem cell microenvironment or niches. Three-dimensional (3D) bioprinting is a promising tissue engineering field that offers new opportunities to precisely place stem cells within their niches layer-by-layer. This review covers bioprinting technologies, the current development of 'bio-inks' and how bioprinting has already been applied to stem-cell culture, as well as their applications for human regenerative medicine. The key considerations for bioink properties such as stiffness, stability and biodegradation, biocompatibility and printability are highlighted. Bioprinting of both adult and pluriopotent stem cells for various types of artificial tissues from liver to brain has been reviewed. 3D bioprinting of stem-cell derived tissues for human regenerative medicine is an exciting emerging area that represents opportunities for new research, industries and products as well as future challenges in clinical translation.This article is part of the theme issue 'Designer human tissue: coming to a lab near you'.


Subject(s)
Bioprinting/methods , Printing, Three-Dimensional , Regenerative Medicine/methods , Stem Cells/pathology , Tissue Engineering/methods , Bioprinting/instrumentation , Brain/physiology , Humans , Liver/physiology , Printing, Three-Dimensional/instrumentation
17.
J Vis Exp ; (137)2018 07 27.
Article in English | MEDLINE | ID: mdl-30102283

ABSTRACT

Human pluripotent stem cells represent a renewable source of human tissue. Our research is focused on generating human liver tissue from human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs). Current differentiation procedures generate human hepatocyte-like cells (HLCs) displaying a mixture of fetal and adult traits. To improve cell phenotype, we have fully defined our differentiation procedure and the cell niche, resulting in the generation of cell populations which display improved gene expression and function. While these studies mark progress, the ability to generate large quantities of multi well plates for screening has been limited by labour intensive procedures and batch to batch variation. To tackle this issue, we have developed a semi-automated platform to differentiate pluripotent stem cells into HLCs. Stem cell seeding and differentiation were performed using liquid handling and automatic pipetting systems in 96-well plate format. Following the differentiation, cell phenotype was analyzed using automated microscopy and a multi well luminometer.


Subject(s)
Hepatocytes/metabolism , Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation , Hepatocytes/cytology , Humans , Mice , Pluripotent Stem Cells/cytology
18.
J Vis Exp ; (121)2017 03 02.
Article in English | MEDLINE | ID: mdl-28287600

ABSTRACT

Human pluripotent stem cells (hPSCs) possess great value for biomedical research. hPSCs can be scaled and differentiated to all cell types found in the human body. The differentiation of hPSCs to human hepatocyte-like cells (HLCs) has been extensively studied, and efficient differentiation protocols have been established. The combination of extracellular matrix and biological stimuli, including growth factors, cytokines, and small molecules, have made it possible to generate HLCs that resemble primary human hepatocytes. However, the majority of procedures still employ undefined components, giving rise to batch-to-batch variation. This serves as a significant barrier to the application of the technology. To tackle this issue, we developed a defined system for hepatocyte differentiation using human recombinant laminins as extracellular matrices in combination with a serum-free differentiation process. Highly efficient hepatocyte specification was achieved, with demonstrated improvements in both HLC function and phenotype. Importantly, this system is easy to scale up using research and GMP-grade hPSC lines promising advances in cell-based modelling and therapies.


Subject(s)
Hepatocytes/cytology , Pluripotent Stem Cells/cytology , Cell Count , Cell Differentiation , Extracellular Matrix/metabolism , Humans
19.
Stem Cells Transl Med ; 5(6): 764-72, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27057006

ABSTRACT

UNLABELLED: The liver performs multiple functions within the human body. It is composed of numerous cell types, which play important roles in organ physiology. Our study centers on the major metabolic cell type of the liver, the hepatocyte, and its susceptibility to damage during drug overdose. In these studies, hepatocytes were generated from a renewable and genetically defined resource. In vitro-derived hepatocytes were extensively profiled and exposed to varying levels of paracetamol and plasma isolated from liver-failure patients, with a view to identifying noncoding microRNAs that could reduce drug- or serum-induced hepatotoxicity. We identified a novel anti-microRNA, which reduced paracetamol-induced hepatotoxicity and glutathione depletion. Additionally, we identified a prosurvival role for anti-microRNA-324 following exposure to plasma collected from liver failure patients. We believe that these studies represent an important advance for the field, demonstrating the power of stem cell-derived systems to model human biology "in a dish" and identify novel noncoding microRNAs, which could be translated to the clinic in the future. SIGNIFICANCE: The liver performs vital functions within the human body and is composed of numerous cell types. The major metabolic cell type of the liver, the hepatocyte, is susceptible to damage during drug overdose. In these studies, hepatocytes were generated from a renewable resource and exposed to varying levels of paracetamol, with a view to identifying interventions that could reduce or attenuate drug-induced liver toxicity. A novel noncoding RNA that reduced paracetamol-induced hepatocyte toxicity was identified. These findings may represent an important advance for the field.


Subject(s)
Hepatocytes/drug effects , MicroRNAs/therapeutic use , Necrosis/therapy , RNA, Untranslated/therapeutic use , Acetaminophen/toxicity , Adult , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/therapy , Female , Hepatocytes/pathology , Humans , Liver/drug effects , Liver/injuries , Liver/pathology , Male , MicroRNAs/genetics , Necrosis/genetics , Primary Cell Culture , RNA, Untranslated/genetics
20.
Methods Mol Biol ; 1250: 105-11, 2015.
Article in English | MEDLINE | ID: mdl-26272137

ABSTRACT

The increase in human liver disease worldwide is a major concern. At present, the only successful mode of treatment for failing liver function is organ transplantation. While highly successful, donor organs are a limited resource that cannot meet current demands. Therefore, alternative liver support strategies have been explored, including the use of the major and metabolic cell within the liver, the hepatocyte. While current approaches using human hepatocytes are very promising, donor material is still required and therefore suffers from similar limitations to whole organ transplantation. One alternative source of human hepatocytes being actively pursued in the field is pluripotent stem cells. Pluripotent stem cells are a scalable and renewable cell-based resource, which can be efficiently differentiated towards hepatocytes, including pluripotent stem cell lines that have been derived under good manufacturing practice conditions. Therefore, it is believed that this approach provides a promising model system for cell scale-up and differentiation. In the future, pluripotent stem cell-derived hepatocytes could be used in the clinic to support failing liver function if they should be deemed fit for purpose.


Subject(s)
Cell Culture Techniques , Cell Differentiation , Culture Media, Serum-Free , Hepatocytes/cytology , Human Embryonic Stem Cells/cytology , Cell Differentiation/drug effects , Hepatocytes/drug effects , Human Embryonic Stem Cells/drug effects , Humans , Intercellular Signaling Peptides and Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL