Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Stem Cell Reports ; 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38964325

ABSTRACT

Culture-acquired variants in human pluripotent stem cells (hPSCs) hinder their applications in research and clinic. However, the mechanisms that underpin selection of variants remain unclear. Here, through analysis of comprehensive karyotyping datasets from over 23,000 hPSC cultures of more than 1,500 lines, we explored how culture conditions shape variant selection. Strikingly, we identified an association of chromosome 1q gains with feeder-free cultures and noted a rise in its prevalence in recent years, coinciding with increased usage of feeder-free regimens. Competition experiments of multiple isogenic lines with and without a chromosome 1q gain confirmed that 1q variants have an advantage in feeder-free (E8/vitronectin), but not feeder-based, culture. Mechanistically, we show that overexpression of MDM4, located on chromosome 1q, drives variants' advantage in E8/vitronectin by alleviating genome damage-induced apoptosis, which is lower in feeder-based conditions. Our study explains condition-dependent patterns of hPSC aberrations and offers insights into the mechanisms of variant selection.

2.
Bioessays ; : e2400062, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38873900

ABSTRACT

Genetic changes arising in human pluripotent stem cells (hPSC) upon culture may bestow unwanted or detrimental phenotypes to cells, thus potentially impacting on the applications of hPSCs for clinical use and basic research. In the 20 years since the first report of culture-acquired genetic aberrations in hPSCs, a characteristic spectrum of recurrent aberrations has emerged. The preponderance of such aberrations implies that they provide a selective growth advantage to hPSCs upon expansion. However, understanding the consequences of culture-acquired variants for specific applications in cell therapy or research has been more elusive. The rapid progress of hPSC-based therapies to clinics is galvanizing the field to address this uncertainty and provide definitive ways both for risk assessment of variants and reducing their prevalence in culture. Here, we aim to provide a timely update on almost 20 years of research on this fascinating, but a still unresolved and concerning, phenomenon.

3.
Nat Commun ; 15(1): 3745, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702304

ABSTRACT

Early childhood tumours arise from transformed embryonic cells, which often carry large copy number alterations (CNA). However, it remains unclear how CNAs contribute to embryonic tumourigenesis due to a lack of suitable models. Here we employ female human embryonic stem cell (hESC) differentiation and single-cell transcriptome and epigenome analysis to assess the effects of chromosome 17q/1q gains, which are prevalent in the embryonal tumour neuroblastoma (NB). We show that CNAs impair the specification of trunk neural crest (NC) cells and their sympathoadrenal derivatives, the putative cells-of-origin of NB. This effect is exacerbated upon overexpression of MYCN, whose amplification co-occurs with CNAs in NB. Moreover, CNAs potentiate the pro-tumourigenic effects of MYCN and mutant NC cells resemble NB cells in tumours. These changes correlate with a stepwise aberration of developmental transcription factor networks. Together, our results sketch a mechanistic framework for the CNA-driven initiation of embryonal tumours.


Subject(s)
Cell Differentiation , DNA Copy Number Variations , N-Myc Proto-Oncogene Protein , Neural Crest , Neuroblastoma , Humans , Neuroblastoma/genetics , Neuroblastoma/pathology , Neural Crest/metabolism , Neural Crest/pathology , Female , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , Chromosome Aberrations , Human Embryonic Stem Cells/metabolism , Transcriptome , Cell Line, Tumor , Gene Expression Regulation, Neoplastic
4.
Cytotherapy ; 26(4): 383-392, 2024 04.
Article in English | MEDLINE | ID: mdl-38349312

ABSTRACT

BACKGROUND AIMS: The appearance of genetically variant populations in human pluripotent stem cell (hPSC) cultures represents a concern for research and clinical applications. Genetic variations may alter hPSC differentiation potential or cause phenotype variation in differentiated cells. Further, variants may have properties such as proliferative rate, or response to the culture environment, that differ from wild-type cells. As such, understanding the behavior of these variants in culture, and any potential operational impact on manufacturing processes, will be necessary to control quality of putative hPSC-based products that include a proportion of variant threshold in their quality specification. METHODS: Here we show a computational model that mathematically describes the growth dynamics between commonly occurring genetically variant hPSCs and their counterpart wild-type cells in culture. RESULTS: We show that our model is capable of representing the growth behaviors of both wild-type and variant hPSCs in individual and co-culture systems. CONCLUSIONS: This representation allows us to identify three critical process parameters that drive critical quality attributes when genetically variant cells are present within the system: total culture density, proportion of variant cells within the culture system and variant cell overgrowth. Lastly, we used our model to predict how the variability of these parameters affects the prevalence of both populations in culture.


Subject(s)
Cell Culture Techniques , Pluripotent Stem Cells , Humans , Cell Differentiation/genetics , Coculture Techniques
5.
ACS Appl Mater Interfaces ; 16(2): 2154-2165, 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-38181419

ABSTRACT

Extracellular matrices interface with cells to promote cell growth and tissue development. Given this critical role, matrix mimetics are introduced to enable biomedical materials ranging from tissue engineering scaffolds and tumor models to organoids for drug screening and implant surface coatings. Traditional microscopy methods are used to evaluate such materials in their ability to support exploitable cell responses, which are expressed in changes in cell proliferation rates and morphology. However, the physical imaging methods do not capture the chemistry of cells at cell-matrix interfaces. Herein, we report hyperspectral imaging to map the chemistry of human primary and embryonic stem cells grown on matrix materials, both native and artificial. We provide the statistical analysis of changes in lipid and protein content of the cells obtained from infrared spectral maps to conclude matrix morphologies as a major determinant of biochemical cell responses. The study demonstrates an effective methodology for evaluating bespoke matrix materials directly at cell-matrix interfaces.


Subject(s)
Biocompatible Materials , Tissue Scaffolds , Humans , Tissue Scaffolds/chemistry , Biocompatible Materials/chemistry , Tissue Engineering/methods , Extracellular Matrix/chemistry , Embryonic Stem Cells
6.
Stem Cell Reports ; 18(9): 1744-1752, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37703820

ABSTRACT

The laboratory culture of human stem cells seeks to capture a cellular state as an in vitro surrogate of a biological system. For the results and outputs from this research to be accurate, meaningful, and durable, standards that ensure reproducibility and reliability of the data should be applied. Although such standards have been previously proposed for repositories and distribution centers, no widely accepted best practices exist for laboratory research with human pluripotent and tissue stem cells. To fill that void, the International Society for Stem Cell Research has developed a set of recommendations, including reporting criteria, for scientists in basic research laboratories. These criteria are designed to be technically and financially feasible and, when implemented, enhance the reproducibility and rigor of stem cell research.


Subject(s)
Stem Cell Research , Humans , Reproducibility of Results
7.
Stem Cell Reports ; 18(3): 782-797, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36801002

ABSTRACT

Chromosome 20 abnormalities are some of the most frequent genomic changes acquired by human pluripotent stem cell (hPSC) cultures worldwide. Yet their effects on differentiation remain largely unexplored. We investigated a recurrent abnormality also found on amniocentesis, the isochromosome 20q (iso20q), during a clinical retinal pigment epithelium differentiation. Here we show that the iso20q abnormality interrupts spontaneous embryonic lineage specification. Isogenic lines revealed that under conditions that promote the spontaneous differentiation of wild-type hPSCs, the iso20q variants fail to differentiate into primitive germ layers and to downregulate pluripotency networks, resulting in apoptosis. Instead, iso20q cells are highly biased for extra-embryonic/amnion differentiation following inhibition of DNMT3B methylation or BMP2 treatment. Finally, directed differentiation protocols can overcome the iso20q block. Our findings reveal in iso20q a chromosomal abnormality that impairs the developmental competency of hPSCs toward germ layers but not amnion, which models embryonic developmental bottlenecks in the presence of aberrations.


Subject(s)
Isochromosomes , Pluripotent Stem Cells , Humans , Cell Differentiation/genetics , Pluripotent Stem Cells/metabolism , Retinal Pigment Epithelium , Germ Layers
8.
Cell Stem Cell ; 29(12): 1624-1636, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36459966

ABSTRACT

It is well established that human pluripotent stem cells (hPSCs) can acquire genetic and epigenetic changes during culture in vitro. Given the increasing use of hPSCs in research and therapy and the vast expansion in the number of hPSC lines available for researchers, the International Society for Stem Cell Research has recognized the need to reassess quality control standards for ensuring the genetic integrity of hPSCs. Here, we summarize current knowledge of the nature of recurrent genetic and epigenetic variants in hPSC culture, the methods for their detection, and what is known concerning their effects on cell behavior in vitro or in vivo. We argue that the potential consequences of low-level contamination of cell therapy products with cells bearing oncogenic variants are essentially unknown at present. We highlight the key challenges facing the field with particular reference to safety assessment of hPSC-derived cellular therapeutics.


Subject(s)
Epigenomics , Pluripotent Stem Cells , Humans , Stem Cell Research , Oncogenes , Epigenesis, Genetic
9.
Curr Protoc ; 2(11): e606, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36426882

ABSTRACT

Human pluripotent stem cells (hPSCs) can be grown in culture indefinitely, making them a valuable tool for use in basic biology, disease modeling, and regenerative medicine. However, over prolonged periods in culture, hPSCs tend to acquire genomic aberrations that confer growth advantages, similar to those seen in some cancers. Monitoring the genomic stability of cultured hPSCs is critical to ensuring their efficacy and safety as a therapeutic tool. Most commonly employed methods for monitoring of hPSC genomes are cytogenetic methods, such as G-banding. Nonetheless, such methods have limited resolution and sensitivity for detecting mosaicism. Single nucleotide polymorphism (SNP) array platforms are a potential alternative that could improve detection of abnormalities. Here, we outline protocols for SNP array whole-genome screening of hPSCs. Moreover, we detail the procedure for assessing the SNP array's sensitivity in detecting low-level mosaic copy-number changes. We show that mosaicism can be confidently identified in samples only once they contain 20% variants, although samples containing 10% variants typically display enough variation to warrant further investigation and confirmation, for example by using a more sensitive targeted method. Finally, we highlight the advantages and limitations of SNP arrays, including a cost comparison of SNP arrays versus other commonly employed methods for detection of genetic changes in hPSC cultures. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: DNA sample preparation for SNP arrays Basic Protocol 2: SNP array hybridization, washing, and scanning Basic Protocol 3: SNP array data analysis Support Protocol: Assessment of SNP array sensitivity for detection of mosaicism.


Subject(s)
Pluripotent Stem Cells , Polymorphism, Single Nucleotide , Humans , Polymorphism, Single Nucleotide/genetics , Cytogenetic Analysis , Mosaicism , Nucleic Acid Hybridization
10.
Dev Cell ; 57(11): 1316-1330.e7, 2022 06 06.
Article in English | MEDLINE | ID: mdl-35597240

ABSTRACT

The changes that drive differentiation facilitate the emergence of abnormal cells that need to be removed before they contribute to further development or the germline. Consequently, in mice in the lead-up to gastrulation, ∼35% of embryonic cells are eliminated. This elimination is caused by hypersensitivity to apoptosis, but how it is regulated is poorly understood. Here, we show that upon exit of naive pluripotency, mouse embryonic stem cells lower their mitochondrial apoptotic threshold, and this increases their sensitivity to cell death. We demonstrate that this enhanced apoptotic response is induced by a decrease in mitochondrial fission due to a reduction in the activity of dynamin-related protein 1 (DRP1). Furthermore, we show that in naive pluripotent cells, DRP1 prevents apoptosis by promoting mitophagy. In contrast, during differentiation, reduced mitophagy levels facilitate apoptosis. Together, these results indicate that during early mammalian development, DRP1 regulation of mitophagy determines the apoptotic response.


Subject(s)
Dynamins/metabolism , Mitophagy , Animals , Apoptosis/physiology , Mammals/metabolism , Mice , Mitochondria/metabolism , Mitochondrial Dynamics/physiology , Mitophagy/physiology
11.
Curr Protoc ; 2(5): e435, 2022 May.
Article in English | MEDLINE | ID: mdl-35621694

ABSTRACT

Cell-cell interactions are required for development and homeostasis in multicellular organisms from insects to mammals. A critical process governed by these interactions is cell competition, which functions throughout development to control tissue composition by eliminating cells that possess a lower fitness status than their neighbors. Human pluripotent stem cells (hPSCs) are a key biological tool in modeling human development and offer further potential as a source of clinically relevant cell populations for regenerative medicine applications. Recently, cell competition has been demonstrated in hPSC cultures and during induced pluripotent stem cell reprogramming. In turn, these findings suggest that hPSCs can be used as a tool to study and model cell-cell interactions during different stages of development and disease. Here, we provide a panel of protocols optimized for hPSCs to investigate the potential role that cell competition may have in determining the fate and composition of cell populations during culture. The protocols entail assessment of the competitive phenotype and the mode through which cell competition may lead to elimination of less-fit cells from mosaic cultures with fitter counterparts. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Electroporation of hPSCs to establish a fluorescent reference cell line Support Protocol 1: Single-cell dissociation of hPSCs Support Protocol 2: Single-cell cloning of fluorescently labeled hPSCs Basic Protocol 2: Separate culture and co-culture proliferation assays Basic Protocol 3: Assessing levels of apoptosis in hPSC cultures using flow cytometry Basic Protocol 4: Transwell assay Support Protocol 3: Immunohistochemistry and image quantification of cleaved caspase-3 Basic Protocol 5: Cell confrontation assay Basic Protocol 6: Cell compression assay Basic Protocol 7: Time-lapse imaging to assess mechanical extrusion.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Animals , Cell Competition , Flow Cytometry/methods , Humans , Mammals , Regenerative Medicine
12.
Methods Mol Biol ; 2416: 267-284, 2022.
Article in English | MEDLINE | ID: mdl-34870842

ABSTRACT

The presence of genetic changes in human pluripotent stem cells (hPSCs) can affect their behavior and impact on the utility of hPSC-based applications in research and clinic. The spectrum of spontaneously arising genetic abnormalities in hPSCs is wide and ranges from numerical and structural chromosomal anomalies down to point mutations. The detection of genetic changes in hPSCs is confounded by the fact that no single method detects all types of abnormalities with the same accuracy and sensitivity, therefore necessitating the use of a combination of different methods. Here, we provide detailed protocols for two methods commonly utilized for the detection of genetic changes in naïve and primed hPSCs: karyotyping by G-banding and fluorescent in situ hybridization (FISH).


Subject(s)
Pluripotent Stem Cells , Cell Differentiation , Chromosome Aberrations , Chromosome Banding , Humans , In Situ Hybridization, Fluorescence , Karyotyping
13.
Dev Cell ; 56(17): 2455-2470.e10, 2021 09 13.
Article in English | MEDLINE | ID: mdl-34407428

ABSTRACT

The appearance of genetic changes in human pluripotent stem cells (hPSCs) presents a concern for their use in research and regenerative medicine. Variant hPSCs that harbor recurrent culture-acquired aneuploidies display growth advantages over wild-type diploid cells, but the mechanisms that yield a drift from predominantly wild-type to variant cell populations remain poorly understood. Here, we show that the dominance of variant clones in mosaic cultures is enhanced through competitive interactions that result in the elimination of wild-type cells. This elimination occurs through corralling and mechanical compression by faster-growing variants, causing a redistribution of F-actin and sequestration of yes-associated protein (YAP) in the cytoplasm that induces apoptosis in wild-type cells. YAP overexpression or promotion of YAP nuclear localization in wild-type cells alleviates their "loser" phenotype. Our results demonstrate that hPSC fate is coupled to mechanical cues imposed by neighboring cells and reveal that hijacking this mechanism allows variants to achieve clonal dominance in cultures.


Subject(s)
Cell Competition/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Pluripotent Stem Cells/cytology , YAP-Signaling Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cells, Cultured , Cytoplasm/metabolism , Humans , Transcription Factors/metabolism
14.
Dev Biol ; 476: 209-217, 2021 08.
Article in English | MEDLINE | ID: mdl-33891964

ABSTRACT

A small number of pluripotent cells within early embryo gives rise to all cells in the adult body, including germ cells. Hence, any mutations occurring in the pluripotent cell population are at risk of being propagated to their daughter cells and could lead to congenital defects or embryonic lethality and pose a risk of being transmitted to future generations. The observation that genetic errors are relatively common in preimplantation embryos, but their levels reduce as development progresses, suggests the existence of mechanisms for clearance of aberrant, unfit or damaged cells. Although early human embryogenesis is largely experimentally inaccessible, pluripotent stem cell (PSC) lines can be derived either from the inner cell mass (ICM) of a blastocyst or by reprogramming somatic cells into an embryonic stem cell-like state. PSCs retain the ability to differentiate into any cell type in vitro and, hence, they represent a unique and powerful tool for studying otherwise intractable stages of human development. The advent of PSCs has also opened up a possibility of developing regenerative medicine therapies, either through PSC differentiation in vitro or by creating interspecies chimeras for organ replacement. Here, we discuss the emerging evidence of cell selection in human PSC populations in vivo and in vitro and we highlight the implications of understanding this phenomenon for human development and regenerative medicine.


Subject(s)
Genetic Fitness/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Blastocyst/metabolism , Cell Differentiation , Chimera/embryology , Chimera/genetics , Embryo, Mammalian , Embryonic Stem Cells , Genetic Fitness/physiology , Humans , Pluripotent Stem Cells/physiology , Regenerative Medicine
15.
Development ; 148(6)2021 03 23.
Article in English | MEDLINE | ID: mdl-33658223

ABSTRACT

The anteroposterior axial identity of motor neurons (MNs) determines their functionality and vulnerability to neurodegeneration. Thus, it is a crucial parameter in the design of strategies aiming to produce MNs from human pluripotent stem cells (hPSCs) for regenerative medicine/disease modelling applications. However, the in vitro generation of posterior MNs corresponding to the thoracic/lumbosacral spinal cord has been challenging. Although the induction of cells resembling neuromesodermal progenitors (NMPs), the bona fide precursors of the spinal cord, offers a promising solution, the progressive specification of posterior MNs from these cells is not well defined. Here, we determine the signals guiding the transition of human NMP-like cells toward thoracic ventral spinal cord neurectoderm. We show that combined WNT-FGF activities drive a posterior dorsal pre-/early neural state, whereas suppression of TGFß-BMP signalling pathways promotes a ventral identity and neural commitment. Based on these results, we define an optimised protocol for the generation of thoracic MNs that can efficiently integrate within the neural tube of chick embryos. We expect that our findings will facilitate the comparison of hPSC-derived spinal cord cells of distinct axial identities.


Subject(s)
Cell Differentiation/genetics , Mesoderm/growth & development , Neural Stem Cells/metabolism , Spinal Cord/growth & development , Animals , Body Patterning/genetics , Bone Morphogenetic Proteins/genetics , Cell Lineage/genetics , Chick Embryo , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Mesoderm/metabolism , Motor Neurons/metabolism , Neural Stem Cells/cytology , Pluripotent Stem Cells/cytology , Signal Transduction/genetics , Spinal Cord/metabolism , Transforming Growth Factor beta/genetics , Wnt Proteins/genetics
16.
Stem Cells Dev ; 30(11): 578-586, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33757297

ABSTRACT

Copy number variants (CNVs) are genomic rearrangements implicated in numerous congenital and acquired diseases, including cancer. The appearance of culture-acquired CNVs in human pluripotent stem cells (PSCs) has prompted concerns for their use in regenerative medicine. A particular problem in PSC is the frequent occurrence of CNVs in the q11.21 region of chromosome 20. However, the exact mechanism of origin of this amplicon remains elusive due to the difficulty in delineating its sequence and breakpoints. Here, we have addressed this problem using long-read Nanopore sequencing of two examples of this CNV, present as duplication and as triplication. In both cases, the CNVs were arranged in a head-to-tail orientation, with microhomology sequences flanking or overlapping the proximal and distal breakpoints. These breakpoint signatures point to a mechanism of microhomology-mediated break-induced replication in CNV formation, with surrounding Alu sequences likely contributing to the instability of this genomic region.


Subject(s)
Nanopore Sequencing , Pluripotent Stem Cells , Chromosomes , DNA Copy Number Variations/genetics , DNA Repair , Humans
17.
Nat Commun ; 11(1): 4989, 2020 10 05.
Article in English | MEDLINE | ID: mdl-33020476

ABSTRACT

We postulate that exit from pluripotency involves intermediates that retain pluripotency while simultaneously exhibiting lineage-bias. Using a MIXL1 reporter, we explore mesoderm lineage-bias within the human pluripotent stem cell compartment. We identify a substate, which at the single cell level coexpresses pluripotent and mesodermal gene expression programmes. Functionally these cells initiate stem cell cultures and exhibit mesodermal bias in differentiation assays. By promoting mesodermal identity through manipulation of WNT signalling while preventing exit from pluripotency using lysophosphatidic acid, we 'trap' and maintain cells in a lineage-biased stem cell state through multiple passages. These cells correspond to a normal state on the differentiation trajectory, the plasticity of which is evidenced by their reacquisition of an unbiased state upon removal of differentiation cues. The use of 'cross-antagonistic' signalling to trap pluripotent stem cell intermediates with different lineage-bias may have general applicability in the efficient production of cells for regenerative medicine.


Subject(s)
Cellular Reprogramming , Mesoderm/metabolism , Pluripotent Stem Cells/cytology , Animals , Cell Differentiation/genetics , Cell Line , Cell Lineage , Cell Plasticity/genetics , Cell Self Renewal , Culture Media , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Mice , Pluripotent Stem Cells/metabolism , Signal Transduction/genetics
18.
Nat Rev Mol Cell Biol ; 21(12): 715-728, 2020 12.
Article in English | MEDLINE | ID: mdl-32968234

ABSTRACT

In the 20 years since human embryonic stem cells, and subsequently induced pluripotent stem cells, were first described, it has become apparent that during long-term culture these cells (collectively referred to as 'pluripotent stem cells' (PSCs)) can acquire genetic changes, which commonly include gains or losses of particular chromosomal regions, or mutations in certain cancer-associated genes, especially TP53. Such changes raise concerns for the safety of PSC-derived cellular therapies for regenerative medicine. Although acquired genetic changes may not be present in a cell line at the start of a research programme, the low sensitivity of current detection methods means that mutations may be difficult to detect if they arise but are present in only a small proportion of the cells. In this Review, we discuss the types of mutations acquired by human PSCs and the mechanisms that lead to their accumulation. Recent work suggests that the underlying mutation rate in PSCs is low, although they also seem to be particularly susceptible to genomic damage. This apparent contradiction can be reconciled by the observations that, in contrast to somatic cells, PSCs are programmed to die in response to genomic damage, which may reflect the requirements of early embryogenesis. Thus, the common genetic variants that are observed are probably rare events that give the cells with a selective growth advantage.


Subject(s)
Clonal Evolution/genetics , Mutation Accumulation , Pluripotent Stem Cells/metabolism , Cell Culture Techniques/methods , Cell Culture Techniques/standards , Cell Differentiation/genetics , Cell- and Tissue-Based Therapy/methods , Cell- and Tissue-Based Therapy/trends , Cells, Cultured , Clonal Evolution/physiology , Human Embryonic Stem Cells/physiology , Humans , Mutation/physiology , Pluripotent Stem Cells/physiology
19.
Stem Cell Reports ; 15(3): 557-565, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32857978

ABSTRACT

The enteric nervous system (ENS) is derived primarily from the vagal neural crest, a migratory multipotent cell population emerging from the dorsal neural tube between somites 1 and 7. Defects in the development and function of the ENS cause a range of enteric neuropathies, including Hirschsprung disease. Little is known about the signals that specify early ENS progenitors, limiting progress in the generation of enteric neurons from human pluripotent stem cells (hPSCs) to provide tools for disease modeling and regenerative medicine for enteric neuropathies. We describe the efficient and accelerated generation of ENS progenitors from hPSCs, revealing that retinoic acid is critical for the acquisition of vagal axial identity and early ENS progenitor specification. These ENS progenitors generate enteric neurons in vitro and, following in vivo transplantation, achieved long-term colonization of the ENS in adult mice. Thus, hPSC-derived ENS progenitors may provide the basis for cell therapy for defects in the ENS.


Subject(s)
Enteric Nervous System/cytology , Neural Crest/cytology , Neural Stem Cells/cytology , Tretinoin/pharmacology , Animals , Cell Line , Humans , Mice , Neural Stem Cells/drug effects , Neurons/cytology , Neurons/drug effects , Signal Transduction/drug effects , Time Factors , Vagus Nerve/cytology
20.
Stem Cell Reports ; 14(6): 1009-1017, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32413278

ABSTRACT

Human pluripotent stem cells (PSCs) are subject to the appearance of recurrent genetic variants on prolonged culture. We have now found that, compared with isogenic differentiated cells, PSCs exhibit evidence of considerably more DNA damage during the S phase of the cell cycle, apparently as a consequence of DNA replication stress marked by slower progression of DNA replication, activation of latent origins of replication, and collapse of replication forks. As in many cancers, which, like PSCs, exhibit a shortened G1 phase and DNA replication stress, the resulting DNA damage may underlie the higher incidence of abnormal and abortive mitoses in PSCs, resulting in chromosomal non-dysjunction or cell death. However, we have found that the extent of DNA replication stress, DNA damage, and consequent aberrant mitoses can be substantially reduced by culturing PSCs in the presence of exogenous nucleosides, resulting in improved survival, clonogenicity, and population growth.


Subject(s)
DNA Replication , Genomic Instability , Nucleosides/pharmacology , Pluripotent Stem Cells/drug effects , Cell Line , Culture Media/chemistry , Culture Media/pharmacology , Cytoprotection , DNA Damage , Humans , Mitosis , Nucleosides/analysis , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...