Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biofabrication ; 16(1)2023 12 11.
Article in English | MEDLINE | ID: mdl-37972398

ABSTRACT

Embryoid bodies (EBs) and self-organizing organoids derived from human pluripotent stem cells (hPSCs) recapitulate tissue development in a dish and hold great promise for disease modeling and drug development. However, current protocols are hampered by cellular stress and apoptosis during cell aggregation, resulting in variability and impaired cell differentiation. Here, we demonstrate that EBs and various organoid models (e.g., brain, gut, kidney) can be optimized by using the small molecule cocktail named CEPT (chroman 1, emricasan, polyamines, trans-ISRIB), a polypharmacological approach that ensures cytoprotection and cell survival. Application of CEPT for just 24 h during cell aggregation has long-lasting consequences affecting morphogenesis, gene expression, cellular differentiation, and organoid function. Various qualification methods confirmed that CEPT treatment enhanced experimental reproducibility and consistently improved EB and organoid fitness as compared to the widely used ROCK inhibitor Y-27632. Collectively, we discovered that stress-free cell aggregation and superior cell survival in the presence of CEPT are critical quality control determinants that establish a robust foundation for bioengineering complex tissue and organ models.


Subject(s)
Embryoid Bodies , Pluripotent Stem Cells , Humans , Embryoid Bodies/metabolism , Reproducibility of Results , Organoids , Cell Differentiation
2.
Stem Cell Reports ; 18(8): 1701-1720, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37451260

ABSTRACT

Human gliogenesis remains poorly understood, and derivation of astrocytes from human pluripotent stem cells (hPSCs) is inefficient and cumbersome. Here, we report controlled glial differentiation from hPSCs that bypasses neurogenesis, which otherwise precedes astrogliogenesis during brain development and in vitro differentiation. hPSCs were first differentiated into radial glial cells (RGCs) resembling resident RGCs of the fetal telencephalon, and modulation of specific cell signaling pathways resulted in direct and stepwise induction of key astroglial markers (NFIA, NFIB, SOX9, CD44, S100B, glial fibrillary acidic protein [GFAP]). Transcriptomic and genome-wide epigenetic mapping and single-cell analysis confirmed RGC-to-astrocyte differentiation, obviating neurogenesis and the gliogenic switch. Detailed molecular and cellular characterization experiments uncovered new mechanisms and markers for human RGCs and astrocytes. In summary, establishment of a glia-exclusive neural lineage progression model serves as a unique serum-free platform of manufacturing large numbers of RGCs and astrocytes for neuroscience, disease modeling (e.g., Alexander disease), and regenerative medicine.


Subject(s)
Astrocytes , Pluripotent Stem Cells , Humans , Astrocytes/metabolism , Ependymoglial Cells/metabolism , Pluripotent Stem Cells/metabolism , Neurogenesis , Cell Differentiation , Glial Fibrillary Acidic Protein/metabolism
3.
Cell Rep Methods ; 3(3): 100420, 2023 03 27.
Article in English | MEDLINE | ID: mdl-37056373

ABSTRACT

SEQUIN is a web-based application (app) that allows fast and intuitive analysis of RNA sequencing data derived for model organisms, tissues, and single cells. Integrated app functions enable uploading datasets, quality control, gene set enrichment, data visualization, and differential gene expression analysis. We also developed the iPSC Profiler, a practical gene module scoring tool that helps measure and compare pluripotent and differentiated cell types. Benchmarking to other commercial and non-commercial products underscored several advantages of SEQUIN. Freely available to the public, SEQUIN empowers scientists using interdisciplinary methods to investigate and present transcriptome data firsthand with state-of-the-art statistical methods. Hence, SEQUIN helps democratize and increase the throughput of interrogating biological questions using next-generation sequencing data with single-cell resolution.


Subject(s)
Software , Transcriptome , RNA-Seq , Transcriptome/genetics , Sequence Analysis, RNA/methods , Gene Regulatory Networks
4.
Stem Cell Reports ; 18(4): 1030-1047, 2023 04 11.
Article in English | MEDLINE | ID: mdl-37044067

ABSTRACT

Development of new non-addictive analgesics requires advanced strategies to differentiate human pluripotent stem cells (hPSCs) into relevant cell types. Following principles of developmental biology and translational applicability, here we developed an efficient stepwise differentiation method for peptidergic and non-peptidergic nociceptors. By modulating specific cell signaling pathways, hPSCs were first converted into SOX10+ neural crest, followed by differentiation into sensory neurons. Detailed characterization, including ultrastructural analysis, confirmed that the hPSC-derived nociceptors displayed cellular and molecular features comparable to native dorsal root ganglion (DRG) neurons, and expressed high-threshold primary sensory neuron markers, transcription factors, neuropeptides, and over 150 ion channels and receptors relevant for pain research and axonal growth/regeneration studies (e.g., TRPV1, NAV1.7, NAV1.8, TAC1, CALCA, GAP43, DPYSL2, NMNAT2). Moreover, after confirming robust functional activities and differential response to noxious stimuli and specific drugs, a robotic cell culture system was employed to produce large quantities of human sensory neurons, which can be used to develop nociceptor-selective analgesics.


Subject(s)
Neurons , Pluripotent Stem Cells , Humans , Neurons/metabolism , Nociceptors , Cell Differentiation , Signal Transduction , Ganglia, Spinal/metabolism , Sensory Receptor Cells
5.
Nat Protoc ; 18(1): 58-80, 2023 01.
Article in English | MEDLINE | ID: mdl-36261632

ABSTRACT

Human pluripotent stem cells (hPSCs) are inherently sensitive cells. Single-cell dissociation and the establishment of clonal cell lines have been long-standing challenges. This inefficiency of cell cloning represents a major obstacle for the standardization and streamlining of gene editing in induced pluripotent stem cells for basic and translational research. Here we describe a chemically defined protocol for robust single-cell cloning using microfluidics-based cell sorting in combination with the CEPT small-molecule cocktail. This advanced strategy promotes the viability and cell fitness of self-renewing stem cells. The use of low-pressure microfluidic cell dispensing ensures gentle and rapid dispensing of single cells into 96- and 384-well plates, while the fast-acting CEPT cocktail minimizes cellular stress and maintains cell structure and function immediately after cell dissociation. The protocol also facilitates clone picking and produces genetically stable clonal cell lines from hPSCs in a safe and cost-efficient fashion. Depending on the proliferation rate of the clone derived from a single cell, this protocol can be completed in 7-14 d and requires experience with aseptic cell culture techniques. Altogether, the relative ease, scalability and robustness of this workflow should boost gene editing in hPSCs and leverage a wide range of applications, including cell line development (e.g., reporter and isogenic cell lines), disease modeling and applications in regenerative medicine.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Humans , Cell Culture Techniques/methods , Cell Line , Cell Differentiation , Cloning, Molecular
7.
Methods Mol Biol ; 2454: 811-827, 2022.
Article in English | MEDLINE | ID: mdl-34128205

ABSTRACT

Human pluripotent stem cells (hPSCs), such as induced pluripotent stem cells (iPSCs), hold great promise for drug discovery, toxicology studies, and regenerative medicine. Here, we describe standardized protocols and experimental procedures that combine automated cell culture for scalable production of hPSCs with quantitative high-throughput screening (qHTS) in miniaturized 384-well plates. As a proof of principle, we established dose-response assessments and determined optimal concentrations of 12 small molecule compounds that are commonly used in the stem cell field. Multi-parametric analysis of readouts from diverse assays including cell viability, mitochondrial membrane potential, plasma membrane integrity, and ATP production was used to distinguish normal biological responses from cellular stress induced by small molecule treatment. Collectively, the establishment of integrated workflows for cell manufacturing, qHTS, high-content imaging, and data analysis provides an end-to-end platform for industrial-scale projects and should leverage the drug discovery process using hPSC-derived cell types.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Cell Culture Techniques/methods , Cell Differentiation/physiology , Drug Evaluation, Preclinical , High-Throughput Screening Assays/methods , Humans
8.
Stem Cell Reports ; 16(12): 3076-3092, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34861164

ABSTRACT

Efficient translation of human induced pluripotent stem cells (hiPSCs) requires scalable cell manufacturing strategies for optimal self-renewal and functional differentiation. Traditional manual cell culture is variable and labor intensive, posing challenges for high-throughput applications. Here, we established a robotic platform and automated all essential steps of hiPSC culture and differentiation under chemically defined conditions. This approach allowed rapid and standardized manufacturing of billions of hiPSCs that can be produced in parallel from up to 90 different patient- and disease-specific cell lines. Moreover, we established automated multi-lineage differentiation and generated functional neurons, cardiomyocytes, and hepatocytes. To validate our approach, we compared robotic and manual cell culture operations and performed comprehensive molecular and cellular characterizations (e.g., single-cell transcriptomics, mass cytometry, metabolism, electrophysiology) to benchmark industrial-scale cell culture operations toward building an integrated platform for efficient cell manufacturing for disease modeling, drug screening, and cell therapy.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Robotics , Automation , Cell Lineage , Cells, Cultured , Embryoid Bodies/cytology , Hepatocytes/cytology , Hepatocytes/virology , Human Embryonic Stem Cells/cytology , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/virology , Neurons/cytology , RNA-Seq , Reference Standards , Single-Cell Analysis , Zika Virus Infection/pathology
9.
Nat Methods ; 18(5): 528-541, 2021 05.
Article in English | MEDLINE | ID: mdl-33941937

ABSTRACT

Human pluripotent stem cells (hPSCs) are capable of extensive self-renewal yet remain highly sensitive to environmental perturbations in vitro, posing challenges to their therapeutic use. There is an urgent need to advance strategies that ensure safe and robust long-term growth and functional differentiation of these cells. Here, we deployed high-throughput screening strategies to identify a small-molecule cocktail that improves viability of hPSCs and their differentiated progeny. The combination of chroman 1, emricasan, polyamines, and trans-ISRIB (CEPT) enhanced cell survival of genetically stable hPSCs by simultaneously blocking several stress mechanisms that otherwise compromise cell structure and function. CEPT provided strong improvements for several key applications in stem-cell research, including routine cell passaging, cryopreservation of pluripotent and differentiated cells, embryoid body (EB) and organoid formation, single-cell cloning, and genome editing. Thus, CEPT represents a unique poly-pharmacological strategy for comprehensive cytoprotection, providing a rationale for efficient and safe utilization of hPSCs.


Subject(s)
Cell Differentiation/drug effects , Cell Survival/drug effects , Cryoprotective Agents/pharmacology , Pluripotent Stem Cells/drug effects , Polypharmacology , Cell Culture Techniques , Cryopreservation/methods , Cryoprotective Agents/chemistry , Gene Expression Regulation/drug effects , High-Throughput Screening Assays , Humans , Pluripotent Stem Cells/physiology , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
10.
bioRxiv ; 2020 Aug 03.
Article in English | MEDLINE | ID: mdl-32793899

ABSTRACT

Efficient translation of human induced pluripotent stem cells (hiPSCs) depends on implementing scalable cell manufacturing strategies that ensure optimal self-renewal and functional differentiation. Currently, manual culture of hiPSCs is highly variable and labor-intensive posing significant challenges for high-throughput applications. Here, we established a robotic platform and automated all essential steps of hiPSC culture and differentiation under chemically defined conditions. This streamlined approach allowed rapid and standardized manufacturing of billions of hiPSCs that can be produced in parallel from up to 90 different patient-and disease-specific cell lines. Moreover, we established automated multi-lineage differentiation to generate primary embryonic germ layers and more mature phenotypes such as neurons, cardiomyocytes, and hepatocytes. To validate our approach, we carefully compared robotic and manual cell culture and performed molecular and functional cell characterizations (e.g. bulk culture and single-cell transcriptomics, mass cytometry, metabolism, electrophysiology, Zika virus experiments) in order to benchmark industrial-scale cell culture operations towards building an integrated platform for efficient cell manufacturing for disease modeling, drug screening, and cell therapy. Combining stem cell-based models and non-stop robotic cell culture may become a powerful strategy to increase scientific rigor and productivity, which are particularly important during public health emergencies (e.g. opioid crisis, COVID-19 pandemic).

11.
Methods Mol Biol ; 1919: 59-72, 2019.
Article in English | MEDLINE | ID: mdl-30656621

ABSTRACT

Human pluripotent stem cells (hPSCs) are characterized by their ability to self-renew and differentiate into any cell type of the human body. To fully utilize the potential of hPSCs for translational research and clinical applications, it is critical to develop rigorous cell differentiation protocols under feeder-free conditions that are efficient, reproducible, and scalable for high-throughput projects. Focusing on neural conversion of hPSCs, here we describe robust small molecule-based procedures that generate neural stem cells (NSCs) in less than a week under chemically defined conditions. These protocols can be used to dissect the mechanisms of neural lineage entry and to further develop systematic protocols that produce the cellular diversity of the central nervous system at industrial scale.


Subject(s)
Cell Differentiation/drug effects , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Animals , Biomarkers , Cell Culture Techniques , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Humans , Immunohistochemistry , Immunophenotyping , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Mice , Neural Stem Cells/metabolism , Pluripotent Stem Cells/metabolism
12.
Oncotarget ; 8(9): 14860-14875, 2017 Feb 28.
Article in English | MEDLINE | ID: mdl-28118603

ABSTRACT

A hallmark of cellular transformation is the evasion of contact-dependent inhibition of growth. To find new therapeutic targets for glioblastoma, we looked for pathways that are inhibited by high cell density in astrocytes but not in glioma cells. Here we report that glioma cells have disabled the normal controls on cholesterol synthesis. At high cell density, astrocytes turn off cholesterol synthesis genes and have low cholesterol levels, but glioma cells keep this pathway on and maintain high cholesterol. Correspondingly, cholesterol pathway upregulation is associated with poor prognosis in glioblastoma patients. Densely-plated glioma cells increase oxygen consumption, aerobic glycolysis, and the pentose phosphate pathway to synthesize cholesterol, resulting in a decrease in reactive oxygen species, TCA cycle intermediates, and ATP. This constitutive cholesterol synthesis is controlled by the cell cycle, as it can be turned off by cyclin-dependent kinase inhibitors and it correlates with disabled cell cycle control though loss of p53 and RB. Finally, glioma cells, but not astrocytes, are sensitive to cholesterol synthesis inhibition downstream of the mevalonate pathway, suggesting that specifically targeting cholesterol synthesis might be an effective treatment for glioblastoma.


Subject(s)
Astrocytes/metabolism , Brain Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Transformation, Neoplastic/pathology , Cholesterol/metabolism , Glioblastoma/pathology , Astrocytes/cytology , Astrocytes/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Count , Cell Division , Cell Transformation, Neoplastic/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glycolysis/drug effects , Humans , Oxygen Consumption/drug effects , Protein Kinase Inhibitors/pharmacology , Tumor Cells, Cultured
13.
J Biol Chem ; 290(1): 56-64, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25391652

ABSTRACT

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) plays roles in both energy maintenance, and stress signaling by forming a protein complex with seven in absentia homolog 1 (Siah1). Mechanisms to coordinate its glycolytic and stress cascades are likely to be very important for survival and homeostatic control of any living organism. Here we report that apoptosis signal-regulating kinase 1 (ASK1), a representative stress kinase, interacts with both GAPDH and Siah1 and is likely able to phosphorylate Siah1 at specific amino acid residues (Thr-70/Thr-74 and Thr-235/Thr-239). Phosphorylation of Siah1 by ASK1 triggers GAPDH-Siah1 stress signaling and activates a key downstream target, p300 acetyltransferase in the nucleus. This novel mechanism, together with the established S-nitrosylation/oxidation of GAPDH at Cys-150, provides evidence of how the stress signaling involving GAPDH is finely regulated. In addition, the present results imply crosstalk between the ASK1 and GAPDH-Siah1 stress cascades.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Nuclear Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Signal Transduction/genetics , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Binding Sites , Gene Expression Regulation , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/genetics , HEK293 Cells , Humans , Hydrogen Peroxide/pharmacology , MAP Kinase Kinase Kinase 5/genetics , Molecular Sequence Data , Nuclear Proteins/genetics , Oxidative Stress , Phosphorylation/drug effects , Protein Binding/drug effects , Recombinant Fusion Proteins/genetics , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...