Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
bioRxiv ; 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38645112

ABSTRACT

Most GWAS loci are presumed to affect gene regulation, however, only ∼43% colocalize with expression quantitative trait loci (eQTLs). To address this colocalization gap, we identify eQTLs, chromatin accessibility QTLs (caQTLs), and histone acetylation QTLs (haQTLs) using molecular samples from three early developmental (EDev) tissues. Through colocalization, we annotate 586 GWAS loci for 17 traits by QTL complexity, QTL phenotype, and QTL temporal specificity. We show that GWAS loci are highly enriched for colocalization with complex QTL modules that affect multiple elements (genes and/or peaks). We also demonstrate that caQTLs and haQTLs capture regulatory variations not associated with eQTLs and explain ∼49% of the functionally annotated GWAS loci. Additionally, we show that EDev-unique QTLs are strongly depleted for colocalizing with GWAS loci. By conducting one of the largest multi-omic QTL studies to date, we demonstrate that many GWAS loci exhibit phenotypic complexity and therefore, are missed by traditional eQTL analyses.

2.
Nanoscale ; 15(32): 13322-13334, 2023 Aug 17.
Article in English | MEDLINE | ID: mdl-37526009

ABSTRACT

Here, rational engineering of doxorubicin prodrug loaded peptide-targeted liposomal nanoparticles to selectively target metastatic breast cancer cells in vivo is described. Glucose-regulated protein 78 (GRP78), a heat shock protein typically localized in the endoplasmic reticulum in healthy cells, has been identified to home to the cell surface in certain cancers, and thus has emerged as a promising therapeutic target. Recent reports indicated GRP78 to be expressed on the cell surface of an aggressive subpopulation of stem-like breast cancer cells that exhibit metastatic potential. In this study, a targeted nanoparticle formulation with a GRP78-binding peptide (Kd of 7.4 ± 1.0 µM) was optimized to selectively target this subpopulation. In vitro studies with breast cancer cell lines showed the targeted nanoparticle formulation (TNPGRP78pep) achieved enhanced cellular uptake, while maintaining selectivity over the control groups. In vivo, TNPGRP78pep loaded with doxorubicin prodrug was evaluated using a lung metastatic mouse model and demonstrated inhibition of breast cancer cell seeding to lungs down at the level of negative control groups. Combined, this study established that specific-targeting of surface GRP78 expressing a subpopulation of aggressive breast cancer cells was able to inhibit breast cancer metastasis to lungs, and underpinned the significance of GRP78 in breast cancer metastasis.


Subject(s)
Neoplasms , Prodrugs , Animals , Mice , Endoplasmic Reticulum Chaperone BiP , Membrane Proteins , Cell Line, Tumor , Glucose , Peptides , Doxorubicin/pharmacology
3.
Stem Cell Res ; 62: 102799, 2022 07.
Article in English | MEDLINE | ID: mdl-35550987

ABSTRACT

Kabuki syndrome (KS) is a rare genetic disorder typically characterized by facial abnormalities, developmental delay, cognitive dysfunction, and organ impairment. In this report, fibroblast cells obtained from a KS patient containing a heterozygous KMT2D c.12592 C>T mutation (p.R4198X) were reprogrammed using non-integrative Sendai virus to generate three induced pluripotent stem cell (iPSC) clones. The iPSC lines retained the KS patient mutation, and displayed normal karyotypes, pluripotency marker expression, and the ability to differentiate into the three germ layers.


Subject(s)
Hematologic Diseases , Induced Pluripotent Stem Cells , Vestibular Diseases , Abnormalities, Multiple , Face/abnormalities , Hematologic Diseases/genetics , Humans , Mutation/genetics , Vestibular Diseases/genetics
4.
Oncogene ; 40(23): 4050-4059, 2021 06.
Article in English | MEDLINE | ID: mdl-33981001

ABSTRACT

The heat shock protein GRP78 typically resides in the endoplasmic reticulum in normal tissues, but it has been shown to be expressed on the cell surface of several cancer cells, and some stem cells, where it can act as a signaling molecule by not-yet-fully defined mechanisms. Although cell surface GRP78 (sGRP78) has emerged as an attractive chemotherapeutic target, understanding how sGRP78 is functioning in cancer has been complicated by the fact that sGRP78 can function in a cell-context dependent manner, with a diverse array of reported binding partners, to regulate a variety of cellular responses. We had previously shown that sGRP78 was important in regulating pluripotent stem cell (PSC) functions, and hypothesized that embryonic-like mechanisms of GRP78 were critical to regulating aggressive breast cancer cell functions. Here, using proteomics we identify Dermcidin (DCD) as a novel sGRP78 binding partner common to both PSCs and breast cancer cells. We show that GRP78 and DCD cooperate to regulate stem cell and cancer cell migration that is dependent on the cell surface functions of these proteins. Finally, we identify Wnt/ß-catenin signaling, a critical pathway in stem cell and cancer cell biology, as an important downstream intermediate in regulating this migration phenotype.


Subject(s)
Breast Neoplasms/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum Chaperone BiP/metabolism , Peptides/metabolism , Wnt Signaling Pathway , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Female , Humans , Stem Cells/metabolism
5.
Stem Cell Res ; 49: 102096, 2020 12.
Article in English | MEDLINE | ID: mdl-33370871

ABSTRACT

Cancer-derived iPSCs have provided valuable insight into oncogenesis, but human cancer cells can often be difficult to reprogram, especially in cases of complex genetic abnormalities. Here we report, to our knowledge, the first successful generation of an iPSC line from a human immortalized acute myeloid leukemia (AML) cell line, the cell line HL-60. This iPSC line retains a majority of the leukemic genotype and displays defects in myeloid differentiation, thus providing a tool for modeling and studying AML.


Subject(s)
Induced Pluripotent Stem Cells , Leukemia, Myeloid, Acute , Cell Differentiation , HL-60 Cells , Hematopoiesis , Humans , Leukemia, Myeloid, Acute/genetics
6.
Sci Rep ; 10(1): 3474, 2020 02 26.
Article in English | MEDLINE | ID: mdl-32103065

ABSTRACT

Reliable approaches to identify stem cell mechanisms that mediate aggressive cancer could have great therapeutic value, based on the growing evidence of embryonic signatures in metastatic cancers. However, how to best identify and target stem-like mechanisms aberrantly acquired by cancer cells has been challenging. We harnessed the power of reprogramming to examine GRP78, a chaperone protein generally restricted to the endoplasmic reticulum in normal tissues, but which is expressed on the cell surface of human embryonic stem cells and many cancer types. We have discovered that (1) cell surface GRP78 (sGRP78) is expressed on iPSCs and is important in reprogramming, (2) sGRP78 promotes cellular functions in both pluripotent and breast cancer cells (3) overexpression of GRP78 in breast cancer cells leads to an induction of a CD24-/CD44+ tumor initiating cell (TIC) population (4) sGRP78+ breast cancer cells are enriched for stemness genes and appear to be a subset of TICs (5) sGRP78+ breast cancer cells show an enhanced ability to seed metastatic organ sites in vivo. These collective findings show that GRP78 has important functions in regulating both pluripotency and oncogenesis, and suggest that sGRP78 marks a stem-like population in breast cancer cells that has increased metastatic potential in vivo.


Subject(s)
Cell Differentiation , Cell Self Renewal , Heat-Shock Proteins/metabolism , Neoplastic Stem Cells/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic , Cellular Reprogramming , Endoplasmic Reticulum Chaperone BiP , Female , HEK293 Cells , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice , Mice, Knockout , Neoplastic Stem Cells/cytology , RNA Interference , RNA, Small Interfering/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transplantation, Heterologous
7.
Stem Cell Res ; 41: 101587, 2019 12.
Article in English | MEDLINE | ID: mdl-31739201

ABSTRACT

Using iPSCs to study cancer has been complicated by the fact that many cancer cells are difficult to reprogram, which has been attributed to the genomic abnormalities present. Acute Myeloid Leukemia (AML) is a complex disease that presents with various types of genomic aberrations that affect prognosis. Here we reprogrammed CD34+ cells from an AML patient containing a rare der(7)t(7;13) translocation associated with poor prognosis, who had relapsed and was refractory to current treatments. The generated AML-iPSCs displayed normal karyotypes and myeloid differentiation potential. These findings have implications for modeling and treating AML disease.


Subject(s)
Bone Marrow/pathology , Cell Differentiation , Drug Resistance, Neoplasm , Induced Pluripotent Stem Cells/pathology , Leukemia, Myeloid, Acute/pathology , Myeloid Cells/pathology , Neoplasm Recurrence, Local/pathology , Aged , Humans , Karyotype , Male , Tumor Cells, Cultured
8.
Genome Biol ; 18(1): 135, 2017 07 20.
Article in English | MEDLINE | ID: mdl-28728561

ABSTRACT

Three recent studies analyzing large-scale collections of human induced pluripotent stem cell lines provide valuable insight into how genetic regulatory variation affects cellular and molecular traits.


Subject(s)
Disease , Genetic Variation , Induced Pluripotent Stem Cells , Models, Genetic , Cell Differentiation , Humans
9.
Cell Stem Cell ; 20(4): 505-517.e6, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28388429

ABSTRACT

Induced pluripotent stem cells (iPSCs) show variable methylation patterns between lines, some of which reflect aberrant differences relative to embryonic stem cells (ESCs). To examine whether this aberrant methylation results from genetic variation or non-genetic mechanisms, we generated human iPSCs from monozygotic twins to investigate how genetic background, clone, and passage number contribute. We found that aberrantly methylated CpGs are enriched in regulatory regions associated with MYC protein motifs and affect gene expression. We classified differentially methylated CpGs as being associated with genetic and/or non-genetic factors (clone and passage), and we found that aberrant methylation preferentially occurs at CpGs associated with clone-specific effects. We further found that clone-specific effects play a strong role in recurrent aberrant methylation at specific CpG sites across different studies. Our results argue that a non-genetic biological mechanism underlies aberrant methylation in iPSCs and that it is likely based on a probabilistic process involving MYC that takes place during or shortly after reprogramming.


Subject(s)
DNA Methylation/genetics , Induced Pluripotent Stem Cells/metabolism , Nucleotide Motifs/genetics , Proto-Oncogene Proteins c-myc/metabolism , Clone Cells , CpG Islands/genetics , Fibroblasts/metabolism , Gene Expression Regulation , Genetic Variation , Genome-Wide Association Study , Humans , Sequence Analysis, RNA , Transcription Factors/metabolism , Twins, Monozygotic/genetics
10.
Stem Cell Reports ; 8(4): 1101-1111, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28410643

ABSTRACT

Reprogramming somatic cells to induced pluripotent stem cells (iPSCs) offers the possibility of studying the molecular mechanisms underlying human diseases in cell types difficult to extract from living patients, such as neurons and cardiomyocytes. To date, studies have been published that use small panels of iPSC-derived cell lines to study monogenic diseases. However, to study complex diseases, where the genetic variation underlying the disorder is unknown, a sizable number of patient-specific iPSC lines and controls need to be generated. Currently the methods for deriving and characterizing iPSCs are time consuming, expensive, and, in some cases, descriptive but not quantitative. Here we set out to develop a set of simple methods that reduce cost and increase throughput in the characterization of iPSC lines. Specifically, we outline methods for high-throughput quantification of surface markers, gene expression analysis of in vitro differentiation potential, and evaluation of karyotype with markedly reduced cost.


Subject(s)
Genetic Variation , High-Throughput Screening Assays/methods , Induced Pluripotent Stem Cells/metabolism , Karyotyping/methods , Myocytes, Cardiac/metabolism , Neurons/metabolism , Biomarkers/metabolism , Cell Differentiation , Cell Line , Cellular Reprogramming/genetics , Cost-Benefit Analysis , Genotype , High-Throughput Screening Assays/economics , High-Throughput Screening Assays/instrumentation , Humans , Induced Pluripotent Stem Cells/cytology , Karyotyping/economics , Myocytes, Cardiac/cytology , Neurons/cytology , Phenotype
11.
Stem Cell Reports ; 8(4): 1086-1100, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28410642

ABSTRACT

Large-scale collections of induced pluripotent stem cells (iPSCs) could serve as powerful model systems for examining how genetic variation affects biology and disease. Here we describe the iPSCORE resource: a collection of systematically derived and characterized iPSC lines from 222 ethnically diverse individuals that allows for both familial and association-based genetic studies. iPSCORE lines are pluripotent with high genomic integrity (no or low numbers of somatic copy-number variants) as determined using high-throughput RNA-sequencing and genotyping arrays, respectively. Using iPSCs from a family of individuals, we show that iPSC-derived cardiomyocytes demonstrate gene expression patterns that cluster by genetic background, and can be used to examine variants associated with physiological and disease phenotypes. The iPSCORE collection contains representative individuals for risk and non-risk alleles for 95% of SNPs associated with human phenotypes through genome-wide association studies. Our study demonstrates the utility of iPSCORE for examining how genetic variants influence molecular and physiological traits in iPSCs and derived cell lines.


Subject(s)
Arrhythmias, Cardiac/genetics , Databases, Factual , Genetic Association Studies , Genetic Variation , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Arrhythmias, Cardiac/ethnology , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Cell Differentiation , Cell Line , Cellular Reprogramming/genetics , Genotype , High-Throughput Nucleotide Sequencing , Humans , Induced Pluripotent Stem Cells/cytology , Multigene Family , Myocytes, Cardiac/cytology , Oligonucleotide Array Sequence Analysis , Phenotype , Polymorphism, Single Nucleotide , Racial Groups
12.
Stem Cells ; 33(7): 2343-50, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25826782

ABSTRACT

Danon disease is a familial cardiomyopathy associated with impaired autophagy due to mutations in the gene encoding lysosomal-associated membrane protein type 2 (LAMP-2). Emerging evidence has highlighted the importance of autophagy in regulating cardiomyocyte bioenergetics, function, and survival. However, the mechanisms responsible for cellular dysfunction and death in cardiomyocytes with impaired autophagic flux remain unclear. To investigate the molecular mechanisms responsible for Danon disease, we created induced pluripotent stem cells (iPSCs) from two patients with different LAMP-2 mutations. Danon iPSC-derived cardiomyocytes (iPSC-CMs) exhibited impaired autophagic flux and key features of heart failure such as increased cell size, increased expression of natriuretic peptides, and abnormal calcium handling compared to control iPSC-CMs. Additionally, Danon iPSC-CMs demonstrated excessive amounts of mitochondrial oxidative stress and apoptosis. Using the sulfhydryl antioxidant N-acetylcysteine to scavenge free radicals resulted in a significant reduction in apoptotic cell death in Danon iPSC-CMs. In summary, we have modeled Danon disease using human iPSC-CMs from patients with mutations in LAMP-2, allowing us to gain mechanistic insight into the pathogenesis of this disease. We demonstrate that LAMP-2 deficiency leads to an impairment in autophagic flux, which results in excessive oxidative stress, and subsequent cardiomyocyte apoptosis. Scavenging excessive free radicals with antioxidants may be beneficial for patients with Danon disease. In vivo studies will be necessary to validate this new treatment strategy.


Subject(s)
Glycogen Storage Disease Type IIb/genetics , Heart Failure/genetics , Myocytes, Cardiac/metabolism , Oxidative Stress/genetics , Apoptosis , Autophagy , Glycogen Storage Disease Type IIb/pathology , Heart Failure/pathology , Humans , Induced Pluripotent Stem Cells
13.
Nat Commun ; 4: 1382, 2013.
Article in English | MEDLINE | ID: mdl-23340422

ABSTRACT

Recent studies indicate that human-induced pluripotent stem cells contain genomic structural variations and point mutations in coding regions. However, these studies have focused on fibroblast-derived human induced pluripotent stem cells, and it is currently unknown whether the use of alternative somatic cell sources with varying reprogramming efficiencies would result in different levels of genetic alterations. Here we characterize the genomic integrity of eight human induced pluripotent stem cell lines derived from five different non-fibroblast somatic cell types. We show that protein-coding mutations are a general feature of the human induced pluripotent stem cell state and are independent of somatic cell source. Furthermore, we analyse a total of 17 point mutations found in human induced pluripotent stem cells and demonstrate that they do not generally facilitate the acquisition of pluripotency and thus are not likely to provide a selective advantage for reprogramming.


Subject(s)
Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/metabolism , Mutation/genetics , Open Reading Frames/genetics , Alleles , Base Sequence , Cell Line , Fibroblasts/cytology , Gene Silencing , Human Umbilical Vein Endothelial Cells , Humans , Molecular Sequence Data , Point Mutation/genetics , Retroviridae , Sequence Analysis, RNA
14.
J Biol Chem ; 287(48): 40767-78, 2012 Nov 23.
Article in English | MEDLINE | ID: mdl-23019325

ABSTRACT

BACKGROUND: Strategies on the basis of doxycycline-inducible lentiviruses in mouse cells allowed the examination of mechanisms governing somatic cell reprogramming. RESULTS: Using a doxycycline-inducible human reprogramming system, we identified unreported miRs enhancing reprogramming efficiency. CONCLUSION: We generated a drug-inducible human reprogramming reporter system as an invaluable tool for genetic or chemical screenings. SIGNIFICANCE: These cellular systems provide a tool to enable the advancement of reprogramming technologies in human cells. Reprogramming of somatic cells into induced pluripotent stem cells is achieved by the expression of defined transcription factors. In the last few years, reprogramming strategies on the basis of doxycycline-inducible lentiviruses in mouse cells became highly powerful for screening purposes when the expression of a GFP gene, driven by the reactivation of endogenous stem cell specific promoters, was used as a reprogramming reporter signal. However, similar reporter systems in human cells have not been generated. Here, we describe the derivation of drug-inducible human fibroblast-like cell lines that express different subsets of reprogramming factors containing a GFP gene under the expression of the endogenous OCT4 promoter. These cell lines can be used to screen functional substitutes for reprogramming factors or modifiers of reprogramming efficiency. As a proof of principle of this system, we performed a screening of a library of pluripotent-enriched microRNAs and identified hsa-miR-519a as a novel inducer of reprogramming efficiency.


Subject(s)
Cell Differentiation , Cytological Techniques/methods , Doxycycline/pharmacology , Genes, Reporter/drug effects , Stem Cells/cytology , Animals , Cell Differentiation/drug effects , Cell Line , Drug Evaluation, Preclinical/methods , Gene Expression/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lentivirus/genetics , Lentivirus/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Stem Cells/metabolism
15.
Proc Natl Acad Sci U S A ; 109(40): 16196-201, 2012 Oct 02.
Article in English | MEDLINE | ID: mdl-22991473

ABSTRACT

Generation of human induced pluripotent stem cells (hiPSCs) by the expression of specific transcription factors depends on successful epigenetic reprogramming to a pluripotent state. Although hiPSCs and human embryonic stem cells (hESCs) display a similar epigenome, recent reports demonstrated the persistence of specific epigenetic marks from the somatic cell type of origin and aberrant methylation patterns in hiPSCs. However, it remains unknown whether the use of different somatic cell sources, encompassing variable levels of selection pressure during reprogramming, influences the level of epigenetic aberrations in hiPSCs. In this work, we characterized the epigenomic integrity of 17 hiPSC lines derived from six different cell types with varied reprogramming efficiencies. We demonstrate that epigenetic aberrations are a general feature of the hiPSC state and are independent of the somatic cell source. Interestingly, we observe that the reprogramming efficiency of somatic cell lines inversely correlates with the amount of methylation change needed to acquire pluripotency. Additionally, we determine that both shared and line-specific epigenetic aberrations in hiPSCs can directly translate into changes in gene expression in both the pluripotent and differentiated states. Significantly, our analysis of different hiPSC lines from multiple cell types of origin allow us to identify a reprogramming-specific epigenetic signature comprised of nine aberrantly methylated genes that is able to segregate hESC and hiPSC lines regardless of the somatic cell source or differentiation state.


Subject(s)
Cellular Reprogramming/physiology , DNA Methylation/genetics , Epigenesis, Genetic/physiology , Induced Pluripotent Stem Cells/physiology , Cell Line , Cellular Reprogramming/genetics , CpG Islands/genetics , Epigenesis, Genetic/genetics , Epigenomics , Fluorescent Antibody Technique , Gene Library , Humans , Microarray Analysis , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA
16.
Curr Opin Hematol ; 19(4): 256-60, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22555392

ABSTRACT

PURPOSE OF REVIEW: With the advent of reprogramming came the possibility of generating patient-specific clinical therapies. The purpose of this review is to discuss the recent key developments and remaining limitations in the stem cell and hematopoietic fields toward the goal of translating induced pluripotent stem cell (iPSC) technologies into the hematology clinic. RECENT FINDINGS: Recent progress in the hematopoietic and reprogramming fields has included identification of hematopoietic stem cells (HSCs) capable of long-term engraftment at the single-cell level, improvements in ex-vivo expansion of HSCs, transdifferentiation of somatic cells into hematopoietic progenitors, and the 'correction' of several disease-specific iPSCs using various gene-targeting strategies. SUMMARY: In light of recent advances, it is the hope that the hurdle of obtaining fully functional HSCs in a laboratory setting will be overcome through either in-vitro differentiation of pluripotent stem cells, ex-vivo expansion of HSCs obtained in vivo, or transdifferentiation from other somatic sources. Equally important will be for the reprogramming field to better understand the causes and consequences of the recently reported genetic/epigenetic variations present in iPSCs, especially within the context of gene-targeted strategies for correcting disease. The progress in the reprogramming and hematopoietic fields provides a strong foundation for future work toward the possible treatment of numerous hematological disorders using iPSC technologies.


Subject(s)
Cell Differentiation , Hematology/methods , Hematopoietic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Cell Culture Techniques/methods , Hematopoietic Stem Cell Transplantation/methods , Humans
17.
Cell Res ; 22(1): 168-77, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22064701

ABSTRACT

Metabolism is vital to every aspect of cell function, yet the metabolome of induced pluripotent stem cells (iPSCs) remains largely unexplored. Here we report, using an untargeted metabolomics approach, that human iPSCs share a pluripotent metabolomic signature with embryonic stem cells (ESCs) that is distinct from their parental cells, and that is characterized by changes in metabolites involved in cellular respiration. Examination of cellular bioenergetics corroborated with our metabolomic analysis, and demonstrated that somatic cells convert from an oxidative state to a glycolytic state in pluripotency. Interestingly, the bioenergetics of various somatic cells correlated with their reprogramming efficiencies. We further identified metabolites that differ between iPSCs and ESCs, which revealed novel metabolic pathways that play a critical role in regulating somatic cell reprogramming. Our findings are the first to globally analyze the metabolome of iPSCs, and provide mechanistic insight into a new layer of regulation involved in inducing pluripotency, and in evaluating iPSC and ESC equivalence.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/metabolism , Metabolome , DNA Methylation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Energy Metabolism , Gene Expression Regulation , Glycolysis , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Induced Pluripotent Stem Cells/cytology , Oxidation-Reduction , Oxidative Phosphorylation , Plasmids/genetics , Plasmids/metabolism , Retroviridae/genetics , Retroviridae/metabolism
18.
Cell Metab ; 14(2): 143-4, 2011 Aug 03.
Article in English | MEDLINE | ID: mdl-21803281

ABSTRACT

Reprogramming involves multiple layers of molecular regulation, yet it remains relatively unknown how the cell's metabolism is changing and/or contributing to this process. In this issue of Cell Metabolism, Folmes et al. (2011) demonstrate that reprogramming induces a bioenergetic transition from an oxidative to a glycolytic state, and provide evidence to suggest that these changes may precede pluripotency.

19.
PLoS One ; 6(5): e19743, 2011.
Article in English | MEDLINE | ID: mdl-21603572

ABSTRACT

The ability to induce somatic cells to pluripotency by ectopic expression of defined transcription factors (e.g. KLF-4, OCT4, SOX2, c-MYC, or KOSM) has transformed the future of regenerative medicine. Here we report somatic cell reprogramming of human umbilical vein endothelial cells (HUVECs), yielding induced pluripotent stem (iPS) cells with the fastest kinetics, and one of the highest reprogramming efficiencies for a human somatic cell to date. HUVEC-derived iPS (Huv-iPS) cell colonies appeared as early as 6 days after a single KOSM infection, and were generated with a 2.5-3% reprogramming efficiency. Furthermore, when HUVEC reprogramming was performed under hypoxic conditions in the presence of a TGF-beta family signaling inhibitor, colony formation increased an additional ∼2.5-fold over standard conditions. Huv-iPS cells were indistinguishable from human embryonic stem (ES) cells with regards to morphology, pluripotent marker expression, and their ability to generate all embryonic germ layers in vitro and in vivo. The high efficiency and rapid kinetics of Huv-iPS cell formation, coupled with the ease by which HUVECs can be collected, expanded and stored, make these cells an attractive somatic source for therapeutic application, and for studying the reprogramming process.


Subject(s)
Cell Culture Techniques/methods , Endothelial Cells/cytology , Induced Pluripotent Stem Cells/cytology , Umbilical Veins/cytology , Biomarkers , Cellular Reprogramming , Embryonic Stem Cells/cytology , Humans
20.
Cell Stem Cell ; 8(4): 347-8, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21474093

ABSTRACT

Several recent reports (Mayshar et al., 2010; Laurent et al., 2011; Lister et al., 2011; Gore et al., 2011; Hussein et al., 2011) uncover genetic and epigenetic alterations in induced pluripotent stem cells, stimulating debate about their future. However, will these important findings really impact what we hope to gain?

SELECTION OF CITATIONS
SEARCH DETAIL
...