Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters








Publication year range
1.
Stem Cell Res ; 81: 103532, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39276527

ABSTRACT

Induced pluripotent stem cells (iPSCs) harboring patient derived SAMD9 mutation offer a unique platform to study the multi-organ involvement observed in this rare disease, referred to as myelodysplasia, infections, restriction of growth, adrenal hypoplasia, genital phenotypes, and enteropathy (MIRAGE) syndrome. The pluripotent nature of iPSCs allows in vitro differentiation into various somatic cell types representing multiple organ systems affected in SAMD9-mutated patients. Hence, in this paper, we present a CRISPR/Cas9-engineered iPSC model carrying SAMD9 c.2948T>G, p.I983S mutation previously reported in two patients with severe MIRAGE syndrome.

2.
Cell Rep ; 42(8): 112868, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37494188

ABSTRACT

Cells maintain and dynamically change their proteomes according to the environment and their needs. Mechanistic target of rapamycin (mTOR) is a key regulator of proteostasis, homeostasis of the proteome. Thus, dysregulation of mTOR leads to changes in proteostasis and the consequent progression of diseases, including cancer. Based on the physiological and clinical importance of mTOR signaling, we investigated mTOR feedback signaling, proteostasis, and cell fate. Here, we reveal that mTOR targeting inhibits eIF4E-mediated cap-dependent translation, but feedback signaling activates a translation initiation factor, eukaryotic translation initiation factor 3D (eIF3D), to sustain alternative non-canonical translation mechanisms. Importantly, eIF3D-mediated protein synthesis enables cell phenotype switching from proliferative to more migratory. eIF3D cooperates with mRNA-binding proteins such as heterogeneous nuclear ribonucleoprotein F (hnRNPF), heterogeneous nuclear ribonucleoprotein K (hnRNPK), and Sjogren syndrome antigen B (SSB) to support selective mRNA translation following mTOR inhibition, which upregulates and activates proteins involved in insulin receptor (INSR)/insulin-like growth factor 1 receptor (IGF1R)/insulin receptor substrate (IRS) and interleukin 6 signal transducer (IL-6ST)/Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling. Our study highlights the mechanisms by which cells establish the dynamic change of proteostasis and the resulting phenotype switch.


Subject(s)
Proteostasis , Receptor, Insulin , RNA, Messenger/metabolism , Receptor, Insulin/metabolism , TOR Serine-Threonine Kinases/metabolism , Sirolimus , Protein Biosynthesis
3.
Front Psychiatry ; 13: 924956, 2022.
Article in English | MEDLINE | ID: mdl-36405918

ABSTRACT

16p13.11 copy number variants (CNVs) have been associated with autism, schizophrenia, psychosis, intellectual disability, and epilepsy. The majority of 16p13.11 deletions or duplications occur within three well-defined intervals, and despite growing knowledge of the functions of individual genes within these intervals, the molecular mechanisms that underlie commonly observed clinical phenotypes remain largely unknown. Patient-derived, induced pluripotent stem cells (iPSCs) provide a platform for investigating the morphological, electrophysiological, and gene-expression changes that result from 16p13.11 CNVs in human-derived neurons. Patient derived iPSCs with varying sizes of 16p13.11 deletions and familial controls were differentiated into cortical neurons for phenotypic analysis. High-content imaging and morphological analysis of patient-derived neurons demonstrated an increase in neurite branching in patients compared with controls. Whole-transcriptome sequencing revealed expression level changes in neuron development and synaptic-related gene families, suggesting a defect in synapse formation. Subsequent quantification of synapse number demonstrated increased numbers of synapses on neurons derived from early-onset patients compared to controls. The identification of common phenotypes among neurons derived from patients with overlapping 16p13.11 deletions will further assist in ascertaining common pathways and targets that could be utilized for screening drug candidates. These studies can help to improve future treatment options and clinical outcomes for 16p13.11 deletion patients.

4.
Stem Cell Res ; 46: 101836, 2020 07.
Article in English | MEDLINE | ID: mdl-32485644

ABSTRACT

Sialidosis is an autosomal recessive lysosomal storage disease, belonging to the glycoproteinoses. The disease is caused by deficiency of the sialic acid-cleaving enzyme, sialidase 1 or neuraminidase 1 (NEU1). Patients with sialidosis are classified based on the age of onset and severity of the clinical symptoms into type I (normomorphic) and type II (dysmorphic). Patient-derived skin fibroblasts from both disease types were reprogrammed using the CytoTune™-iPS 2.0 Sendai Reprogramming Kit. iPSCs were characterized for pluripotency, three germ-layer differentiation, normal karyotype and absence of viral components. These cell lines represent a valuable resource to model sialidosis and to screen for therapeutics.


Subject(s)
Induced Pluripotent Stem Cells , Mucolipidoses , Cell Differentiation , Fibroblasts , Humans , Mucolipidoses/genetics , Mutation , Neuraminidase/genetics
5.
Biochem Biophys Rep ; 20: 100656, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31467990

ABSTRACT

Naïve pluripotent stem cells (PSCs) display a distinctive phenotype when compared to their "primed" counterparts, including, but not limited to, increased potency to differentiate and more robust mitochondrial respiration. The cultivation and maintenance of naïve PSCs have been notoriously challenging, requiring the use of complex cytokine cocktails. NME7AB is a newly discovered embryonic stem cell growth factor that is expressed exclusively in the first few days of human blastocyst development. It has been previously reported that growing primed induced PSCs (iPSCs) in bFGF-depleted medium with NME7AB as the only added growth factor facilitates the regression of these cells to their naïve state. Here, we confirm this regression by demonstrating the reactivation of mitochondrial function in the induced naïve-like PSCs and increased ATP production in these cells, as compared to that in primed iPSCs.

6.
Nat Commun ; 10(1): 3623, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31399583

ABSTRACT

Coordinated regulation of the lysosomal and autophagic systems ensures basal catabolism and normal cell physiology, and failure of either system causes disease. Here we describe an epigenetic rheostat orchestrated by c-MYC and histone deacetylases that inhibits lysosomal and autophagic biogenesis by concomitantly repressing the expression of the transcription factors MiT/TFE and FOXH1, and that of lysosomal and autophagy genes. Inhibition of histone deacetylases abates c-MYC binding to the promoters of lysosomal and autophagy genes, granting promoter occupancy to the MiT/TFE members, TFEB and TFE3, and/or the autophagy regulator FOXH1. In pluripotent stem cells and cancer, suppression of lysosomal and autophagic function is directly downstream of c-MYC overexpression and may represent a hallmark of malignant transformation. We propose that, by determining the fate of these catabolic systems, this hierarchical switch regulates the adaptive response of cells to pathological and physiological cues that could be exploited therapeutically.


Subject(s)
Autophagy/physiology , Epigenesis, Genetic , Lysosomes/metabolism , Organelle Biogenesis , Polytetrafluoroethylene/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Autophagy/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Binding Sites , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Histone Deacetylase 2/metabolism , Histone Deacetylases/metabolism , Humans , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , Stem Cells , Transcription, Genetic
8.
Exp Cell Res ; 379(1): 55-64, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30922922

ABSTRACT

Metabolic studies of human pluripotent stem cells (hPSCs) have focused on how the cells produce energy through the catabolic pathway. The less-studied anabolic pathway, by which hPSCs expend energy in the form of adenosine triphosphate (ATP), is not yet fully understood. Compared to fully differentiated somatic cells, hPSCs undergo significant changes not only in their gene expression but also in their production and/or expenditure of ATP. Here, we investigate how hPSCs tightly control their energy homeostasis by studying the main energy-consuming process, mRNA translation. In addition, change of subcellular organelles regarding energy homeostasis has been investigated. Lysosomes are organelles that play an important role in the elimination of unnecessary cellular materials by digestion and in the recycling system of the cell. We have found that hPSCs control their lysosome numbers in part by regulating lysosomal gene/protein expression. Thus, because the levels of mRNA translation rate are lower in hPSCs than in somatic cells, not only the global translational machinery but also the lysosomal recycling machinery is suppressed in hPSCs. Overall, the results of our study suggest that hPSCs reprogram gene expression and signaling to regulate energy-consuming processes and energy-controlling organelles.


Subject(s)
Energy Metabolism/physiology , Organelles/metabolism , Pluripotent Stem Cells/metabolism , Adenosine Triphosphate/metabolism , Cell Differentiation/physiology , Cells, Cultured , Gene Expression/physiology , Homeostasis/physiology , Humans , Lysosomes/metabolism , Protein Biosynthesis/physiology , RNA, Messenger/metabolism , Signal Transduction/physiology
9.
Proc Natl Acad Sci U S A ; 116(8): 2967-2976, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30728292

ABSTRACT

ERK is a key coordinator of the epithelial-to-mesenchymal transition (EMT) in that a variety of EMT-inducing factors activate signaling pathways that converge on ERK to regulate EMT transcription programs. However, the mechanisms by which ERK controls the EMT program are not well understood. Through an analysis of the global changes of gene expression mediated by ERK2, we identified the transcription factor FoxO1 as a potential mediator of ERK2-induced EMT, and thus we investigated the mechanism by which ERK2 regulates FoxO1. Additionally, our analysis revealed that ERK2 induced the expression of Dock10, a Rac1/Cdc42 GEF, during EMT. We demonstrate that the activation of the Rac1/JNK signaling axis downstream of Dock10 leads to an increase in FoxO1 expression and EMT. Taken together, our study uncovers mechanisms by which epithelial cells acquire less proliferative but more migratory mesenchymal properties and reveals potential therapeutic targets for cancers evolving into a metastatic disease state.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Forkhead Box Protein O1/genetics , Guanine Nucleotide Exchange Factors/genetics , Mitogen-Activated Protein Kinase 1/genetics , Cell Line, Tumor , Gene Expression Regulation/genetics , Humans , MAP Kinase Signaling System/genetics , Transcriptional Activation/genetics , rac1 GTP-Binding Protein/genetics
10.
BMB Rep ; 52(5): 324-329, 2019 May.
Article in English | MEDLINE | ID: mdl-30293549

ABSTRACT

Recent progress in cellular reprogramming technology and lineage-specific cell differentiation has provided great opportunities for translational research. Because virus-based gene delivery is not a practical reprogramming protocol, protein-based reprogramming has been receiving attention as a safe way to generate reprogrammed cells. However, the poor efficiency of the cellular uptake of reprogramming proteins is still a major obstacle. Here, we reported key factors which improve the cellular uptake of these proteins. Purified red fluorescent proteins fused with 9xLysine (dsRED-9K) as a cell penetrating peptide were efficiently delivered into the diverse primary cells. Protein delivery was improved by the addition of amodiaquine. Furthermore, purified dsRED-9K was able to penetrate all cell lineages derived from mouse embryonic stem cells efficiently. Our data may provide important insights into the design of protein-based reprogramming or differentiation protocols [BMB Reports 2019; 52(5): 324-329].


Subject(s)
Cell-Penetrating Peptides/metabolism , Cellular Reprogramming Techniques/methods , Polylysine/metabolism , Amodiaquine/pharmacology , Animals , Cell Culture Techniques , Cell Differentiation/genetics , Cell-Penetrating Peptides/pharmacology , Cellular Reprogramming/genetics , Embryonic Stem Cells/cytology , Fibroblasts/metabolism , Gene Transfer Techniques , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Peptides/therapeutic use , Polylysine/therapeutic use , Transcription Factors/metabolism
11.
Mol Psychiatry ; 23(11): 2167-2183, 2018 11.
Article in English | MEDLINE | ID: mdl-29449635

ABSTRACT

Accumulating evidence suggests that cerebellar dysfunction early in life is associated with autism spectrum disorder (ASD), but the molecular mechanisms underlying the cerebellar deficits at the cellular level are unclear. Tuberous sclerosis complex (TSC) is a neurocutaneous disorder that often presents with ASD. Here, we developed a cerebellar Purkinje cell (PC) model of TSC with patient-derived human induced pluripotent stem cells (hiPSCs) to characterize the molecular mechanisms underlying cerebellar abnormalities in ASD and TSC. Our results show that hiPSC-derived PCs from patients with pathogenic TSC2 mutations displayed mTORC1 pathway hyperactivation, defects in neuronal differentiation and RNA regulation, hypoexcitability and reduced synaptic activity when compared with those derived from controls. Our gene expression analyses revealed downregulation of several components of fragile X mental retardation protein (FMRP) targets in TSC2-deficient hiPSC-PCs. We detected decreased expression of FMRP, glutamate receptor δ2 (GRID2), and pre- and post-synaptic markers such as synaptophysin and PSD95 in the TSC2-deficient hiPSC-PCs. The mTOR inhibitor rapamycin rescued the deficits in differentiation, synaptic dysfunction, and hypoexcitability of TSC2 mutant hiPSC-PCs in vitro. Our findings suggest that these gene expression changes and cellular abnormalities contribute to aberrant PC function during development in TSC affected individuals.


Subject(s)
Purkinje Cells/metabolism , Tuberous Sclerosis/metabolism , Adult , Autism Spectrum Disorder/complications , Autism Spectrum Disorder/metabolism , Cerebellar Diseases/metabolism , Cerebellum/metabolism , Child , Child, Preschool , Female , Fragile X Mental Retardation Protein/drug effects , Fragile X Mental Retardation Protein/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Models, Biological , Purkinje Cells/pathology , Sirolimus/pharmacology , Synapses/metabolism , Synapses/physiology , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis/physiopathology , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics
12.
Biochem Biophys Res Commun ; 492(2): 154-160, 2017 10 14.
Article in English | MEDLINE | ID: mdl-28802578

ABSTRACT

Induced pluripotent stem cells (iPSCs) technology is a method for generating pluripotent stem cells in vitro from fully differentiated cells such as fibroblast cells. The potential applications of iPSC technology in cell therapy and disease modeling could influence current medical practices. Despite current advances in iPSC technology, many patient-derived reprogrammed cells are not suitable for clinical trial because most protocols rely on virus-based techniques, which pose the risk of integration of the viral genome into the chromosomes. Therefore, non-viral methods such as mRNA and protein-based reprogramming are promising alternatives when generating clinically safe iPSCs. In a previous study, we generated human iPSCs using cell extracts with cell penetration peptide (CPP) for the delivery of reprogramming proteins [Kim et al. Cell Stem Cells, 2009]. In here, we show that the expression of reprogramming factors in mammalian cells and subsequent purification of these factors by FLAG-Tag could reprogram fibroblasts into iPSCs.


Subject(s)
Cellular Reprogramming Techniques/methods , Cellular Reprogramming , Fibroblasts/cytology , Induced Pluripotent Stem Cells/cytology , Cells, Cultured , Fibroblasts/metabolism , Gene Expression , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism
13.
BMB Rep ; 50(9): 435-436, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28683850

ABSTRACT

Primed human pluripotent stem cells (hPSCs) are highly dependent on glycolysis rather than oxidative phosphorylation, which is similar to the metabolic switch that occurs in cancer cells. However, the molecular mechanisms that underlie this metabolic reprogramming in hPSCs and its relevance to pluripotency remain unclear. Cha et al. (2017) recently revealed that downregulation of SIRT2 by miR-200c enhances acetylation of glycolytic enzymes and glycolysis, which in turn facilitates cellular reprogramming, suggesting that SIRT2 is a key enzyme linking the metabolic switch and pluripotency in hPSCs. [BMB Reports 2017; 50(9): 435-436].


Subject(s)
Pluripotent Stem Cells/metabolism , Sirtuin 2/metabolism , Acetylation , Cellular Reprogramming/genetics , Cellular Reprogramming/physiology , Glycolysis/genetics , Glycolysis/physiology , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Oxidative Phosphorylation , Pluripotent Stem Cells/cytology , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/physiology , Sirtuin 2/genetics
14.
Nat Cell Biol ; 19(5): 445-456, 2017 May.
Article in English | MEDLINE | ID: mdl-28436968

ABSTRACT

A hallmark of cancer cells is the metabolic switch from oxidative phosphorylation (OXPHOS) to glycolysis, a phenomenon referred to as the 'Warburg effect', which is also observed in primed human pluripotent stem cells (hPSCs). Here, we report that downregulation of SIRT2 and upregulation of SIRT1 is a molecular signature of primed hPSCs and that SIRT2 critically regulates metabolic reprogramming during induced pluripotency by targeting glycolytic enzymes including aldolase, glyceraldehyde-3-phosphate dehydrogenase, phosphoglycerate kinase, and enolase. Remarkably, knockdown of SIRT2 in human fibroblasts resulted in significantly decreased OXPHOS and increased glycolysis. In addition, we found that miR-200c-5p specifically targets SIRT2, downregulating its expression. Furthermore, SIRT2 overexpression in hPSCs significantly affected energy metabolism, altering stem cell functions such as pluripotent differentiation properties. Taken together, our results identify the miR-200c-SIRT2 axis as a key regulator of metabolic reprogramming (Warburg-like effect), via regulation of glycolytic enzymes, during human induced pluripotency and pluripotent stem cell function.


Subject(s)
Cell Differentiation , Energy Metabolism , MicroRNAs/metabolism , Pluripotent Stem Cells/enzymology , Sirtuin 2/metabolism , Acetylation , Cell Lineage , Cell Survival , Cellular Reprogramming , Computational Biology , Databases, Genetic , Gene Expression Regulation, Enzymologic , Glycolysis , HEK293 Cells , Humans , MicroRNAs/genetics , Oxidative Phosphorylation , Phenotype , Protein Processing, Post-Translational , Signal Transduction , Sirtuin 1/genetics , Sirtuin 1/metabolism , Sirtuin 2/genetics , Time Factors , Transfection
15.
J Exp Med ; 214(3): 681-697, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28183733

ABSTRACT

Disruption of myelination during development has been implicated in a range of neurodevelopmental disorders including tuberous sclerosis complex (TSC). TSC patients with autism display impairments in white matter integrity. Similarly, mice lacking neuronal Tsc1 have a hypomyelination phenotype. However, the mechanisms that underlie these phenotypes remain unknown. In this study, we demonstrate that neuronal TSC1/2 orchestrates a program of oligodendrocyte maturation through the regulated secretion of connective tissue growth factor (CTGF). We characterize oligodendrocyte maturation both in vitro and in vivo. We find that neuron-specific Tsc1 deletion results in an increase in CTGF secretion that non-cell autonomously stunts oligodendrocyte development and decreases the total number of oligodendrocytes. Genetic deletion of CTGF from neurons, in turn, mitigates the TSC-dependent hypomyelination phenotype. These results show that the mechanistic target of rapamycin (mTOR) pathway in neurons regulates CTGF production and secretion, revealing a paracrine mechanism by which neuronal signaling regulates oligodendrocyte maturation and myelination in TSC. This study highlights the role of mTOR-dependent signaling between neuronal and nonneuronal cells in the regulation of myelin and identifies an additional therapeutic avenue for this disease.


Subject(s)
Connective Tissue Growth Factor/physiology , Myelin Sheath/physiology , Neurons/physiology , Tuberous Sclerosis/physiopathology , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Oligodendroglia/physiology , Rats , TOR Serine-Threonine Kinases/physiology , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/physiology
17.
Cell Rep ; 17(4): 1053-1070, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27760312

ABSTRACT

Tuberous sclerosis complex (TSC) is a neurodevelopmental disease caused by TSC1 or TSC2 mutations and subsequent activation of the mTORC1 kinase. Upon mTORC1 activation, anabolic metabolism, which requires mitochondria, is induced, yet at the same time the principal pathway for mitochondrial turnover, autophagy, is compromised. How mTORC1 activation impacts mitochondrial turnover in neurons remains unknown. Here, we demonstrate impaired mitochondrial homeostasis in neuronal in vitro and in vivo models of TSC. We find that Tsc1/2-deficient neurons accumulate mitochondria in cell bodies, but are depleted of axonal mitochondria, including those supporting presynaptic sites. Axonal and global mitophagy of damaged mitochondria is impaired, suggesting that decreased turnover may act upstream of impaired mitochondrial metabolism. Importantly, blocking mTORC1 or inducing mTOR-independent autophagy restores mitochondrial homeostasis. Our study clarifies the complex relationship between the TSC-mTORC1 pathway, autophagy, and mitophagy, and defines mitochondrial homeostasis as a therapeutic target for TSC and related diseases.


Subject(s)
Mitochondrial Dynamics , Mitophagy , Models, Biological , Neurons/metabolism , Neurons/pathology , Tuberous Sclerosis/metabolism , Tuberous Sclerosis/pathology , Animals , Autophagy , Axons/metabolism , Cell Respiration , Humans , Lysosomes/metabolism , Membrane Potential, Mitochondrial , Mice , Mutation/genetics , Pluripotent Stem Cells/metabolism , Presynaptic Terminals/metabolism , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism
18.
Cold Spring Harb Mol Case Stud ; 2(5): a001008, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27626066

ABSTRACT

We describe a child with onset of command auditory hallucinations and behavioral regression at 6 yr of age in the context of longer standing selective mutism, aggression, and mild motor delays. His genetic evaluation included chromosomal microarray analysis and whole-exome sequencing. Sequencing revealed a previously unreported heterozygous de novo mutation c.385G>A in ATP1A3, predicted to result in a p.V129M amino acid change. This gene codes for a neuron-specific isoform of the catalytic α-subunit of the ATP-dependent transmembrane sodium-potassium pump. Heterozygous mutations in this gene have been reported as causing both sporadic and inherited forms of alternating hemiplegia of childhood and rapid-onset dystonia parkinsonism. We discuss the literature on phenotypes associated with known variants in ATP1A3, examine past functional studies of the role of ATP1A3 in neuronal function, and describe a novel clinical presentation associated with mutation of this gene.

19.
Biochem Biophys Res Commun ; 450(1): 802-7, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24952159

ABSTRACT

Post-translational modifications (PTMs) of histones such as phosphorylation, acetylation, and ubiquitination, collectively referred to as the "histone-code", have been known to regulate gene expression and chromatin condensation for over a decade. They are also implicated in processes such as DNA repair and apoptosis. However, the study of the phosphorylation of histones has been mainly focused on chromosome condensation and mitosis. Therefore, the phosphorylation of histones in apoptosis is not fully understood. It was recently demonstrated by Tang et al. that histones are released from nucleosome during apoptosis, an observation that is in agreement with our findings. In addition to the release of histones, the dephosphorylation of histone H3 at Thr-3 and Ser-10 was observed during apoptosis in some cancer cells. Our data suggest that the modification and release of histones could serve markers of apoptosis in human cancer cells. We also suggest that the released histones, especially H3, could be translocated to mitochondria during apoptosis.


Subject(s)
Apoptosis/physiology , Histones/metabolism , Mitochondria/metabolism , Protein Processing, Post-Translational/physiology , Staurosporine/pharmacology , Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Humans , Jurkat Cells , Mitochondria/drug effects , Protein Processing, Post-Translational/drug effects
20.
Stem Cells Transl Med ; 3(5): 599-609, 2014 May.
Article in English | MEDLINE | ID: mdl-24763686

ABSTRACT

It has recently been shown that genomic integrity (with respect to copy number variants [CNVs]) is compromised in human induced pluripotent stem cells (iPSCs) generated by viral-based ectopic expression of specific transcription factors (e.g., Oct4, Sox2, Klf4, and c-Myc). However, it is unclear how different methods for iPSC generation compare with one another with respect to CNV formation. Because array-based methods remain the gold standard for detecting unbalanced structural variants (i.e., CNVs), we have used this approach to comprehensively identify CNVs in iPSC as a proxy for determining whether our modified protein-based method minimizes genomic instability compared with retro- and lentiviral methods. In this study, we established an improved method for protein reprogramming by using partially purified reprogramming proteins, resulting in more efficient generation of iPSCs from C57/BL6J mouse hepatocytes than using protein extracts. We also developed a robust and unbiased 1 M custom array CGH platform to identify novel CNVs and previously described hot spots for CNV formation, allowing us to detect CNVs down to the size of 1.9 kb. The genomic integrity of these protein-based mouse iPSCs (p-miPSCs) was compared with miPSCs developed from viral-based strategies (i.e., retroviral: retro-miPSCs or lentiviral: lenti-miPSCs). We identified an increased CNV content in lenti-miPSCs and retro-miPSCs (29∼53 CNVs) compared with p-miPSCs (9∼10 CNVs), indicating that our improved protein-based reprogramming method maintains genomic integrity better than current viral reprogramming methods. Thus, our study, for the first time to our knowledge, demonstrates that reprogramming methods significantly influence the genomic integrity of resulting iPSCs.


Subject(s)
Genomic Instability , Induced Pluripotent Stem Cells , Lentivirus , Transcription Factors , Transduction, Genetic , Animals , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Mice , Transcription Factors/biosynthesis , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL