Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Rev Mol Cell Biol ; 17(3): 170-82, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26818440

ABSTRACT

Experimental modelling of human disorders enables the definition of the cellular and molecular mechanisms underlying diseases and the development of therapies for treating them. The availability of human pluripotent stem cells (PSCs), which are capable of self-renewal and have the potential to differentiate into virtually any cell type, can now help to overcome the limitations of animal models for certain disorders. The ability to model human diseases using cultured PSCs has revolutionized the ways in which we study monogenic, complex and epigenetic disorders, as well as early- and late-onset diseases. Several strategies are used to generate such disease models using either embryonic stem cells (ES cells) or patient-specific induced PSCs (iPSCs), creating new possibilities for the establishment of models and their use in drug screening.


Subject(s)
Genetic Diseases, Inborn , Human Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Stem Cell Transplantation/methods , Allografts , Animals , Autografts , Disease Models, Animal , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/therapy , Humans
2.
Cell ; 153(5): 939-40, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23706730

ABSTRACT

Induction of pluripotency in somatic cells has been achieved by myriad combinations of transcription factors that belong to the core pluripotency circuitry. In this issue, Shu et al. report reprogramming with lineage specifiers, lending support to the view of the pluripotent state as a fine balance between competing differentiation forces.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Transcription Factors/metabolism , Animals
4.
Hum Mol Genet ; 31(17): 2899-2917, 2022 08 25.
Article in English | MEDLINE | ID: mdl-35394024

ABSTRACT

Cellular proliferation depends on the accurate and timely replication of the genome. Several genetic diseases are caused by mutations in key DNA replication genes; however, it remains unclear whether these genes influence the normal program of DNA replication timing. Similarly, the factors that regulate DNA replication dynamics are poorly understood. To systematically identify trans-acting modulators of replication timing, we profiled replication in 184 cell lines from three cell types, encompassing 60 different gene knockouts or genetic diseases. Through a rigorous approach that considers the background variability of replication timing, we concluded that most samples displayed normal replication timing. However, mutations in two genes showed consistently abnormal replication timing. The first gene was RIF1, a known modulator of replication timing. The second was MCM10, a highly conserved member of the pre-replication complex. Cells from a single patient carrying MCM10 mutations demonstrated replication timing variability comprising 46% of the genome and at different locations than RIF1 knockouts. Replication timing alterations in the mutated MCM10 cells were predominantly comprised of replication delays and initiation site gains and losses. Taken together, this study demonstrates the remarkable robustness of the human replication timing program and reveals MCM10 as a novel candidate modulator of DNA replication timing.


Subject(s)
DNA Replication Timing , Minichromosome Maintenance Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , DNA Replication/genetics , DNA Replication Timing/genetics , Humans , Minichromosome Maintenance Proteins/genetics , Replication Origin
5.
Genome Res ; 31(12): 2155-2169, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34810218

ABSTRACT

Haploid human embryonic stem cells (ESCs) provide a powerful genetic system but diploidize at high rates. We hypothesized that diploidization results from aberrant DNA replication. To test this, we profiled DNA replication timing in isogenic haploid and diploid ESCs. The greatest difference was the earlier replication of the X Chromosome in haploids, consistent with the lack of X-Chromosome inactivation. We also identified 21 autosomal regions that had delayed replication in haploids, extending beyond the normal S phase and into G2/M. Haploid-delays comprised a unique set of quiescent genomic regions that are also underreplicated in polyploid placental cells. The same delays were observed in female ESCs with two active X Chromosomes, suggesting that increased X-Chromosome dosage may cause delayed autosomal replication. We propose that incomplete replication at the onset of mitosis could prevent cell division and result in re-entry into the cell cycle and whole genome duplication.

6.
EMBO J ; 38(12)2019 06 17.
Article in English | MEDLINE | ID: mdl-31088843

ABSTRACT

Human pluripotent stem cells (hPSCs) are being increasingly utilized worldwide in investigating human development, and modeling and discovering therapies for a wide range of diseases as well as a source for cellular therapy. Yet, since the first isolation of human embryonic stem cells (hESCs) 20 years ago, followed by the successful reprogramming of human-induced pluripotent stem cells (hiPSCs) 10 years later, various studies shed light on abnormalities that sometimes accumulate in these cells in vitro Whereas genetic aberrations are well documented, epigenetic alterations are not as thoroughly discussed. In this review, we highlight frequent epigenetic aberrations found in hPSCs, including alterations in DNA methylation patterns, parental imprinting, and X chromosome inactivation. We discuss the potential origins of these abnormalities in hESCs and hiPSCs, survey the different methods for detecting them, and elaborate on their potential consequences for the different utilities of hPSCs.


Subject(s)
Epigenesis, Genetic/physiology , Pluripotent Stem Cells/physiology , Cell Differentiation/genetics , Cellular Reprogramming/genetics , DNA Methylation/physiology , Genomic Imprinting/genetics , Humans , Induced Pluripotent Stem Cells/physiology , X Chromosome Inactivation/physiology
7.
Exp Eye Res ; 233: 109542, 2023 08.
Article in English | MEDLINE | ID: mdl-37331647

ABSTRACT

Retinoblastoma (Rb) is a rare malignant disorder affecting the developing retina of children under the age of five. Chemotherapeutic agents used for treating Rb have been associated with defects of the retinal pigment epithelium (RPE), such as hyperplasia, gliosis, and mottling. Herein, we have developed two pluripotent stem cell (PSC)-RPE models to assess the cytotoxicity of known Rb chemotherapeutics such as Melphalan, Topotecan and TW-37. Our findings demonstrate that these drugs alter the RPE by decreasing the monolayer barrier's trans-epithelial resistance and affecting the cells' phagocytic activity. Transcriptional analyses demonstrate an altered expression of genes involved in melanin and retinol processing, tight junction and apical-basal polarity pathways in both models. When applied within the clinical range, none of the drug treatments caused significant cytotoxic effects, changes to the apical-basal polarity, tight junction network or cell cycle. Together, our results demonstrate that although the most commonly used Rb chemotherapeutic drugs do not cause cytotoxicity in RPE, their application in vitro leads to compromised phagocytosis and strength of the barrier function, in addition to changes in gene expression that could alter the visual cycle in vivo. Our data demonstrate that widely used Rb chemotherapeutic drugs can have a deleterious impact on RPE cells and thus great care has to be exercised with regard to their delivery so the adjacent healthy RPE is not damaged during the course of tumor eradication.


Subject(s)
Retinal Neoplasms , Retinoblastoma , Child , Humans , Retinal Pigment Epithelium/metabolism , Retinoblastoma/drug therapy , Retinoblastoma/genetics , Retinoblastoma/metabolism , Retina , Retinal Neoplasms/drug therapy , Retinal Neoplasms/genetics , Retinal Neoplasms/metabolism , Gene Expression , Cell Differentiation
8.
Nature ; 545(7653): 229-233, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28445466

ABSTRACT

Human pluripotent stem cells (hPS cells) can self-renew indefinitely, making them an attractive source for regenerative therapies. This expansion potential has been linked with the acquisition of large copy number variants that provide mutated cells with a growth advantage in culture. The nature, extent and functional effects of other acquired genome sequence mutations in cultured hPS cells are not known. Here we sequence the protein-coding genes (exomes) of 140 independent human embryonic stem cell (hES cell) lines, including 26 lines prepared for potential clinical use. We then apply computational strategies for identifying mutations present in a subset of cells in each hES cell line. Although such mosaic mutations were generally rare, we identified five unrelated hES cell lines that carried six mutations in the TP53 gene that encodes the tumour suppressor P53. The TP53 mutations we observed are dominant negative and are the mutations most commonly seen in human cancers. We found that the TP53 mutant allelic fraction increased with passage number under standard culture conditions, suggesting that the P53 mutations confer selective advantage. We then mined published RNA sequencing data from 117 hPS cell lines, and observed another nine TP53 mutations, all resulting in coding changes in the DNA-binding domain of P53. In three lines, the allelic fraction exceeded 50%, suggesting additional selective advantage resulting from the loss of heterozygosity at the TP53 locus. As the acquisition and expansion of cancer-associated mutations in hPS cells may go unnoticed during most applications, we suggest that careful genetic characterization of hPS cells and their differentiated derivatives be carried out before clinical use.


Subject(s)
Genes, Dominant/genetics , Genes, p53 , Human Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mutation/genetics , Selection, Genetic , Tumor Suppressor Protein p53/genetics , Alleles , Cell Count , Cell Differentiation/genetics , Cell Division/genetics , Cell Line , DNA/metabolism , DNA Mutational Analysis , Exome/genetics , Human Embryonic Stem Cells/cytology , Humans , Loss of Heterozygosity/genetics , Mosaicism , Neoplasms/genetics , Protein Domains , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism
9.
Nature ; 532(7597): 107-11, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26982723

ABSTRACT

Diploidy is a fundamental genetic feature in mammals, in which haploid cells normally arise only as post-meiotic germ cells that serve to ensure a diploid genome upon fertilization. Gamete manipulation has yielded haploid embryonic stem (ES) cells from several mammalian species, but haploid human ES cells have yet to be reported. Here we generated and analysed a collection of human parthenogenetic ES cell lines originating from haploid oocytes, leading to the successful isolation and maintenance of human ES cell lines with a normal haploid karyotype. Haploid human ES cells exhibited typical pluripotent stem cell characteristics, such as self-renewal capacity and a pluripotency-specific molecular signature. Moreover, we demonstrated the utility of these cells as a platform for loss-of-function genetic screening. Although haploid human ES cells resembled their diploid counterparts, they also displayed distinct properties including differential regulation of X chromosome inactivation and of genes involved in oxidative phosphorylation, alongside reduction in absolute gene expression levels and cell size. Surprisingly, we found that a haploid human genome is compatible not only with the undifferentiated pluripotent state, but also with differentiated somatic fates representing all three embryonic germ layers both in vitro and in vivo, despite a persistent dosage imbalance between the autosomes and X chromosome. We expect that haploid human ES cells will provide novel means for studying human functional genomics and development.


Subject(s)
Cell Differentiation , Genetic Association Studies/methods , Haploidy , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Cell Self Renewal , Cell Separation , Cell Size , Chromosomes, Human, X/genetics , Diploidy , Down-Regulation/genetics , Gene Deletion , Germ Layers/cytology , Humans , Karyotyping , Oocytes/metabolism , Oxidative Phosphorylation , Parthenogenesis , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , X Chromosome Inactivation/genetics
10.
Nature ; 525(7570): 469-78, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26399828

ABSTRACT

Stem cells self-renew and generate specialized progeny through differentiation, but vary in the range of cells and tissues they generate, a property called developmental potency. Pluripotent stem cells produce all cells of an organism, while multipotent or unipotent stem cells regenerate only specific lineages or tissues. Defining stem-cell potency relies upon functional assays and diagnostic transcriptional, epigenetic and metabolic states. Here we describe functional and molecular hallmarks of pluripotent stem cells, propose a checklist for their evaluation, and illustrate how forensic genomics can validate their provenance.


Subject(s)
Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genomics , Humans
12.
Stem Cells ; 37(9): 1130-1135, 2019 09.
Article in English | MEDLINE | ID: mdl-31021472

ABSTRACT

This report summarizes the recent activity of the International Stem Cell Banking Initiative held at Harvard Stem Cell Institute, Boston, MA, USA, on June 18, 2017. In this meeting, we aimed to find consensus on ongoing issues of quality control (QC), safety, and efficacy of human pluripotent stem cell banks and their derivative cell therapy products for the global harmonization. In particular, assays for the QC testing such as pluripotency assays test and general QC testing criteria were intensively discussed. Moreover, the recent activities of global stem cell banking centers and the regulatory bodies were briefly summarized to provide an overview on global developments and issues. Stem Cells 2019;37:1130-1135.


Subject(s)
Pluripotent Stem Cells/cytology , Stem Cells/cytology , Tissue Banks/standards , Boston , Cell- and Tissue-Based Therapy/methods , Humans , Induced Pluripotent Stem Cells/cytology , International Cooperation , Quality Control
13.
Nature ; 510(7506): 533-6, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24776804

ABSTRACT

The transfer of somatic cell nuclei into oocytes can give rise to pluripotent stem cells that are consistently equivalent to embryonic stem cells, holding promise for autologous cell replacement therapy. Although methods to induce pluripotent stem cells from somatic cells by transcription factors are widely used in basic research, numerous differences between induced pluripotent stem cells and embryonic stem cells have been reported, potentially affecting their clinical use. Because of the therapeutic potential of diploid embryonic stem-cell lines derived from adult cells of diseased human subjects, we have systematically investigated the parameters affecting efficiency of blastocyst development and stem-cell derivation. Here we show that improvements to the oocyte activation protocol, including the use of both kinase and translation inhibitors, and cell culture in the presence of histone deacetylase inhibitors, promote development to the blastocyst stage. Developmental efficiency varied between oocyte donors, and was inversely related to the number of days of hormonal stimulation required for oocyte maturation, whereas the daily dose of gonadotropin or the total number of metaphase II oocytes retrieved did not affect developmental outcome. Because the use of concentrated Sendai virus for cell fusion induced an increase in intracellular calcium concentration, causing premature oocyte activation, we used diluted Sendai virus in calcium-free medium. Using this modified nuclear transfer protocol, we derived diploid pluripotent stem-cell lines from somatic cells of a newborn and, for the first time, an adult, a female with type 1 diabetes.


Subject(s)
Cell Nucleus/genetics , Cellular Reprogramming , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Diploidy , Oocytes/cytology , Pluripotent Stem Cells/cytology , Adult , Blastocyst/drug effects , Cell Fusion , Chromosomes, Mammalian/metabolism , Female , Histone Deacetylase Inhibitors/pharmacology , Humans , Infant, Newborn , Metaphase , Oocytes/metabolism , Oogenesis , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/pathology , Sendai virus , Spindle Apparatus/metabolism
14.
PLoS Genet ; 13(8): e1006979, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28837588

ABSTRACT

Human pluripotent stem cells (hPSCs) are an important player in disease modeling and regenerative medicine. Nonetheless, multiple studies uncovered their inherent genetic instability upon prolonged culturing, where specific chromosomal aberrations provide cells with a growth advantage. These positively selected modifications have dramatic effects on multiple cellular characteristics. Epigenetic aberrations also possess the potential of changing gene expression and altering cellular functions. In the current study we assessed the landscape of DNA methylation aberrations during prolonged culturing of hPSCs, and defined a set of genes which are recurrently hypermethylated and silenced. We further focused on one of these genes, testis-specific Y-encoded like protein 5 (TSPYL5), and demonstrated that when silenced, differentiation-related genes and tumor-suppressor genes are downregulated, while pluripotency- and growth promoting genes are upregulated. This process is similar to the hypermethylation-mediated inactivation of certain genes during tumor development. Our analysis highlights the existence and importance of recurrent epigenetic aberrations in hPSCs during prolonged culturing.


Subject(s)
Chromosome Aberrations , Epigenesis, Genetic/genetics , Genomic Instability/genetics , Pluripotent Stem Cells/cytology , Cell Culture Techniques , Cell Differentiation/genetics , DNA Methylation/genetics , Gene Expression Regulation/genetics , Humans , Regenerative Medicine
15.
Nature ; 540(7632): 211-212, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27919070

Subject(s)
Stem Cells , Humans
18.
Stem Cells ; 33(3): 1013-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25377277

ABSTRACT

Pluripotent-specific inhibitors (PluriSIns) make a powerful tool to study the mechanisms controlling the survival of human pluripotent stem cells (hPSCs). Here, we characterize the mechanism of action of PluriSIn#2, a compound that selectively eliminates undifferentiated hPSCs, while sparing various other cell types derived from them. Toxicogenomic analysis predicts this compound to be a topoisomerase inhibitor. Gene expression analyses reveal that one of the human topoisomerase enzymes, topoisomerase II alpha (TOP2A), is uniquely expressed in hPSCs: TOP2A is highly expressed in undifferentiated cells, is downregulated during their differentiation, and its expression depends on the expression of core pluripotency transcription factors. Furthermore, siRNA-based knockdown of TOP2A in undifferentiated hPSCs results in their cell death, revealing that TOP2A expression is required for the survival of these cells. We find that PluriSIn#2 does not directly inhibit TOP2A enzymatic activity, but rather selectively represses its transcription, thereby significantly reducing TOP2A protein levels. As undifferentiated hPSCs require TOP2A activity for their survival, TOP2A inhibition by PluriSIn#2 thus causes their cell death. Therefore, TOP2A dependency can be harnessed for the selective elimination of tumorigenic hPSCs from culture.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Topoisomerase Inhibitors/pharmacology , Antigens, Neoplasm/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/enzymology , Humans , Pluripotent Stem Cells/enzymology , Poly-ADP-Ribose Binding Proteins , Small Molecule Libraries/pharmacology
19.
Nat Biotechnol ; 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38195986

ABSTRACT

To survey cancer-related mutations in human pluripotent stem cells and their derivatives, we analyzed >2,200 transcriptomes from 146 independent lines in the NCBI's Sequence Read Archive. Twenty-two per cent of samples had at least one cancer-related mutation; of these, 64% had TP53 mutations, which conferred a pronounced selective advantage, perturbed target gene expression and altered cellular differentiation. These findings underscore the need for robust surveillance of cancer-related mutations in pluripotent cells, especially in clinical applications.

20.
Stem Cells ; 30(4): 612-22, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22328490

ABSTRACT

Mouse pluripotent stem cells (PSCs) are the best studied pluripotent system and are regarded as the "gold standard" to which human PSCs are compared. However, while the genomic integrity of human PSCs has recently drawn much attention, mouse PSCs have not been systematically evaluated in this regard. The genomic stability of PSCs is a matter of profound significance, as it affects their pluripotency, differentiation, and tumorigenicity. We thus performed a thorough analysis of the genomic integrity of 325 samples of mouse PSCs, including 127 induced pluripotent stem cell (iPSC) samples. We found that genomic aberrations occur frequently in mouse embryonic stem cells of various mouse strains, add in mouse iPSCs of various cell origins and derivation techniques. Four hotspots of chromosomal aberrations were detected: full trisomy 11 (with a minimally recurrent gain in 11qE2), full trisomy 8, and deletions in chromosomes 10qB and 14qC-14qE. The most recurrent aberration in mouse PSCs, gain 11qE2, turned out to be fully syntenic to the common aberration 17q25 in human PSCs, while other recurrent aberrations were found to be species specific. Analysis of chromosomal aberrations in 74 samples of rhesus macaque PSCs revealed a gain in chromosome 16q, syntenic to the hotspot in human 17q. Importantly, these common aberrations jeopardize the interpretation of published comparisons of PSCs, which were unintentionally conducted between normal and aberrant cells. Therefore, this work emphasizes the need to carefully monitor genomic integrity of PSCs from all species, for their proper use in biomedical research.


Subject(s)
Chromosome Aberrations , Conserved Sequence/genetics , Evolution, Molecular , Genome/genetics , Pluripotent Stem Cells/metabolism , Animals , Gene Expression Regulation , Humans , Macaca mulatta/genetics , Mice , Species Specificity , Synteny/genetics
SELECTION OF CITATIONS
SEARCH DETAIL