Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
2.
Cell Stem Cell ; 28(5): 846-862.e8, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33784497

ABSTRACT

The retinal pigment epithelium (RPE)-choriocapillaris (CC) complex in the eye is compromised in age-related macular degeneration (AMD) and related macular dystrophies (MDs), yet in vitro models of RPE-CC complex that enable investigation of AMD/MD pathophysiology are lacking. By incorporating iPSC-derived cells into a hydrogel-based extracellular matrix, we developed a 3D RPE-CC model that recapitulates key features of both healthy and AMD/MD eyes and provides modular control over RPE and CC layers. Using this 3D RPE-CC model, we demonstrated that both RPE- and mesenchyme-secreted factors are necessary for the formation of fenestrated CC-like vasculature. Our data show that choroidal neovascularization (CNV) and CC atrophy occur in the absence of endothelial cell dysfunction and are not necessarily secondary to drusen deposits underneath RPE cells, and CC atrophy and/or CNV can be initiated systemically by patient serum or locally by mutant RPE-secreted factors. Finally, we identify FGF2 and matrix metalloproteinases as potential therapeutic targets for AMD/MDs.


Subject(s)
Choroid Diseases , Induced Pluripotent Stem Cells , Macular Degeneration , Choroid , Humans , Retinal Pigment Epithelium
3.
PLoS Comput Biol ; 17(3): e1008778, 2021 03.
Article in English | MEDLINE | ID: mdl-33647016

ABSTRACT

Human pluripotent stem cells hold significant promise for regenerative medicine. However, long differentiation protocols and immature characteristics of stem cell-derived cell types remain challenges to the development of many therapeutic applications. In contrast to the slow differentiation of human stem cells in vitro that mirrors a nine-month gestation period, mouse stem cells develop according to a much faster three-week gestation timeline. Here, we tested if co-differentiation with mouse pluripotent stem cells could accelerate the differentiation speed of human embryonic stem cells. Following a six-week RNA-sequencing time course of neural differentiation, we identified 929 human genes that were upregulated earlier and 535 genes that exhibited earlier peaked expression profiles in chimeric cell cultures than in human cell cultures alone. Genes with accelerated upregulation were significantly enriched in Gene Ontology terms associated with neurogenesis, neuron differentiation and maturation, and synapse signaling. Moreover, chimeric mixed samples correlated with in utero human embryonic samples earlier than human cells alone, and acceleration was dose-dependent on human-mouse co-culture ratios. The altered gene expression patterns and developmental rates described in this report have implications for accelerating human stem cell differentiation and the use of interspecies chimeric embryos in developing human organs for transplantation.


Subject(s)
Chimerism , Human Embryonic Stem Cells , Neurogenesis , Pluripotent Stem Cells , Animals , Cells, Cultured , Computational Biology , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/physiology , Humans , Mice , Neurogenesis/genetics , Neurogenesis/physiology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Species Specificity , Transcriptome/genetics
4.
Proc Natl Acad Sci U S A ; 117(45): 27854-27861, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33106430

ABSTRACT

Understanding the mechanisms of nanoparticle interaction with cell membranes is essential for designing materials for applications such as bioimaging and drug delivery, as well as for assessing engineered nanomaterial safety. Much attention has focused on nanoparticles that bind strongly to biological membranes or induce membrane damage, leading to adverse impacts on cells. More subtle effects on membrane function mediated via changes in biophysical properties of the phospholipid bilayer have received little study. Here, we combine electrophysiology measurements, infrared spectroscopy, and molecular dynamics simulations to obtain insight into a mode of nanoparticle-mediated modulation of membrane protein function that was previously only hinted at in prior work. Electrophysiology measurements on gramicidin A (gA) ion channels embedded in planar suspended lipid bilayers demonstrate that anionic gold nanoparticles (AuNPs) reduce channel activity and extend channel lifetimes without disrupting membrane integrity, in a manner consistent with changes in membrane mechanical properties. Vibrational spectroscopy indicates that AuNP interaction with the bilayer does not perturb the conformation of membrane-embedded gA. Molecular dynamics simulations reinforce the experimental findings, showing that anionic AuNPs do not directly interact with embedded gA channels but perturb the local properties of lipid bilayers. Our results are most consistent with a mechanism in which anionic AuNPs disrupt ion channel function in an indirect manner by altering the mechanical properties of the surrounding bilayer. Alteration of membrane mechanical properties represents a potentially important mechanism by which nanoparticles induce biological effects, as the function of many embedded membrane proteins depends on phospholipid bilayer biophysical properties.


Subject(s)
Ion Channels/metabolism , Lipid Bilayers/chemistry , Metal Nanoparticles/chemistry , Anions/metabolism , Cell Membrane/metabolism , Cell Membrane/physiology , Gold/chemistry , Gold/pharmacology , Gramicidin/chemistry , Hydrophobic and Hydrophilic Interactions , Ion Channels/chemistry , Lipid Bilayers/metabolism , Membrane Proteins/metabolism , Membranes/metabolism , Molecular Conformation , Molecular Dynamics Simulation , Phosphatidylcholines/chemistry , Phospholipids/chemistry , Phospholipids/metabolism
5.
Stem Cell Reports ; 13(4): 669-683, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31522977

ABSTRACT

Here we elucidate the effect of Alzheimer disease (AD)-predisposing genetic backgrounds, APOE4, PSEN1ΔE9, and APPswe, on functionality of human microglia-like cells (iMGLs). We present a physiologically relevant high-yield protocol for producing iMGLs from induced pluripotent stem cells. Differentiation is directed with small molecules through primitive erythromyeloid progenitors to re-create microglial ontogeny from yolk sac. The iMGLs express microglial signature genes and respond to ADP with intracellular Ca2+ release distinguishing them from macrophages. Using 16 iPSC lines from healthy donors, AD patients and isogenic controls, we reveal that the APOE4 genotype has a profound impact on several aspects of microglial functionality, whereas PSEN1ΔE9 and APPswe mutations trigger minor alterations. The APOE4 genotype impairs phagocytosis, migration, and metabolic activity of iMGLs but exacerbates their cytokine secretion. This indicates that APOE4 iMGLs are fundamentally unable to mount normal microglial functionality in AD.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Apolipoprotein E4/genetics , Cell Differentiation/genetics , Induced Pluripotent Stem Cells/metabolism , Microglia/metabolism , Phenotype , Presenilin-1/genetics , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Apolipoprotein E4/metabolism , Calcium/metabolism , Cells, Cultured , Chemokines/metabolism , Cytokines/metabolism , Hematopoiesis , Humans , Induced Pluripotent Stem Cells/cytology , Inflammation Mediators/metabolism , Microglia/cytology , Mutation , Phagocytosis , Presenilin-1/metabolism , Proteolysis
6.
Adv Healthc Mater ; 8(2): e1801186, 2019 01.
Article in English | MEDLINE | ID: mdl-30565891

ABSTRACT

Vascularization is an important strategy to overcome diffusion limits and enable the formation of complex, physiologically relevant engineered tissues and organoids. Self-assembly is a technique to generate in vitro vascular networks, but engineering the necessary network morphology and function remains challenging. Here, autofluorescence multiphoton microscopy (aMPM), a label-free imaging technique, is used to quantitatively evaluate in vitro vascular network morphology. Vascular networks are generated using human embryonic stem cell-derived endothelial cells and primary human pericytes encapsulated in synthetic poly(ethylene glycol)-based hydrogels. Two custom-built bioreactors are used to generate distinct fluid flow patterns during vascular network formation: recirculating flow or continuous flow. aMPM is used to image these 3D vascular networks without the need for fixation, labels, or dyes. Image processing and analysis algorithms are developed to extract quantitative morphological parameters from these label-free images. It is observed with aMPM that both bioreactors promote formation of vascular networks with lower network anisotropy compared to static conditions, and the continuous flow bioreactor induces more branch points compared to static conditions. Importantly, these results agree with trends observed with immunocytochemistry. These studies demonstrate that aMPM allows label-free monitoring of vascular network morphology to streamline optimization of growth conditions and provide quality control of engineered tissues.


Subject(s)
Hydrogels/chemistry , Imaging, Three-Dimensional/methods , Microscopy, Fluorescence, Multiphoton/methods , Pericytes/cytology , Bioreactors , Blood Vessels , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Endothelial Cells/cytology , Endothelial Cells/physiology , Humans , Microscopy, Confocal , NAD/metabolism , NADP/metabolism , Neovascularization, Physiologic , Pericytes/physiology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Polyethylene Glycols/chemistry
7.
Article in English | MEDLINE | ID: mdl-29104816

ABSTRACT

The physiological relevance of Matrigel as a cell-culture substrate and in angiogenesis assays is often called into question. Here, we describe an array-based method for the identification of synthetic hydrogels that promote the formation of robust in vitro vascular networks for the detection of putative vascular disruptors, and that support human embryonic stem cell expansion and pluripotency. We identified hydrogel substrates that promoted endothelial-network formation by primary human umbilical vein endothelial cells and by endothelial cells derived from human induced pluripotent stem cells, and used the hydrogels with endothelial networks to identify angiogenesis inhibitors. The synthetic hydrogels show superior sensitivity and reproducibility over Matrigel when evaluating known inhibitors, as well as in a blinded screen of a subset of 38 chemicals, selected according to predicted vascular disruption potential, from the Toxicity ForeCaster library of the US Environmental Protection Agency. The identified synthetic hydrogels should be suitable alternatives to Matrigel for common cell-culture applications.

8.
Proc Natl Acad Sci U S A ; 114(30): E6072-E6078, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28696312

ABSTRACT

Here, we report the derivation of arterial endothelial cells from human pluripotent stem cells that exhibit arterial-specific functions in vitro and in vivo. We combine single-cell RNA sequencing of embryonic mouse endothelial cells with an EFNB2-tdTomato/EPHB4-EGFP dual reporter human embryonic stem cell line to identify factors that regulate arterial endothelial cell specification. The resulting xeno-free protocol produces cells with gene expression profiles, oxygen consumption rates, nitric oxide production levels, shear stress responses, and TNFα-induced leukocyte adhesion rates characteristic of arterial endothelial cells. Arterial endothelial cells were robustly generated from multiple human embryonic and induced pluripotent stem cell lines and have potential applications for both disease modeling and regenerative medicine.


Subject(s)
Arteries/cytology , Endothelial Cells/transplantation , Neovascularization, Physiologic , Pluripotent Stem Cells/physiology , Tissue Engineering/methods , Animals , CRISPR-Cas Systems , Cell Line , Endothelial Cells/cytology , Humans , Mice , Myocardial Infarction/therapy , Sequence Analysis, RNA
9.
Exp Biol Med (Maywood) ; 242(17): 1679-1689, 2017 11.
Article in English | MEDLINE | ID: mdl-28599598

ABSTRACT

The aim of the present study was to test sample reproducibility for model neural tissues formed on synthetic hydrogels. Human embryonic stem (ES) cell-derived precursor cells were cultured on synthetic poly(ethylene glycol) (PEG) hydrogels to promote differentiation and self-organization into model neural tissue constructs. Neural progenitor, vascular, and microglial precursor cells were combined on PEG hydrogels to mimic developmental timing, which produced multicomponent neural constructs with 3D neuronal and glial organization, organized vascular networks, and microglia with ramified morphologies. Spearman's rank correlation analysis of global gene expression profiles and a comparison of coefficient of variation for expressed genes demonstrated that replicate neural constructs were highly uniform to at least day 21 for samples from independent experiments. We also demonstrate that model neural tissues formed on PEG hydrogels using a simplified neural differentiation protocol correlated more strongly to in vivo brain development than samples cultured on tissue culture polystyrene surfaces alone. These results provide a proof-of-concept demonstration that 3D cellular models that mimic aspects of human brain development can be produced from human pluripotent stem cells with high sample uniformity between experiments by using standard culture techniques, cryopreserved cell stocks, and a synthetic extracellular matrix. Impact statement Pluripotent stem (PS) cells have been characterized by an inherent ability to self-organize into 3D "organoids" resembling stomach, intestine, liver, kidney, and brain tissues, offering a potentially powerful tool for modeling human development and disease. However, organoid formation must be quantitatively reproducible for applications such as drug and toxicity screening. Here, we report a strategy to produce uniform neural tissue constructs with reproducible global gene expression profiles for replicate samples from multiple experiments.


Subject(s)
Brain/cytology , Models, Biological , Neural Stem Cells/cytology , Pluripotent Stem Cells/cytology , Tissue Engineering/methods , Cell Culture Techniques/methods , Cell Differentiation/physiology , Cells, Cultured , Humans , Hydrogels , Polyethylene Glycols
10.
Stem Cell Reports ; 8(4): 907-918, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28343999

ABSTRACT

A defined protocol for efficiently deriving endothelial cells from human pluripotent stem cells was established and vascular morphogenesis was used as a model system to understand how synthetic hydrogels influence global biological function compared with common 2D and 3D culture platforms. RNA sequencing demonstrated that gene expression profiles were similar for endothelial cells and pericytes cocultured in polyethylene glycol (PEG) hydrogels or Matrigel, while monoculture comparisons identified distinct vascular signatures for each cell type. Endothelial cells cultured on tissue-culture polystyrene adopted a proliferative phenotype compared with cells cultured on or encapsulated in PEG hydrogels. The proliferative phenotype correlated to increased FAK-ERK activity, and knockdown or inhibition of ERK signaling reduced proliferation and expression for cell-cycle genes while increasing expression for "3D-like" vasculature development genes. Our results provide insight into the influence of 2D and 3D culture formats on global biological processes that regulate cell function.


Subject(s)
Endothelial Cells/cytology , Pericytes/cytology , Pluripotent Stem Cells/cytology , Tissue Engineering/methods , Transcriptome , Cell Culture Techniques/methods , Cell Cycle , Cell Differentiation , Cell Line , Cell Proliferation , Cells, Cultured , Collagen/chemistry , Drug Combinations , Endothelial Cells/metabolism , Humans , Hydrogels/chemistry , Laminin/chemistry , MAP Kinase Signaling System , Neovascularization, Physiologic , Pericytes/metabolism , Pluripotent Stem Cells/metabolism , Polyethylene Glycols/chemistry , Polystyrenes/chemistry , Proteoglycans/chemistry , Tissue Scaffolds/chemistry
11.
Dev Biol ; 423(2): 101-110, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28179190

ABSTRACT

How species-specific developmental timing is controlled is largely unknown. By following human embryonic stem (ES) cell and mouse epiblast stem (EpiS) cell differentiation through detailed RNA-sequencing time courses, here we show that pluripotent stem cells closely retain in vivo species-specific developmental timing in vitro. In identical neural differentiation conditions in vitro, gene expression profiles are accelerated in mouse EpiS cells compared to human ES cells with relative rates of differentiation closely reflecting the rates of progression through the Carnegie stages in utero. Dynamic Time Warping analysis identified 3389 genes that were regulated more quickly in mouse EpiS cells and identified none that were regulated more quickly in human ES cells. Interestingly, we also find that human ES cells differentiated in teratomas maintain the same rate of differentiation observed in vitro in spite of being grown in a mouse host. These results suggest the existence of a cell autonomous, species-specific developmental clock that pluripotent stem cells maintain even out of context of an intact embryo.


Subject(s)
Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Embryonic Stem Cells/cytology , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Mice, SCID , Neurons/cytology , Species Specificity , Teratoma/pathology , Time Factors
12.
Biomacromolecules ; 18(3): 709-718, 2017 03 13.
Article in English | MEDLINE | ID: mdl-28157290

ABSTRACT

As a result of improved relevance to in vivo physiology, in vitro studies are increasingly performed in diverse, three-dimensional (3D) biomaterials. However, material-cell type pairing effects on cytokine availability remain unclear. We cultured five cell types in agarose, alginate, collagen, Matrigel, or RGD-functionalized polyethylene glycol (PEG) hydrogels. We measured 21 cytokines in the conditioned media, and we identified differences in measured cytokine levels that were cell-type- or material-dependent. We further evaluated our data using principal component analysis. Interestingly, component one identified two classes of biomaterials with characteristic cytokine expression levels. Component two identified cell-type-dependent differences in cytokines related to the wound response. Although elements of soluble cytokine availability are shared despite parameter differences, material and cellular properties variably influenced cytokine levels, underlining the influence of biomaterial-cell type pairings on in vitro assay outcomes. Relationships between material properties, cellular responses, and cytokine availability in 3D in vitro models warrant further investigation.


Subject(s)
Biocompatible Materials/chemistry , Cytokines/biosynthesis , Cell Culture Techniques , Cell Line, Tumor , Culture Media/chemistry , Epithelial Cells/metabolism , Humans , Hydrogels/chemistry , Polyethylene Glycols/chemistry , Principal Component Analysis
13.
Acta Biomater ; 39: 12-24, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27181878

ABSTRACT

UNLABELLED: Activation of vascular endothelial cells (ECs) by growth factors initiates a cascade of events during angiogenesis in vivo consisting of EC tip cell selection, sprout formation, EC stalk cell proliferation, and ultimately vascular stabilization by support cells. Although EC functional assays can recapitulate one or more aspects of angiogenesis in vitro, they are often limited by undefined substrates and lack of dependence on key angiogenic signaling axes. Here, we designed and characterized a chemically-defined model of endothelial sprouting behavior in vitro using human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs). We rapidly encapsulated iPSC-ECs at high density in poly(ethylene glycol) (PEG) hydrogel spheres using thiol-ene chemistry and subsequently encapsulated cell-dense hydrogel spheres in a cell-free hydrogel layer. The hydrogel sprouting array supported pro-angiogenic phenotype of iPSC-ECs and supported growth factor-dependent proliferation and sprouting behavior. iPSC-ECs in the sprouting model responded appropriately to several reference pharmacological angiogenesis inhibitors of vascular endothelial growth factor, NF-κB, matrix metalloproteinase-2/9, protein kinase activity, and ß-tubulin, which confirms their functional role in endothelial sprouting. A blinded screen of 38 putative vascular disrupting compounds from the US Environmental Protection Agency's ToxCast library identified six compounds that inhibited iPSC-EC sprouting and five compounds that were overtly cytotoxic to iPSC-ECs at a single concentration. The chemically-defined iPSC-EC sprouting model (iSM) is thus amenable to enhanced-throughput screening of small molecular libraries for effects on angiogenic sprouting and iPSC-EC toxicity assessment. STATEMENT OF SIGNIFICANCE: Angiogenesis assays that are commonly used for drug screening and toxicity assessment applications typically utilize natural substrates like Matrigel(TM) that are difficult to spatially pattern, costly, ill-defined, and may exhibit lot-to-lot variability. Herein, we describe a novel angiogenic sprouting assay using chemically-defined, bioinert poly(ethylene glycol) hydrogels functionalized with biomimetic peptides to promote cell attachment and degradation in a reproducible format that may mitigate the need for natural substrates. The quantitative assay of angiogenic sprouting here enables precise control over the initial conditions and can be formulated into arrays for screening. The sprouting assay here was dependent on key angiogenic signaling axes in a screen of angiogenesis inhibitors and a blinded screen of putative vascular disrupting compounds from the US-EPA.


Subject(s)
Cell Differentiation , Endothelial Cells/metabolism , Hydrogels/chemistry , Induced Pluripotent Stem Cells/metabolism , Polyethylene Glycols/chemistry , Endothelial Cells/cytology , Humans , Hydrogels/chemical synthesis , Induced Pluripotent Stem Cells/cytology
14.
Acta Biomater ; 35: 32-41, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26945632

ABSTRACT

Here, we describe an in vitro strategy to model vascular morphogenesis where human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) are encapsulated in peptide-functionalized poly(ethylene glycol) (PEG) hydrogels, either on standard well plates or within a passive pumping polydimethylsiloxane (PDMS) tri-channel microfluidic device. PEG hydrogels permissive towards cellular remodeling were fabricated using thiol-ene photopolymerization to incorporate matrix metalloproteinase (MMP)-degradable crosslinks and CRGDS cell adhesion peptide. Time lapse microscopy, immunofluorescence imaging, and RNA sequencing (RNA-Seq) demonstrated that iPSC-ECs formed vascular networks through mechanisms that were consistent with in vivo vasculogenesis and angiogenesis when cultured in PEG hydrogels. Migrating iPSC-ECs condensed into clusters, elongated into tubules, and formed polygonal networks through sprouting. Genes upregulated for iPSC-ECs cultured in PEG hydrogels relative to control cells on tissue culture polystyrene (TCP) surfaces included adhesion, matrix remodeling, and Notch signaling pathway genes relevant to in vivo vascular development. Vascular networks with lumens were stable for at least 14days when iPSC-ECs were encapsulated in PEG hydrogels that were polymerized within the central channel of the microfluidic device. Therefore, iPSC-ECs cultured in peptide-functionalized PEG hydrogels offer a defined platform for investigating vascular morphogenesis in vitro using both standard and microfluidic formats. STATEMENT OF SIGNIFICANCE: Human induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) cultured in synthetic hydrogels self-assemble into capillary networks through mechanisms consistent with in vivo vascular morphogenesis.


Subject(s)
Blood Vessels/physiology , Endothelial Cells/cytology , Hydrogels/pharmacology , Induced Pluripotent Stem Cells/cytology , Tissue Engineering/methods , Blood Vessels/drug effects , Capillaries/drug effects , Capillaries/physiology , Cell Adhesion/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism
15.
Sci Rep ; 5: 14566, 2015 Sep 28.
Article in English | MEDLINE | ID: mdl-26411797

ABSTRACT

Botulinum neurotoxin (BoNT) detection provides a useful model for validating cell-based neurotoxicity screening approaches, as sensitivity is dependent on functionally competent neurons and clear quantitative endpoints are available for correlating results to approved animal testing protocols. Here, human induced pluripotent stem cell (iPSC)-derived neuronal cells were cultured on chemically-defined poly(ethylene glycol) (PEG) hydrogels formed by "thiol-ene" photopolymerization and tested as a cell-based neurotoxicity assay by determining sensitivity to active BoNT/A1. BoNT/A1 sensitivity was comparable to the approved in vivo mouse bioassay for human iPSC-derived neurons and neural stem cells (iPSC-NSCs) cultured on PEG hydrogels or treated tissue culture polystyrene (TCP) surfaces. However, maximum sensitivity for BoNT detection was achieved two weeks earlier for iPSC-NSCs that were differentiated and matured on PEG hydrogels compared to TCP. Therefore, chemically-defined synthetic hydrogels offer benefits over standard platforms when optimizing culture conditions for cell-based screening and achieve sensitivities comparable to an approved animal testing protocol.


Subject(s)
Botulinum Toxins/pharmacology , Cell Differentiation/drug effects , Drug Evaluation, Preclinical/methods , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Neurons/cytology , Neurons/drug effects , Animals , Botulinum Toxins, Type A/pharmacology , Cells, Cultured , Drug Discovery/methods , Humans , Hydrogels/chemistry , Mice , Polyethylene Glycols/chemistry
16.
Proc Natl Acad Sci U S A ; 112(40): 12516-21, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26392547

ABSTRACT

Human pluripotent stem cell-based in vitro models that reflect human physiology have the potential to reduce the number of drug failures in clinical trials and offer a cost-effective approach for assessing chemical safety. Here, human embryonic stem (ES) cell-derived neural progenitor cells, endothelial cells, mesenchymal stem cells, and microglia/macrophage precursors were combined on chemically defined polyethylene glycol hydrogels and cultured in serum-free medium to model cellular interactions within the developing brain. The precursors self-assembled into 3D neural constructs with diverse neuronal and glial populations, interconnected vascular networks, and ramified microglia. Replicate constructs were reproducible by RNA sequencing (RNA-Seq) and expressed neurogenesis, vasculature development, and microglia genes. Linear support vector machines were used to construct a predictive model from RNA-Seq data for 240 neural constructs treated with 34 toxic and 26 nontoxic chemicals. The predictive model was evaluated using two standard hold-out testing methods: a nearly unbiased leave-one-out cross-validation for the 60 training compounds and an unbiased blinded trial using a single hold-out set of 10 additional chemicals. The linear support vector produced an estimate for future data of 0.91 in the cross-validation experiment and correctly classified 9 of 10 chemicals in the blinded trial.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Neural Stem Cells/cytology , Pluripotent Stem Cells/cytology , Brain/cytology , Brain/growth & development , Brain/metabolism , Cell Communication/drug effects , Cell Communication/genetics , Cells, Cultured , Culture Media, Serum-Free/pharmacology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Gene Ontology , Humans , Hydrogels/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , Models, Biological , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neurogenesis/genetics , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Polyethylene Glycols/pharmacology , Support Vector Machine , Tissue Engineering/methods , Xenobiotics/classification , Xenobiotics/pharmacology
17.
Stem Cell Rev Rep ; 11(3): 511-25, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25190668

ABSTRACT

Here we describe a strategy to model blood vessel development using a well-defined induced pluripotent stem cell-derived endothelial cell type (iPSC-EC) cultured within engineered platforms that mimic the 3D microenvironment. The iPSC-ECs used here were first characterized by expression of endothelial markers and functional properties that included VEGF responsiveness, TNF-α-induced upregulation of cell adhesion molecules (MCAM/CD146; ICAM1/CD54), thrombin-dependent barrier function, shear stress-induced alignment, and 2D and 3D capillary-like network formation in Matrigel. The iPSC-ECs also formed 3D vascular networks in a variety of engineering contexts, yielded perfusable, interconnected lumen when co-cultured with primary human fibroblasts, and aligned with flow in microfluidics devices. iPSC-EC function during tubule network formation, barrier formation, and sprouting was consistent with that of primary ECs, and the results suggest a VEGF-independent mechanism for sprouting, which is relevant to therapeutic anti-angiogenesis strategies. Our combined results demonstrate the feasibility of using a well-defined, stable source of iPSC-ECs to model blood vessel formation within a variety of contexts using standard in vitro formats.


Subject(s)
Blood Vessels/growth & development , Cell Differentiation/genetics , Induced Pluripotent Stem Cells , Neovascularization, Physiologic/genetics , Blood Vessels/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Humans , Tumor Necrosis Factor-alpha/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis
18.
Biomater Sci ; 2(5): 745-756, 2014 May.
Article in English | MEDLINE | ID: mdl-25386339

ABSTRACT

Here, we aimed to investigate migration of a model tumor cell line (HT-1080 fibrosarcoma cells, HT-1080s) using synthetic biomaterials to systematically vary peptide ligand density and substrate stiffness. A range of substrate elastic moduli were investigated by using poly(ethylene glycol) (PEG) hydrogel arrays (0.34 - 17 kPa) and self-assembled monolayer (SAM) arrays (~0.1-1 GPa), while cell adhesion was tuned by varying the presentation of Arg-Gly-Asp (RGD)-containing peptides. HT-1080 motility was insensitive to cell adhesion ligand density on RGD-SAMs, as they migrated with similar speed and directionality for a wide range of RGD densities (0.2-5% mol fraction RGD). Similarly, HT-1080 migration speed was weakly dependent on adhesion on 0.34 kPa PEG surfaces. On 13 kPa surfaces, a sharp initial increase in cell speed was observed at low RGD concentration, with no further changes observed as RGD concentration was increased further. An increase in cell speed ~ two-fold for the 13 kPa relative to the 0.34 kPa PEG surface suggested an important role for substrate stiffness in mediating motility, which was confirmed for HT-1080s migrating on variable modulus PEG hydrogels with constant RGD concentration. Notably, despite ~ two-fold changes in cell speed over a wide range of moduli, HT-1080s adopted rounded morphologies on all surfaces investigated, which contrasted with well spread primary human mesenchymal stem cells (hMSCs). Taken together, our results demonstrate that HT-1080s are morphologically distinct from primary mesenchymal cells (hMSCs) and migrate with minimal dependence on cell adhesion for surfaces within a wide range of moduli, whereas motility is strongly influenced by matrix mechanical properties.

19.
Biomater Sci ; 2(7): 1024-1034, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-25105013

ABSTRACT

To address the challenges associated with defined control over matrix properties in 3D cell culture systems, we employed a peptide functionalized poly(ethylene glycol) (PEG) hydrogel matrix in which mechanical modulus and adhesive properties were tuned. An HT-1080 human fibrosarcoma cell line was chosen as a model for probing matrix influences on tumor cell migration using the PEG hydrogel platform. HT-1080 speed varied with a complex dependence on both matrix modulus and Cys-Arg-Gly-Asp-Ser (CRGDS) adhesion ligand concentration, with regimes in which motility increased, decreased, or was minimally altered being observed. We further investigated cell motility by forming matrix interfaces that mimic aspects of tissue boundaries that might be encountered during invasion by taking advantage of the spatial control of the thiol-ene photochemistry to form patterned regions of low and high cross-linking densities. HT-1080s in 100 Pa regions of patterned PEG hydrogels tended to reverse direction or aggregate at the interface when they encountered a 360 Pa boundary. In contrast, HT-1080s were apparently unimpeded when migrating from the stiff to the soft regions of PEG peptide hydrogels, which may indicate that cells are capable of "reverse durotaxis" within at least some matrix regimes. Taken together, our results identified matrix regimes in which HT-1080 motility was both positively and negatively influenced by cell adhesion or matrix modulus.

20.
Biosensors (Basel) ; 4(1): 28-44, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24791214

ABSTRACT

Micro-scale printing and patterning of living cells has multiple applications including tissue engineering, cell signaling assays, and the fabrication of cell-based biosensors. In this work, a molecular printing instrument, the Bioforce Nano eNabler, was modified to enable micron-scale -quill-pen based printing of mammalian cells in a 3D hyaluronan/gelatin based hydrogel. Specifically, photo-initiated -thiol-ene click chemistry was used to couple the thiol groups of thiolated hyaluronan/thiolated gelatin to the alkene groups of 4-arm polyethylene glycol (PEG)-norbornene molecules. Rapid photopolymerization enabled direct printing and controlled curing of living cells within the hydrogel matrix. The resulting hydrogels were biocompatible with human adipose-derived stem cells, NIH-3T3 cells, and mouse embryonic stem cells. The utility of this printing approach was also explored for cell-based biosensors. Micro-printed cells expressing a redox sensitive variant of the green fluorescent protein (roGFP-R12) showed a measurable fluorescent response to addition of oxidizing and then reducing agents. This work represents a novel approach to micron-scale cell patterning, and its potential for living, cell-based biosensors.

SELECTION OF CITATIONS
SEARCH DETAIL
...