Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Annu Rev Chem Biomol Eng ; 8: 525-547, 2017 06 07.
Article in English | MEDLINE | ID: mdl-28460183

ABSTRACT

High-throughput (HT) techniques built upon laboratory automation technology and coupled to statistical experimental design and parallel experimentation have enabled the acceleration of chemical process development across multiple industries. HT technologies are often applied to interrogate wide, often multidimensional experimental spaces to inform the design and optimization of any number of unit operations that chemical engineers use in process development. In this review, we outline the evolution of HT technology and provide a comprehensive overview of how HT automation is used throughout different industries, with a particular focus on chemical and pharmaceutical process development. In addition, we highlight the common strategies of how HT automation is incorporated into routine development activities to maximize its impact in various academic and industrial settings.


Subject(s)
High-Throughput Screening Assays/methods , Technology, Pharmaceutical/methods , Animals , Bioengineering/instrumentation , Bioengineering/methods , Drug Discovery/instrumentation , Drug Discovery/methods , Equipment Design , High-Throughput Screening Assays/instrumentation , Humans , Technology, Pharmaceutical/instrumentation
2.
Methods Mol Biol ; 1307: 319-27, 2016.
Article in English | MEDLINE | ID: mdl-24500899

ABSTRACT

Human pluripotent stem cells (hPSCs), under the right conditions, can be engineered to generate populations of any somatic cell type. Knowledge of what mechanisms govern differentiation towards a particular lineage is often quite useful for efficiently producing somatic cell populations from hPSCs. Here, we have outlined a strategy for deriving populations of simple epithelial cells, as well as more mature epidermal keratinocyte progenitors, from hPSCs by exploiting a mechanism previously shown to direct epithelial differentiation of hPSCs. Specifically, we describe how to direct epithelial differentiation of hPSCs using an Src family kinase inhibitor, SU6656, which has been shown to modulate ß-catenin translocation to the cell membrane and thus promote epithelial differentiation. The differentiation platform outlined here produces cells with the ability to terminally differentiate to epidermal keratinocytes in culture through a stable simple epithelial cell intermediate that can be expanded in culture for numerous (>10) passages.


Subject(s)
Cell Culture Techniques/methods , Epithelial Cells/cytology , Pluripotent Stem Cells/cytology , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , src-Family Kinases/antagonists & inhibitors , Cell Line , Epithelial Cells/drug effects , Flow Cytometry , Fluorescent Antibody Technique , Humans , Pluripotent Stem Cells/drug effects , src-Family Kinases/metabolism
3.
Biotechnol Bioeng ; 110(11): 3024-37, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23740478

ABSTRACT

Human pluripotent stem cells (hPSCs) have an unparalleled potential for tissue engineering applications including regenerative therapies and in vitro cell-based models for studying normal and diseased tissue morphogenesis, or drug and toxicological screens. While numerous hPSC differentiation methods have been developed to generate various somatic cell types, the potential of hPSC-based technologies is hinged on the ability to translate these established lab-scale differentiation systems to large-scale processes to meet the industrial and clinical demands for these somatic cell types. Here, we demonstrate a strategy for investigating the efficiency and scalability of hPSC differentiation platforms. Using two previously reported epithelial differentiation systems as models, we fit an ODE-based kinetic model to data representing dynamics of various cell subpopulations present in our culture. This fit was performed by estimating rate constants of each cell subpopulation's cell fate decisions (self-renewal, differentiation, death). Sensitivity analyses on predicted rate constants indicated which cell fate decisions had the greatest impact on overall epithelial cell yield in each differentiation process. In addition, we found that the final cell yield was limited by the self-renewal rate of either the progenitor state or the final differentiated state, depending on the differentiation protocol. Also, the relative impact of these cell fate decision rates was highly dependent on the maximum capacity of the cell culture system. Overall, we outline a novel approach for quantitative analysis of established laboratory-scale hPSC differentiation systems and this approach may ease development to produce large quantities of cells for tissue engineering applications.


Subject(s)
Cell Differentiation , Cell Proliferation , Pluripotent Stem Cells/physiology , Cell Culture Techniques/methods , Computational Biology , Humans , Models, Biological
4.
Tissue Eng Part A ; 19(19-20): 2233-41, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23672433

ABSTRACT

A satisfactory in vitro model of vocal fold mucosa does not exist, thus precluding a systematic, controlled study of vocal fold biology and biomechanics. We sought to create a valid, reproducible three-dimensional (3D) in vitro model of human origin of vocal fold mucosa of human origin. We hypothesized that coculture of human embryonic stem cell (hESC)-derived simple epithelial cells with primary vocal fold fibroblasts under appropriate conditions would elicit morphogenesis of progenitor cells into vocal fold epithelial-like cells and creation of a basement membrane. Using an in vitro prospective study design, hESCs were differentiated into cells that coexpressed the simple epithelial cell marker, keratin 18 (K18), and the transcription factor, p63. These simple epithelial cells were cocultured with primary vocal fold fibroblasts seeded in a collagen gel scaffold. The cells were cultured for 3 weeks in a keratinocyte medium at an air-liquid interface. After that time, the engineered mucosa demonstrated a stratified, squamous epithelium and a continuous basement membrane recapitulating the key morphologic and phenotypic characteristics of native vocal fold mucosa. hESC-derived epithelial cells exhibited positive staining for vocal fold stratified, squamous epithelial markers, keratin 13 (K13) and 14 (K14), as well as tight junctions, adherens junctions, gap junctions, and desmosomes. Despite the presence of components critical for epithelial structural integrity, the epithelium demonstrated greater permeability than native tissue indicating compromised functional integrity. While further work is warranted to improve functional barrier integrity, this study demonstrates that hESC-derived epithelial progenitor cells can be engineered to create a replicable 3D in vitro model of vocal fold mucosa featuring a multilayered, terminally differentiated epithelium.


Subject(s)
Embryonic Stem Cells/cytology , Epithelial Cells/cytology , Mucous Membrane/cytology , Cell Differentiation/physiology , Humans
5.
Tissue Eng Part C Methods ; 19(12): 949-60, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23560510

ABSTRACT

Human pluripotent stem cells (hPSCs) have an unparalleled potential to generate limitless quantities of any somatic cell type. However, current methods for producing populations of various somatic cell types from hPSCs are generally not standardized and typically incorporate undefined cell culture components often resulting in variable differentiation efficiencies and poor reproducibility. To address this, we have developed a defined approach for generating epithelial progenitor and epidermal cells from hPSCs. In doing so, we have identified an optimal starting cell density to maximize yield and maintain high purity of K18+/p63+ simple epithelial progenitors. In addition, we have shown that the use of synthetic, defined substrates in lieu of Matrigel and gelatin can successfully facilitate efficient epithelial differentiation, maintaining a high (>75%) purity of K14+/p63+ keratinocyte progenitor cells and at a two to threefold higher yield than a previously reported undefined differentiation method. These K14+/p63+ cells also exhibited a higher expansion potential compared to cells generated using an undefined differentiation protocol and were able to terminally differentiate and recapitulate an epidermal tissue architecture in vitro. In summary, we have demonstrated the production of populations of functional epithelial and epidermal cells from multiple hPSC lines using a new, completely defined differentiation strategy.


Subject(s)
Cell Differentiation/physiology , Endothelial Cells , Epithelial Cells , Pluripotent Stem Cells , Cell Line , Endothelial Cells/cytology , Endothelial Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
6.
PLoS One ; 8(3): e60016, 2013.
Article in English | MEDLINE | ID: mdl-23527294

ABSTRACT

Human pluripotent stem cells (hPSCs) provide unprecedented opportunities to study the earliest stages of human development in vitro and have the potential to provide unlimited new sources of cells for regenerative medicine. Although previous studies have reported cytokeratin 14+/p63+ keratinocyte generation from hPSCs, the multipotent progenitors of epithelial lineages have not been described and the developmental pathways regulating epithelial commitment remain largely unknown. Here we report membrane localization of ß-catenin during retinoic acid (RA)--induced epithelial differentiation. In addition hPSC treatment with the Src family kinase inhibitor SU6656 modulated ß-catenin localization and produced an enriched population of simple epithelial cells under defined culture conditions. SU6656 strongly upregulated expression of cytokeratins 18 and 8 (K18/K8), which are expressed in simple epithelial cells, while repressing expression of the pluripotency gene Oct4. This homogeneous population of K18+K8+Oct4- simple epithelial precursor cells can further differentiate into cells expressing keratinocyte or corneal-specific markers. These enriched hPSC-derived simple epithelial cells may provide a ready source for development and toxicology cell models and may serve as a progenitor for epithelial cell transplantation applications.


Subject(s)
Cell Differentiation/physiology , Epithelial Cells/cytology , Gene Expression Regulation/drug effects , Indoles/pharmacology , Pluripotent Stem Cells/cytology , Sulfonamides/pharmacology , src-Family Kinases/antagonists & inhibitors , Blotting, Western , Cadherins/metabolism , Cell Differentiation/drug effects , Cells, Cultured , DNA Primers/genetics , Flow Cytometry , Fluorescent Antibody Technique , Humans , Keratin-18/metabolism , Keratin-8/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/physiology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tretinoin/pharmacology , beta Catenin/metabolism
7.
Biotechnol Adv ; 31(7): 1002-19, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23510904

ABSTRACT

Human pluripotent stem cells (hPSCs), including both embryonic stem cells and induced pluripotent stem cells, offer a potential cell source for research, drug screening, and regenerative medicine applications due to their unique ability to self-renew or differentiate to any somatic cell type. Before the full potential of hPSCs can be realized, robust protocols must be developed to direct their fate. Cell fate decisions are based on components of the surrounding microenvironment, including soluble factors, substrate or extracellular matrix, cell-cell interactions, mechanical forces, and 2D or 3D architecture. Depending on their spatio-temporal context, these components can signal hPSCs to either self-renew or differentiate to cell types of the ectoderm, mesoderm, or endoderm. Researchers working at the interface of engineering and biology have identified various factors which can affect hPSC fate, often based on lessons from embryonic development, and they have utilized this information to design in vitro niches which can reproducibly direct hPSC fate. This review highlights culture systems that have been engineered to promote self-renewal or differentiation of hPSCs, with a focus on studies that have elucidated the contributions of specific microenvironmental cues in the context of those culture systems. We propose the use of microsystem technologies for high-throughput screening of spatial-temporal presentation of cues, as this has been demonstrated to be a powerful approach for differentiating hPSCs to desired cell types.


Subject(s)
Pluripotent Stem Cells , Stem Cell Niche , Tissue Engineering , Animals , Biotechnology , Cell Differentiation , Germ Layers , Humans , Mice
8.
Microsc Res Tech ; 73(9): 866-77, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20232467

ABSTRACT

Polycationic polymers have been used to condense therapeutic DNA into submicron particles, offering protection from shear-induced or enzymatic degradation. However, the spontaneous nature of this self-assembly process gives rise to the formation of multimolecular aggregates, resulting in significant polyplex heterogeneity. Additionally, cytotoxicity issues and serum instability have limited the in vivo efficacy of such systems. One way these issues can be addressed is through the inclusion of poly(ethylene glycol) (PEG). PEG has known steric effects that inhibit polyplex self-aggregation. A variety of PEGylated gene delivery formulations have been previously pursued in an effort to take advantage of this material's benefits. Because of such interest, our aim was to further explore the consequences of PEG inclusion on the structure and activity of gene delivery vehicle formulations. We explored the complexation of plasmid DNA with varying ratios of a PEGylated trilysine peptide (PEG-K(3)) and 25-kDa polyethylenimine (PEI). Atomic force and scanning electron microscopy were utilized to assess the polyplex size and shape and revealed that a critical threshold of PEG was necessary to promote the formation of homogeneous polyplexes. Flow cytometry and fluorescence microscopy analyses suggested that the presence of PEG inhibited transfection efficiency as a consequence of changes in intracellular trafficking and promoted an increased reliance on energy-independent mechanisms of cellular uptake. These studies provide new information on the role of PEG in delivery vehicle design and lay the foundation for future work aimed at elucidating the details of the intracellular transport of PEGylated polyplexes.


Subject(s)
DNA/chemistry , Gene Transfer Techniques/instrumentation , Polyethylene Glycols/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , DNA/genetics , Molecular Structure , Plasmids/chemistry , Plasmids/genetics , Transfection/instrumentation
SELECTION OF CITATIONS
SEARCH DETAIL
...