Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.237
Filter
2.
Stem Cell Reports ; 19(8): 1122-1136, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39094561

ABSTRACT

Reactive astrocytes are known to exert detrimental effects upon neurons in several neurodegenerative diseases, yet our understanding of how astrocytes promote neurotoxicity remains incomplete, especially in human systems. In this study, we leveraged human pluripotent stem cell (hPSC) models to examine how reactivity alters astrocyte function and mediates neurodegeneration. hPSC-derived astrocytes were induced to a reactive phenotype, at which point they exhibited a hypertrophic profile and increased complement C3 expression. Functionally, reactive astrocytes displayed decreased intracellular calcium, elevated phagocytic capacity, and decreased contribution to the blood-brain barrier. Subsequently, co-culture of reactive astrocytes with a variety of neuronal cell types promoted morphological and functional alterations. Furthermore, when reactivity was induced in astrocytes from patient-specific hPSCs (glaucoma, Alzheimer's disease, and amyotrophic lateral sclerosis), the reactive state exacerbated astrocytic disease-associated phenotypes. These results demonstrate how reactive astrocytes modulate neurodegeneration, significantly contributing to our understanding of a role for reactive astrocytes in neurodegenerative diseases.


Subject(s)
Astrocytes , Coculture Techniques , Pluripotent Stem Cells , Astrocytes/metabolism , Humans , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Complement C3/metabolism , Cell Differentiation , Neurons/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Phagocytosis , Blood-Brain Barrier/metabolism , Glaucoma/pathology , Glaucoma/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Calcium/metabolism , Phenotype
3.
Stem Cell Reports ; 19(8): 1137-1155, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39094563

ABSTRACT

Cell size is a crucial physical property that significantly impacts cellular physiology and function. However, the influence of cell size on stem cell specification remains largely unknown. Here, we investigated the dynamic changes in cell size during the differentiation of human pluripotent stem cells into definitive endoderm (DE). Interestingly, cell size exhibited a gradual decrease as DE differentiation progressed with higher stiffness. Furthermore, the application of hypertonic pressure or chemical to accelerate the reduction in cell size significantly and specifically enhanced DE differentiation. By functionally intervening in mechanosensitive elements, we have identified actomyosin activity as a crucial mediator of both DE differentiation and cell size reduction. Mechanistically, the reduction in cell size induces actomyosin-dependent angiomotin (AMOT) nuclear translocation, which suppresses Yes-associated protein (YAP) activity and thus facilitates DE differentiation. Together, our study has established a novel connection between cell size diminution and DE differentiation, which is mediated by AMOT nuclear translocation. Additionally, our findings suggest that the application of osmotic pressure can effectively promote human endodermal lineage differentiation.


Subject(s)
Actomyosin , Angiomotins , Cell Differentiation , Cell Size , Endoderm , Signal Transduction , Transcription Factors , YAP-Signaling Proteins , Humans , Endoderm/cytology , Endoderm/metabolism , Actomyosin/metabolism , YAP-Signaling Proteins/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Osmotic Pressure , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Cell Nucleus/metabolism
4.
Int J Mol Sci ; 25(15)2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39125777

ABSTRACT

Allogeneic natural killer (NK) cell therapy has been effective in treating cancer. Many studies have tested NK cell therapy using human pluripotent stem cells (hPSCs). However, the impacts of the origin of PSC-NK cells on competence are unclear. In this study, several types of hPSCs, including human-induced PSCs (hiPSCs) generated from CD34+, CD3-CD56+, and CD56- cells in umbilical cord blood (UCB), three lines of human embryonic stem cells (hESCs, ES-1. ES-2 and ES-3) and MHC I knockout (B2M-KO)-ESCs were used to differentiate into NK cells and their capacities were analyzed. All PSC types could differentiate into NK cells. Among the iPSC-derived NK cells (iPSC-NKs) and ESC-derived NK cells (ES-NKs), 34+ iPSCs and ES-3 had a higher growth rate and cytotoxicity, respectively, ES-3 also showed better efficacy than 34+ iPSCs. B2M-KO was similar to the wild type. These results suggest that the screening for differentiation of PSCs into NK cells prior to selecting the PSC lines for the development of NK cell immunotherapy is an essential process for universal allotransplantation, including the chimeric antigen receptor (CAR).


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , Killer Cells, Natural , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Cell Line
5.
Cell Mol Life Sci ; 81(1): 366, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39172275

ABSTRACT

BACKGROUND: Chromosome stability is crucial for homeostasis of pluripotent stem cells (PSCs) and early-stage embryonic development. Chromosomal defects may raise carcinogenic risks in regenerative medicine when using PSCs as original materials. However, the detailed mechanism regarding PSCs chromosome stability maintenance is not fully understood. METHODS: Mouse embryonic stem cells (line D3) and human embryonic stem cells (line H9) were cultured under standard conditions. To confirm the loading of RetSat protein on mitotic chromosomes of PSCs, immunostaining was performed in PSCs spontaneous differentiation assay and iPSC reprogramming assay from mouse embryonic fibroblasts (MEFs), respectively. In addition, qPCR, immunoprecipitation, LC-MS/MS and immunoblotting were used to study the expression of RetSat, and interactions of RetSat with cohesin/condensin components. RNA sequencing and teratoma formation assay was conducted to evaluate the carcinogenic risk of mouse embryonic stem cells with RetSat deletion. RESULTS: We reported a PSC high-expressing gene, RetSat, plays key roles in chromosome stabilization. We identified RetSat protein localizing onto mitotic chromosomes specifically in stemness positive cells such as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). We found dramatic chromosome instability, e.g. chromosome bridging, lagging and interphase micronuclei in mouse and human ESCs when down regulating RetSat. RetSat knock-out mouse ESCs upregulated cancer associated gene pathways, and displayed higher tumorigenic capacities in teratoma formation assay. Mechanistically, we confirmed that RetSat interacts with cohesin/condensin components Smc1a and Nudcd2. RetSat deletion impaired the chromosome loading dosage of Smc1a, Smc3 and Nudcd2. CONCLUSIONS: In summary, we reported RetSat to be a key stabilizer of chromosome condensation in pluripotent stem cells. This highlights the crucial roles of RetSat in early-stage embryonic development, and potential value of RetSat as an effective biomarker for assessing the quality of pluripotent stem cells.


Subject(s)
Chromosomal Instability , Chromosome Segregation , Mitosis , Pluripotent Stem Cells , Animals , Mice , Humans , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Cell Differentiation/genetics , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/cytology
6.
PLoS One ; 19(8): e0308743, 2024.
Article in English | MEDLINE | ID: mdl-39121095

ABSTRACT

Human retinal organoids have become indispensable tools for retinal disease modeling and drug screening. Despite its versatile applications, the long timeframe for their differentiation and maturation limits the throughput of such research. Here, we successfully shortened this timeframe by accelerating human retinal organoid development using unique pharmacological approaches. Our method comprised three key steps: 1) a modified self-formed ectodermal autonomous multizone (SEAM) method, including dual SMAD inhibition and bone morphogenetic protein 4 treatment, for initial neural retinal induction; 2) the concurrent use of a Sonic hedgehog agonist SAG, activin A, and all-trans retinoic acid for rapid retinal cell specification; and 3) switching to SAG treatment alone for robust retinal maturation and lamination. The generated retinal organoids preserved typical morphological features of mature retinal organoids, including hair-like surface structures and well-organized outer layers. These features were substantiated by the spatial immunostaining patterns of several retinal cell markers, including rhodopsin and L/M opsin expression in the outermost layer, which was accompanied by reduced ectopic cone photoreceptor generation. Importantly, our method required only 90 days for retinal organoid maturation, which is approximately two-thirds the time necessary for other conventional methods. These results indicate that thoroughly optimized pharmacological interventions play a pivotal role in rapid and precise photoreceptor development during human retinal organoid differentiation and maturation. Thus, our present method may expedite human retinal organoid research, eventually contributing to the development of better treatment options for various degenerative retinal diseases.


Subject(s)
Activins , Cell Differentiation , Hedgehog Proteins , Organoids , Retina , Signal Transduction , Tretinoin , Humans , Activins/pharmacology , Activins/metabolism , Organoids/drug effects , Organoids/metabolism , Organoids/cytology , Hedgehog Proteins/metabolism , Tretinoin/pharmacology , Retina/metabolism , Retina/cytology , Retina/drug effects , Signal Transduction/drug effects , Cell Differentiation/drug effects , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
7.
ACS Biomater Sci Eng ; 10(7): 4525-4540, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38973308

ABSTRACT

Lumenogenesis within the epiblast represents a critical step in early human development, priming the embryo for future specification and patterning events. However, little is known about the specific mechanisms that drive this process due to the inability to study the early embryo in vivo. While human pluripotent stem cell (hPSC)-based models recapitulate many aspects of the human epiblast, most approaches for generating these 3D structures rely on ill-defined, reconstituted basement membrane matrices. Here, we designed synthetic, nonadhesive polyethylene glycol (PEG) hydrogel matrices to better understand the role of matrix mechanical cues in iPSC morphogenesis, specifically elastic modulus. First, we identified a narrow range of hydrogel moduli that were conducive to the hPSC viability, pluripotency, and differentiation. We then used this platform to investigate the effects of the hydrogel modulus on lumenogenesis, finding that matrices of intermediate stiffness yielded the most epiblast-like aggregates. Conversely, stiffer matrices impeded lumen formation and apico-basal polarization, while the softest matrices yielded polarized but aberrant structures. Our approach offers a simple, modular platform for modeling the human epiblast and investigating the role of matrix cues in its morphogenesis.


Subject(s)
Cell Differentiation , Hydrogels , Morphogenesis , Polyethylene Glycols , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Cell Differentiation/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Germ Layers/cytology , Elastic Modulus , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects
8.
Methods Mol Biol ; 2805: 51-87, 2024.
Article in English | MEDLINE | ID: mdl-39008174

ABSTRACT

We describe a scalable method for the robust generation of 3D pancreatic islet-like organoids from human pluripotent stem cells using suspension bioreactors. Our protocol involves a 6-stage, 20-day directed differentiation process, resulting in the production of 104-105 organoids. These organoids comprise α- and ß-like cells that exhibit glucose-responsive insulin and glucagon secretion. We detail methods for culturing, passaging, and cryopreserving stem cells as suspended clusters and for differentiating them through specific growth media and exogenous factors added in a stepwise manner. Additionally, we address quality control measures, troubleshooting strategies, and functional assays for research applications.


Subject(s)
Bioreactors , Cell Culture Techniques , Cell Differentiation , Islets of Langerhans , Organoids , Pluripotent Stem Cells , Humans , Organoids/cytology , Organoids/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Cell Culture Techniques/methods , Cryopreservation/methods
9.
Nat Commun ; 15(1): 5898, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003323

ABSTRACT

Studying human fetal lungs can inform how developmental defects and disease states alter the function of the lungs. Here, we sequenced >150,000 single cells from 19 healthy human pseudoglandular fetal lung tissues ranging between gestational weeks 10-19. We capture dynamic developmental trajectories from progenitor cells that express abundant levels of the cystic fibrosis conductance transmembrane regulator (CFTR). These cells give rise to multiple specialized epithelial cell types. Combined with spatial transcriptomics, we show temporal regulation of key signalling pathways that may drive the temporal and spatial emergence of specialized epithelial cells including ciliated and pulmonary neuroendocrine cells. Finally, we show that human pluripotent stem cell-derived fetal lung models contain CFTR-expressing progenitor cells that capture similar lineage developmental trajectories as identified in the native tissue. Overall, this study provides a comprehensive single-cell atlas of the developing human lung, outlining the temporal and spatial complexities of cell lineage development and benchmarks fetal lung cultures from human pluripotent stem cell differentiations to similar developmental window.


Subject(s)
Cell Differentiation , Cystic Fibrosis Transmembrane Conductance Regulator , Epithelial Cells , Fetus , Lung , Humans , Lung/embryology , Lung/cytology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fetus/cytology , Fetus/embryology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cell Plasticity , Cell Lineage , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Single-Cell Analysis , Transcriptome , Female , Gene Expression Regulation, Developmental , Signal Transduction
10.
Nat Commun ; 15(1): 6344, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39068220

ABSTRACT

Dysfunction of pancreatic δ cells contributes to the etiology of diabetes. Despite their important role, human δ cells are scarce, limiting physiological studies and drug discovery targeting δ cells. To date, no directed δ-cell differentiation method has been established. Here, we demonstrate that fibroblast growth factor (FGF) 7 promotes pancreatic endoderm/progenitor differentiation, whereas FGF2 biases cells towards the pancreatic δ-cell lineage via FGF receptor 1. We develop a differentiation method to generate δ cells from human stem cells by combining FGF2 with FGF7, which synergistically directs pancreatic lineage differentiation and modulates the expression of transcription factors and SST activators during endoderm/endocrine precursor induction. These δ cells display mature RNA profiles and fine secretory granules, secrete somatostatin in response to various stimuli, and suppress insulin secretion from in vitro co-cultured ß cells and mouse ß cells upon transplantation. The generation of human pancreatic δ cells from stem cells in vitro would provide an unprecedented cell source for drug discovery and cell transplantation studies in diabetes.


Subject(s)
Cell Differentiation , Pluripotent Stem Cells , Humans , Animals , Mice , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/cytology , Somatostatin-Secreting Cells/metabolism , Somatostatin-Secreting Cells/cytology , Endoderm/cytology , Endoderm/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Pancreas/cytology , Pancreas/metabolism , Somatostatin/metabolism , Cell Lineage , Insulin/metabolism , Insulin Secretion
11.
Nat Commun ; 15(1): 5894, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003281

ABSTRACT

Remarkable advances in protocol development have been achieved to manufacture insulin-secreting islets from human pluripotent stem cells (hPSCs). Distinct from current approaches, we devised a tunable strategy to generate islet spheroids enriched for major islet cell types by incorporating PDX1+ cell budding morphogenesis into staged differentiation. In this process that appears to mimic normal islet morphogenesis, the differentiating islet spheroids organize with endocrine cells that are intermingled or arranged in a core-mantle architecture, accompanied with functional heterogeneity. Through in vitro modelling of human pancreas development, we illustrate the importance of PDX1 and the requirement for EphB3/4 signaling in eliciting cell budding morphogenesis. Using this new approach, we model Mitchell-Riley syndrome with RFX6 knockout hPSCs illustrating unexpected morphogenesis defects in the differentiation towards islet cells. The tunable differentiation system and stem cell-derived islet models described in this work may facilitate addressing fundamental questions in islet biology and probing human pancreas diseases.


Subject(s)
Cell Differentiation , Homeodomain Proteins , Islets of Langerhans , Morphogenesis , Pluripotent Stem Cells , Spheroids, Cellular , Trans-Activators , Humans , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Trans-Activators/metabolism , Trans-Activators/genetics , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Signal Transduction , Receptors, Eph Family/metabolism , Receptors, Eph Family/genetics
12.
Int J Mol Sci ; 25(14)2024 Jul 21.
Article in English | MEDLINE | ID: mdl-39063213

ABSTRACT

Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have emerged as a promising tool for studying cardiac physiology and drug responses. However, their use is largely limited by an immature phenotype and lack of high-throughput analytical methodology. In this study, we developed a high-throughput testing platform utilizing hPSC-CMs to assess the cardiotoxicity and effectiveness of drugs. Following an optimized differentiation and maturation protocol, hPSC-CMs exhibited mature CM morphology, phenotype, and functionality, making them suitable for drug testing applications. We monitored intracellular calcium dynamics using calcium imaging techniques to measure spontaneous calcium oscillations in hPSC-CMs in the presence or absence of test compounds. For the cardiotoxicity test, hPSC-CMs were treated with various compounds, and calcium flux was measured to evaluate their effects on calcium dynamics. We found that cardiotoxic drugs withdrawn due to adverse drug reactions, including encainide, mibefradil, and cetirizine, exhibited toxicity in hPSC-CMs but not in HEK293-hERG cells. Additionally, in the effectiveness test, hPSC-CMs were exposed to ATX-II, a sodium current inducer for mimicking long QT syndrome type 3, followed by exposure to test compounds. The observed changes in calcium dynamics following drug exposure demonstrated the utility of hPSC-CMs as a versatile model system for assessing both cardiotoxicity and drug efficacy. Overall, our findings highlight the potential of hPSC-CMs in advancing drug discovery and development, which offer a physiologically relevant platform for the preclinical screening of novel therapeutics.


Subject(s)
Cell Differentiation , Drug Evaluation, Preclinical , Myocytes, Cardiac , Pluripotent Stem Cells , Humans , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Drug Evaluation, Preclinical/methods , Cell Differentiation/drug effects , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Cardiovascular Agents/pharmacology , Calcium/metabolism , Cardiotoxicity , High-Throughput Screening Assays/methods , HEK293 Cells , Calcium Signaling/drug effects
13.
Mol Genet Genomics ; 299(1): 74, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39085666

ABSTRACT

Derivation of hypoimmunogenic human cells from genetically manipulated pluripotent stem cells holds great promise for future transplantation medicine and adoptive immunotherapy. Disruption of beta-2-microglobulin (B2M) in pluripotent stem cells followed by differentiation into specialized cell types is a promising approach to derive hypoimmunogenic cells. Given the attractive features of CRISPR/Cas9-based gene editing tool and baculoviral delivery system, baculovirus can deliver CRISPR/Cas9 components for site-specific gene editing of B2M. Herein, we report the development of a baculoviral CRISPR/Cas9 vector system for the B2M locus disruption in human cells. When tested in human embryonic stem cells (hESCs), the B2M gene knockdown/out was successfully achieved, leading to the stable down-regulation of human leukocyte antigen class I expression on the cell surface. Fibroblasts derived from the B2M gene-disrupted hESCs were then used as stimulator cells in the co-cultures with human peripheral blood mononuclear cells. These fibroblasts triggered significantly reduced alloimmune responses as assessed by sensitive Elispot assays. The B2M-negative hESCs maintained the pluripotency and the ability to differentiate into three germ lineages in vitro and in vivo. These findings demonstrated the feasibility of using the baculoviral-CRISPR/Cas9 system to establish B2M-disrupted pluripotent stem cells. B2M knockdown/out sufficiently leads to hypoimmunogenic conditions, thereby supporting the potential use of B2M-negative cells as universal donor cells for allogeneic cell therapy.


Subject(s)
Baculoviridae , CRISPR-Cas Systems , Cell Differentiation , Gene Editing , Genetic Vectors , Pluripotent Stem Cells , beta 2-Microglobulin , Humans , beta 2-Microglobulin/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Baculoviridae/genetics , Gene Editing/methods , Genetic Vectors/genetics , Cell Differentiation/genetics , Gene Knockout Techniques/methods , Animals , Fibroblasts/metabolism , Fibroblasts/cytology , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Mice
14.
Cell Stem Cell ; 31(8): 1113-1126.e6, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38981471

ABSTRACT

Emerging human pluripotent stem cell (hPSC)-based embryo models are useful for studying human embryogenesis. Particularly, there are hPSC-based somitogenesis models using free-floating culture that recapitulate somite formation. Somitogenesis in vivo involves intricately orchestrated biochemical and biomechanical events. However, none of the current somitogenesis models controls biochemical gradients or biomechanical signals in the culture, limiting their applicability to untangle complex biochemical-biomechanical interactions that drive somitogenesis. Herein, we develop a human somitogenesis model by confining hPSC-derived presomitic mesoderm (PSM) tissues in microfabricated trenches. Exogenous microfluidic morphogen gradients imposed on the PSM tissues cause axial patterning and trigger spontaneous rostral-to-caudal somite formation. A mechanical theory is developed to explain the size dependency between somites and the PSM. The microfluidic somitogenesis model is further exploited to reveal regulatory roles of cellular and tissue biomechanics in somite formation. This study presents a useful microengineered, hPSC-based model for understanding the biochemical and biomechanical events that guide somite formation.


Subject(s)
Microfluidics , Models, Biological , Pluripotent Stem Cells , Somites , Humans , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Somites/cytology , Somites/metabolism , Microfluidics/methods , Embryonic Development , Mesoderm/cytology , Cell Differentiation
15.
Stem Cell Res Ther ; 15(1): 213, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39020441

ABSTRACT

BACKGROUND: Commonly used media for the differentiation of human pluripotent stem cells into cardiomyocytes (hPSC-CMs) contain high concentrations of proteins, in particular albumin, which is prone to quality variations and presents a substantial cost factor, hampering the clinical translation of in vitro-generated cardiomyocytes for heart repair. To overcome these limitations, we have developed chemically defined, entirely protein-free media based on RPMI, supplemented with L-ascorbic acid 2-phosphate (AA-2P) and either the non-ionic surfactant Pluronic F-68 or a specific polyvinyl alcohol (PVA). METHODS AND RESULTS: Both media compositions enable the efficient, directed differentiation of embryonic and induced hPSCs, matching the cell yields and cardiomyocyte purity ranging from 85 to 99% achieved with the widely used protein-based CDM3 medium. The protein-free differentiation approach was readily up-scaled to a 2000 mL process scale in a fully controlled stirred tank bioreactor in suspension culture, producing > 1.3 × 109 cardiomyocytes in a single process run. Transcriptome analysis, flow cytometry, electrophysiology, and contractile force measurements revealed that the mass-produced cardiomyocytes differentiated in protein-free medium exhibit the expected ventricular-like properties equivalent to the well-established characteristics of CDM3-control cells. CONCLUSIONS: This study promotes the robustness and upscaling of the cardiomyogenic differentiation process, substantially reduces media costs, and provides an important step toward the clinical translation of hPSC-CMs for heart regeneration.


Subject(s)
Cell Differentiation , Culture Media , Myocytes, Cardiac , Humans , Cell Differentiation/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Culture Media/chemistry , Culture Media/pharmacology , Cell Culture Techniques/methods , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/drug effects , Cells, Cultured
16.
Development ; 151(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39069943

ABSTRACT

Naïve epiblast cells in the embryo and pluripotent stem cells in vitro undergo developmental progression to a formative state competent for lineage specification. During this transition, transcription factors and chromatin are rewired to encode new functional features. Here, we examine the role of mitogen-activated protein kinase (ERK1/2) signalling in pluripotent state transition. We show that a primary consequence of ERK activation in mouse embryonic stem cells is elimination of Nanog, which precipitates breakdown of the naïve state gene regulatory network. Variability in pERK dynamics results in heterogeneous loss of Nanog and metachronous state transition. Knockdown of Nanog allows exit without ERK activation. However, transition to formative pluripotency does not proceed and cells collapse to an indeterminate identity. This outcome is due to failure to maintain expression of the central pluripotency factor Oct4. Thus, during formative transition ERK signalling both dismantles the naïve state and preserves pluripotency. These results illustrate how a single signalling pathway can both initiate and secure transition between cell states.


Subject(s)
MAP Kinase Signaling System , Nanog Homeobox Protein , Octamer Transcription Factor-3 , Pluripotent Stem Cells , Animals , Nanog Homeobox Protein/metabolism , Nanog Homeobox Protein/genetics , Mice , Octamer Transcription Factor-3/metabolism , Octamer Transcription Factor-3/genetics , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Cell Differentiation/genetics , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Germ Layers/metabolism , Germ Layers/cytology , Gene Regulatory Networks , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics
17.
Stem Cell Res ; 79: 103482, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38959701

ABSTRACT

The recently issued ISSCR standards in stem cell research recommend registration of human pluripotent stem cell lines (hPSCs). Registration is critical to establishing stem cell provenance and connecting cell lines to data derived on those lines. In this study, we sought to understand common barriers to registration by conducting interviews with forty-eight Australian stem cell stakeholders, including researchers, clinicians, and industry professionals. Australian stem cell researchers do not routinely register their lines, and only a third of those Australian lines captured by an international registry have fully completed the registration process. Most registered Australian cell lines lack complete information about their ethical provenance or key pluripotency characteristics. Incomplete registration is poorly aligned with the goals of open science on which registries are founded. Users also expressed concerns about the quality of the incomplete information provided to the resource. Registration was considered negatively, for instance as a hurdle or barrier to publication, which impacted on user perceptions of usefulness of registration and lowered the likelihood that they would engage with registries to find resources. Broader adoption of registration by journals, and continued advocacy by stem cell societies, will be important levers for awareness and engagement with registration. Although the Australian community represents a small fraction of potential registry users, the results of this study suggest ways for journals, registries, funders, and the international stem cell community to improve registration compliance.


Subject(s)
Registries , Research Personnel , Humans , Australia , Stem Cell Research , Cell Line , Pluripotent Stem Cells/cytology
18.
Cell Stem Cell ; 31(7): 945-946, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38971145

ABSTRACT

Dattani et al.1 developed a method for inducing hypoblast-like cells from human naive pluripotent stem cells. They elucidated the requirement for FGF signaling in human hypoblast specialization at a specific time window, which was previously controversial.


Subject(s)
Fibroblast Growth Factors , Signal Transduction , Humans , Fibroblast Growth Factors/metabolism , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Cell Differentiation
19.
J Gene Med ; 26(7): e3716, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38961849

ABSTRACT

BACKGROUND: Differentiation of pluripotent stem cells into desired lineages is the key aspect of regenerative medicine and cell-based therapy. Although RNA interference (RNAi) technology is exploited extensively for this, methods for long term silencing of the target genes leading to differentiation remain a challenge. Sustained knockdown of the target gene by RNAi is often inefficient as a result of low delivery efficiencies, protocol induced toxicity and safety concerns related to viral vectors. Earlier, we established octa-arginine functionalized hydroxyapatite nano vehicles (R8HNPs) for delivery of small interfering RNA (siRNA) against a pluripotency marker gene in mouse embryonic stem cells. Although we demonstrated excellent knockdown efficiency of the target gene, sustained gene silencing leading to differentiation was yet to be achieved. METHODS: To establish a sustained non-viral gene silencing protocol using R8HNP, we investigated various methods of siRNA delivery: double delivery of adherent cells (Adh-D), suspension delivery followed by adherent delivery (Susp + Adh), single delivery in suspension (Susp-S) and multiple deliveries in suspension (Susp-R). Sustained knockdown of a pluripotent marker gene followed by differentiation was analysed by reverse transcriptase-PCR, fluoresence-activated cell sorting and immunofluorescence techniques. Impact on cell viability as a result of repeated exposure of the R8HNP was also tested. RESULTS: Amongst the protocols tested, the most efficient knockdown of the target gene for a prolonged period of time was obtained by repeated suspension delivery of the R8HNP-siRNA conjugate. The long-term silencing of a pluripotency marker gene resulted in differentiation of R1 ESCs predominantly towards the extra embryonic and ectodermal lineages. Cells displayed excellent tolerance to repeated exposures of R8HNPs. CONCLUSIONS: The results demonstrate that R8HNPs are promising, biocompatible, non-viral alternatives for prolonged gene silencing and obtaining differentiated cells for therapeutics.


Subject(s)
Cell Differentiation , Durapatite , Mouse Embryonic Stem Cells , RNA, Small Interfering , Animals , Mice , Durapatite/chemistry , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/drug effects , RNA, Small Interfering/genetics , Gene Silencing , Biocompatible Materials/chemistry , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Nanoparticles/chemistry , Transduction, Genetic , RNA Interference , Gene Knockdown Techniques
20.
Cell Mol Life Sci ; 81(1): 297, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992309

ABSTRACT

Muse cells, identified as cells positive for the pluripotent surface marker SSEA-3, are pluripotent-like endogenous stem cells located in the bone marrow (BM), peripheral blood, and organ connective tissues. The detailed characteristics of SSEA-3(+) cells in extraembryonic tissue, however, are unknown. Here, we demonstrated that similar to human-adult tissue-Muse cells collected from the BM, adipose tissue, and dermis as SSEA-3(+), human-umbilical cord (UC)-SSEA-3(+) cells express pluripotency markers, differentiate into triploblastic-lineage cells at a single cell level, migrate to damaged tissue, and exhibit low telomerase activity and non-tumorigenicity. Notably, ~ 20% of human-UC-SSEA-3(+) cells were negative for X-inactive specific transcript (XIST), a naïve pluripotent stem cell characteristic, whereas all human adult tissue-Muse cells are XIST-positive. Single-cell RNA sequencing revealed that the gene expression profile of human-UC-SSEA-3(+) cells was more similar to that of human post-implantation blastocysts than human-adult tissue-Muse cells. The DNA methylation level showed the same trend, and notably, the methylation levels in genes particularly related to differentiation were lower in human-UC-SSEA-3(+) cells than in human-adult tissue-Muse cells. Furthermore, human-UC-SSEA-3(+) cells newly express markers specific to extraembryonic-, germline-, and hematopoietic-lineages after differentiation induction in vitro whereas human-adult tissue-Muse cells respond only partially to the induction. Among various stem/progenitor cells in living bodies, those that exhibit properties similar to post-implantation blastocysts in a naïve state have not yet been found in humans. Easily accessible human-UC-SSEA-3(+) cells may be a valuable tool for studying early-stage human development and human reproductive medicine.


Subject(s)
Blastocyst , Cell Differentiation , Stage-Specific Embryonic Antigens , Umbilical Cord , Humans , Stage-Specific Embryonic Antigens/metabolism , Umbilical Cord/cytology , Blastocyst/cytology , Blastocyst/metabolism , Antigens, Tumor-Associated, Carbohydrate/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Single-Cell Analysis , Telomerase/metabolism , Telomerase/genetics , Female
SELECTION OF CITATIONS
SEARCH DETAIL