Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Stem Cell Res ; 72: 103192, 2023 10.
Article in English | MEDLINE | ID: mdl-37660555

ABSTRACT

Type 2 Long QT Syndrome (LQT2) is a rare genetic heart rhythm disorder causing life-threatening arrhythmias. We derived induced pluripotent stem cell (iPSC) lines from two patients with LQT2, aged 18 and 6, both carrying a heterozygous missense mutation on the 3rd and 11th exons of KCNH2. The iPSC lines exhibited normal genomes, expressed pluripotent markers, and differentiated into trilineage embryonic layers. These patient-specific iPSC lines provide a valuable model to study the molecular and functional impact of the hERG channel gene mutation in LQT2 and to develop personalized therapeutic approaches for this syndrome.


Subject(s)
Induced Pluripotent Stem Cells , Long QT Syndrome , Humans , Induced Pluripotent Stem Cells/metabolism , ERG1 Potassium Channel/genetics , Long QT Syndrome/metabolism , Arrhythmias, Cardiac/metabolism , Mutation
2.
Aging Cell ; 21(11): e13714, 2022 11.
Article in English | MEDLINE | ID: mdl-36251933

ABSTRACT

Recent advances in cell reprogramming showed that OSKM induction is able to improve cell physiology in vitro and in vivo. Here, we show that a single short reprogramming induction is sufficient to prevent musculoskeletal functions deterioration of mice, when applied in early life. In addition, in old age, treated mice have improved tissue structures in kidney, spleen, skin, and lung, with an increased lifespan of 15% associated with organ-specific differential age-related DNA methylation signatures rejuvenated by the treatment. Altogether, our results indicate that a single short reprogramming early in life might initiate and propagate an epigenetically related mechanism to promote a healthy lifespan.


Subject(s)
Cellular Reprogramming , Longevity , Mice , Animals , Longevity/genetics , Cellular Reprogramming/genetics , Health Status
3.
Stem Cell Res ; 60: 102727, 2022 04.
Article in English | MEDLINE | ID: mdl-35245853

ABSTRACT

Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT) is a genetic disorder characterized by ventricular tachycardia, that can cause the heart to stop beating leading to death. The prevalence is 1/10.000 and in approximately 60% of cases, the syndrome can be due to a mutation of the cardiac ryanodine receptor gene (RyR2). We derived an induced pluripotent stem cell (iPSC) line from an 11-year-old patient blood-cells, carrying a heterozygous missense mutation on the 8th exon of the RyR2 N-terminal part. This reprogramed CPVT line displayed normal karyotype, expressed pluripotent markers and had a capacity to differentiate in trilineage embryonic layers.


Subject(s)
Induced Pluripotent Stem Cells , Tachycardia, Ventricular , Child , Humans , Induced Pluripotent Stem Cells/metabolism , Mutation/genetics , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Tachycardia, Ventricular/genetics
4.
Stem Cell Res ; 53: 102297, 2021 05.
Article in English | MEDLINE | ID: mdl-33780731

ABSTRACT

The study of molecular mechanism driving osteoarticular diseases like osteoarthritis or osteoporosis is impaired by the low accessibility to mesenchymal stem cells (MSC) from healthy donors (HD) for differential multi-omics analysis. Advances in cell reprogramming have, however, provided both a new source of human cells for laboratory research and a strategy to erase epigenetic marks involved in cell identity and the development of diseases. To unravel the pathological signatures on the MSC at the origin of cellular drifts during the formation of bone and cartilage, we previously developed iPSC from MSC of osteoarthritis donors. Here we present the derivation of three iPSCs from healthy age matched donors to model the disease and further identify (epi)genomic signatures of the pathology.


Subject(s)
Induced Pluripotent Stem Cells , Mesenchymal Stem Cells , Pluripotent Stem Cells , Aged , Cell Differentiation , Cell Line , Cellular Reprogramming , Humans
5.
Stem Cell Res ; 49: 102106, 2020 12.
Article in English | MEDLINE | ID: mdl-33370874

ABSTRACT

Congenital myasthenic syndromes (CMS) are a class of inherited disorders affecting the neuromuscular junction, a synapse whose activity is essential for movement. CMS with acetylcholinesterase (AChE) deficiency are caused by mutations in COLQ, a collagen that anchors AChE in the synapse. To study the pathophysiological mechanisms of the disease in human cells, we have generated iPSC from a patient's Peripheral Blood Mononuclear cells (PBMC) by reprogramming these cells using a non-integrative method using Sendai viruses bearing the four Yamanaka factors Oct3/4, Sox2, Klf4, and L-Myc.


Subject(s)
Cell Line , Induced Pluripotent Stem Cells , Myasthenic Syndromes, Congenital , Acetylcholinesterase/genetics , Collagen , Humans , Kruppel-Like Factor 4 , Leukocytes, Mononuclear , Muscle Proteins/genetics , Mutation/genetics , Myasthenic Syndromes, Congenital/genetics
6.
Stem Cell Res ; 49: 102094, 2020 12.
Article in English | MEDLINE | ID: mdl-33246213

ABSTRACT

Duchenne Muscular Dystrophy (DMD) is a X-linked degenerative pathology with a prevalence of 1/3600-6000 boys due to the absence of functional dystrophin in muscles. This muscular disease leads to skeletal muscle damages, respiratory failure and in the later stages dilated cardiomyopathy (DCM) leading to heart failure. We generated iPSC lines from three different DMD patients carrying respectively deletions of exons 1, 52 and 55 in the dystrophin gene. The reprogrammed iPSC lines showed expression of pluripotent markers, capacity to differentiate in trilineage embryonic layers and a normal karyotype.


Subject(s)
Cell Line , Induced Pluripotent Stem Cells , Muscular Dystrophy, Duchenne , Dystrophin/genetics , Exons , Humans , Male , Muscles , Muscular Dystrophy, Duchenne/genetics
7.
Stem Cell Res ; 45: 101807, 2020 05.
Article in English | MEDLINE | ID: mdl-32416578

ABSTRACT

Rothmund-Thomson Syndrome (RTS) is a rare autosomal recessive disease that manifests several clinical features of accelerated aging. These findings include atrophic skin and pigment changes, alopecia, osteopenia, cataracts, and an increased incidence of cancer for patients. Mutations in RECQL4 gene are responsible for cases of RTS. RECQL4 belongs to the RECQ DNA helicase family which has been shown to participate in many aspects of DNA metabolism. To be able to study the cellular defects related to the pathology, we derived an induced pluripotent cell line from RTS patient fibroblasts, with the ability to re-differentiate into the three embryonic germ layers.


Subject(s)
Induced Pluripotent Stem Cells , Rothmund-Thomson Syndrome , Skin Abnormalities , Fibroblasts , Humans , Mutation , Rothmund-Thomson Syndrome/genetics
8.
Stem Cell Res ; 44: 101721, 2020 04.
Article in English | MEDLINE | ID: mdl-32200258

ABSTRACT

Mesenchymal stem cells (MSCs) are a unique population of adult stem cells that can differentiate into many cell types. As such, MSCs represent an interesting source of stem cells for use in the clinical treatment of a variety of disorders involving tissue regeneration. It is therefore crucial to investigate further, whether MSCs from patients with bone or cartilage diseases are able to provide iPSCs lines with efficient differentiation ability into MSC derivatives. For this purpose, we derived 3 stable iPSC lines from the MSCs of 3 elderly patients with osteoarthritis (OA) able to re-differentiate into MSC to make bone, cartilage and adipose tissue.


Subject(s)
Induced Pluripotent Stem Cells , Mesenchymal Stem Cells , Osteoarthritis , Pluripotent Stem Cells , Adult , Aged , Cell Differentiation , Cell Line , Humans , Osteoarthritis/therapy
9.
Stem Cell Res ; 43: 101696, 2020 03.
Article in English | MEDLINE | ID: mdl-31918214

ABSTRACT

Bloom syndrome is characterized by severe pre- and postnatal growth deficiency, immune abnormalities, sensitivity to sunlight, insulin resistance, and a high risk for many cancers that occur at an early age. The diagnosis is established on characteristic clinical features and/or presence of biallelic pathogenic variants in the BLM gene. An increased frequency of sister-chromatid exchanges is also observed and can be useful to diagnose BS patients with weak or no clinical features. For the first time, we derived an induced pluripotent cell line from a Bloom syndrome patient retaining the specific sister-chromatid exchange feature as a unique tool to model the pathology.


Subject(s)
Bloom Syndrome/genetics , Induced Pluripotent Stem Cells/metabolism , Sister Chromatid Exchange/genetics , Adolescent , Animals , Female , Humans
10.
Stem Cell Res ; 39: 101515, 2019 08.
Article in English | MEDLINE | ID: mdl-31404747

ABSTRACT

Werner syndrome (WS) is a rare human autosomal recessive disorder characterized by early onset of aging-associated diseases, chromosomal instability, and cancer predisposition, without therapeutic treatment solution. Major clinical symptoms of WS include common age-associated diseases, such as insulin-resistant diabetes mellitus, and atherosclerosis. WRN, the gene responsible for the disease, encodes a RECQL-type DNA helicase with a role in telomere metabolism. We derived a stable iPSC line from 53 years old patient's PBMC, with a normal karyotype, but exhibiting a short telomere length, as a major aspect of the cellular phenotype involved in the pathology.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Leukocytes, Mononuclear/cytology , Werner Syndrome/genetics , Cells, Cultured , Flow Cytometry , Fluorescent Antibody Technique , Genetic Predisposition to Disease/genetics , Humans , Karyotyping , Leukocytes, Mononuclear/metabolism , Microsatellite Repeats/genetics , Telomere/genetics
11.
Cell Cycle ; 17(13): 1667-1681, 2018.
Article in English | MEDLINE | ID: mdl-29963964

ABSTRACT

Organismal aging entails a gradual decline of normal physiological functions and a major contributor to this decline is withdrawal of the cell cycle, known as senescence. Senescence can result from telomere diminution leading to a finite number of population doublings, known as replicative senescence (RS), or from oncogene overexpression, as a protective mechanism against cancer. Senescence is associated with large-scale chromatin re-organization and changes in gene expression. Replication stress is a complex phenomenon, defined as the slowing or stalling of replication fork progression and/or DNA synthesis, which has serious implications for genome stability, and consequently in human diseases. Aberrant replication fork structures activate the replication stress response leading to the activation of dormant origins, which is thought to be a safeguard mechanism to complete DNA replication on time. However, the relationship between replicative stress and the changes in the spatiotemporal program of DNA replication in senescence progression remains unclear. Here, we studied the DNA replication program during senescence progression in proliferative and pre-senescent cells from donors of various ages by single DNA fiber combing of replicated DNA, origin mapping by sequencing short nascent strands and genome-wide profiling of replication timing (TRT). We demonstrate that, progression into RS leads to reduced replication fork rates and activation of dormant origins, which are the hallmarks of replication stress. However, with the exception of a delay in RT of the CREB5 gene in all pre-senescent cells, RT was globally unaffected by replication stress during entry into either oncogene-induced or RS. Consequently, we conclude that RT alterations associated with physiological and accelerated aging, do not result from senescence progression. Our results clarify the interplay between senescence, aging and replication programs and demonstrate that RT is largely resistant to replication stress.


Subject(s)
Cellular Senescence , DNA Replication Timing , Stress, Physiological , Cyclic AMP Response Element-Binding Protein A/metabolism , Fibroblasts/cytology , Humans , Lamins/metabolism , Oncogenes , Progeria/pathology , Protein Domains
12.
Proc Natl Acad Sci U S A ; 114(51): E10972-E10980, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29196523

ABSTRACT

Progeroid syndromes are rare genetic disorders that phenotypically resemble natural aging. Different causal mutations have been identified, but no molecular alterations have been identified that are in common to these diseases. DNA replication timing (RT) is a robust cell type-specific epigenetic feature highly conserved in the same cell types from different individuals but altered in disease. Here, we characterized DNA RT program alterations in Hutchinson-Gilford progeria syndrome (HGPS) and Rothmund-Thomson syndrome (RTS) patients compared with natural aging and cellular senescence. Our results identified a progeroid-specific RT signature that is common to cells from three HGPS and three RTS patients and distinguishes them from healthy individuals across a wide range of ages. Among the RT abnormalities, we identified the tumor protein p63 gene (TP63) as a gene marker for progeroid syndromes. By using the redifferentiation of four patient-derived induced pluripotent stem cells as a model for the onset of progeroid syndromes, we tracked the progression of RT abnormalities during development, revealing altered RT of the TP63 gene as an early event in disease progression of both HGPS and RTS. Moreover, the RT abnormalities in progeroid patients were associated with altered isoform expression of TP63 Our findings demonstrate the value of RT studies to identify biomarkers not detected by other methods, reveal abnormal TP63 RT as an early event in progeroid disease progression, and suggest TP63 gene regulation as a potential therapeutic target.


Subject(s)
DNA Replication Timing , Progeria/genetics , Aged, 80 and over , Biomarkers , Child , Fibroblasts/metabolism , Gene Expression , Genomics/methods , Humans , Infant, Newborn , Lamin Type A/genetics , Lamin Type A/metabolism , Progeria/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
13.
Curr Protoc Cell Biol ; 65: 22.20.1-17, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25447077

ABSTRACT

Recent advances in genome-sequencing technology have led to the complete mapping of DNA replication initiation sites in the human genome. This thorough origin mapping facilitates understanding of the relationship between replication initiation events, transcription, and chromatin modifications, and allows the characterization of consensus sequences of potential replication origins. This unit provides a detailed protocol for isolation and sequence analysis of nascent DNA strands. Two variations of the protocol based on non-overlapping assumptions are described below, addressing potential bias issues for whole-genome analyses.


Subject(s)
Chromosome Mapping/methods , Chromosomes, Human/genetics , DNA Replication , Replication Origin , Animals , Humans
14.
Curr Protoc Cell Biol ; 64: 22.18.1-13, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25181303

ABSTRACT

Understanding the regulatory principles ensuring complete DNA replication in each cell division is critical for deciphering the mechanisms that maintain genomic stability. Recent advances in genome sequencing technology facilitated complete mapping of DNA replication sites and helped move the field from observing replication patterns at a handful of single loci to analyzing replication patterns genome-wide. These advances address issues, such as the relationship between replication initiation events, transcription, and chromatin modifications, and identify potential replication origin consensus sequences. This unit summarizes the technological and fundamental aspects of replication profiling and briefly discusses novel insights emerging from mining large datasets, published in the last 3 years, and also describes DNA replication dynamics on a whole-genome scale.


Subject(s)
Chromosome Mapping/methods , Chromosomes , DNA Replication/physiology , Eukaryotic Cells/metabolism , Replication Origin/physiology , Chromosomes/genetics , Chromosomes/metabolism
15.
Curr Gene Ther ; 13(2): 93-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23317057

ABSTRACT

Induced pluripotent stem cells (iPSCs) and embryonic stem cells (ESCs) are two types of pluripotent stem cells that hold great promise for biomedical research and medical applications. iPSCs were initially favorably compared to ESCs. This view was first based on ethical arguments (the generation of iPSCs does not require the destruction of an embryo) and on immunological reasons (it is easier to derive patient HLA-matched iPSCs than ESCs). However, several reports suggest that iPSCs might be characterized by higher occurrence of epigenetic and genetic aberrations than ESCs as a consequence of the reprogramming process. We focus here on the DNA integrity of pluripotent stem cells and examine the three main sources of genomic abnormalities in iPSCs: (1) genomic variety of the parental cells, (2) cell reprogramming, and (3) in vitro cell culture. Recent reports claim that it is possible to generate mouse or human iPSC lines with a mutation level similar to that of the parental cells, suggesting that "genome-friendly" reprogramming techniques can be developed. The issue of iPSC DNA integrity clearly highlights the crucial need of guidelines to define the acceptable level of genomic integrity of pluripotent stem cells for biomedical applications. We discuss here the main issues that such guidelines should address.


Subject(s)
DNA/genetics , Embryonic Stem Cells/cytology , Genomic Instability , Induced Pluripotent Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Embryonic Stem Cells/immunology , Epigenesis, Genetic , Humans , Induced Pluripotent Stem Cells/immunology , Mice
16.
PLoS One ; 6(10): e25761, 2011.
Article in English | MEDLINE | ID: mdl-22022444

ABSTRACT

We have previously shown that human embryonic stem cells can be differentiated into embryonic and fetal type of red blood cells that sequentially express three types of hemoglobins recapitulating early human erythropoiesis. We report here that we have produced iPS from three somatic cell types: adult skin fibroblasts as well as embryonic and fetal mesenchymal stem cells. We show that regardless of the age of the donor cells, the iPS produced are fully reprogrammed into a pluripotent state that is undistinguishable from that of hESCs by low and high-throughput expression and detailed analysis of globin expression patterns by HPLC. This suggests that reprogramming with the four original Yamanaka pluripotency factors leads to complete erasure of all functionally important epigenetic marks associated with erythroid differentiation regardless of the age or the tissue type of the donor cells, at least as detected in these assays. The ability to produce large number of erythroid cells with embryonic and fetal-like characteristics is likely to have many translational applications.


Subject(s)
Cell Culture Techniques/methods , Embryo, Mammalian/cytology , Erythrocytes/cytology , Fetus/cytology , Induced Pluripotent Stem Cells/cytology , Adult , Cell Differentiation/genetics , Cell Line , Erythrocytes/metabolism , Gene Expression Regulation , Hematopoiesis/genetics , Humans , Induced Pluripotent Stem Cells/metabolism
17.
Genome Res ; 21(9): 1438-49, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21750104

ABSTRACT

In metazoans, thousands of DNA replication origins (Oris) are activated at each cell cycle. Their genomic organization and their genetic nature remain elusive. Here, we characterized Oris by nascent strand (NS) purification and a genome-wide analysis in Drosophila and mouse cells. We show that in both species most CpG islands (CGI) contain Oris, although methylation is nearly absent in Drosophila, indicating that this epigenetic mark is not crucial for defining the activated origin. Initiation of DNA synthesis starts at the borders of CGI, resulting in a striking bimodal distribution of NS, suggestive of a dual initiation event. Oris contain a unique nucleotide skew around NS peaks, characterized by G/T and C/A overrepresentation at the 5' and 3' of Ori sites, respectively. Repeated GC-rich elements were detected, which are good predictors of Oris, suggesting that common sequence features are part of metazoan Oris. In the heterochromatic chromosome 4 of Drosophila, Oris correlated with HP1 binding sites. At the chromosome level, regions rich in Oris are early replicating, whereas Ori-poor regions are late replicating. The genome-wide analysis was coupled with a DNA combing analysis to unravel the organization of Oris. The results indicate that Oris are in a large excess, but their activation does not occur at random. They are organized in groups of site-specific but flexible origins that define replicons, where a single origin is activated in each replicon. This organization provides both site specificity and Ori firing flexibility in each replicon, allowing possible adaptation to environmental cues and cell fates.


Subject(s)
DNA Replication/genetics , Genomics , Replication Origin/genetics , Animals , Base Sequence , Binding Sites/genetics , Cell Line , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Mapping , Conserved Sequence/genetics , CpG Islands , Drosophila/genetics , Heterochromatin/genetics , Mice , Promoter Regions, Genetic , Transcription, Genetic
18.
Genome Res ; 19(12): 2288-99, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19767418

ABSTRACT

The organization of mammalian DNA replication is poorly understood. We have produced high-resolution dynamic maps of the timing of replication in human erythroid, mesenchymal, and embryonic stem (ES) cells using TimEX, a method that relies on gaussian convolution of massive, highly redundant determinations of DNA copy-number variations during S phase to produce replication timing profiles. We first obtained timing maps of 3% of the genome using high-density oligonucleotide tiling arrays and then extended the TimEX method genome-wide using massively parallel sequencing. We show that in untransformed human cells, timing of replication is highly regulated and highly synchronous, and that many genomic segments are replicated in temporal transition regions devoid of initiation, where replication forks progress unidirectionally from origins that can be hundreds of kilobases away. Absence of initiation in one transition region is shown at the molecular level by single molecule analysis of replicated DNA (SMARD). Comparison of ES and erythroid cells replication patterns revealed that these cells replicate about 20% of their genome in different quarters of S phase. Importantly, we detected a strong inverse relationship between timing of replication and distance to the closest expressed gene. This relationship can be used to predict tissue-specific timing of replication profiles from expression data and genomic annotations. We also provide evidence that early origins of replication are preferentially located near highly expressed genes, that mid-firing origins are located near moderately expressed genes, and that late-firing origins are located far from genes.


Subject(s)
DNA Replication Timing , DNA Replication , Embryonic Stem Cells , Erythroid Cells , Gene Expression Profiling , Mesenchymal Stem Cells , S Phase , Cell Differentiation , DNA/biosynthesis , DNA/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , Gene Dosage , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Normal Distribution
19.
PLoS One ; 4(6): e5956, 2009 Jun 18.
Article in English | MEDLINE | ID: mdl-19536296

ABSTRACT

Insertional mutagenesis has emerged as a major obstacle for gene therapy based on vectors that integrate randomly in the genome. Reducing the genotoxicity of genomic viral integration can, in first approximation, be equated with reducing the risk of oncogene activation, at least in the case of therapeutic payloads that have no known oncogenic potential, such as the globin genes. An attractive solution to the problem of oncogene activation is the inclusion of insulators/enhancer-blockers in the viral vectors. In this study we have used Recombinase-Mediated Cassette Exchange to characterize the effect of integration of globin therapeutic cassettes in the presence or absence of the chicken HS4 and three other putative insulators inserted near Stil, Tal1 and MAP17, three well-known cellular proto-oncogenes in the SCL/Tal1 locus. We show that insertion of a Locus Control Region-driven globin therapeutic globin transgene had a dramatic activating effect on Tal1 and Map17, the two closest genes, a minor effect on Stil, and no effect on Cyp4x1, a non-expressed gene. Of the four element tested, cHS4 was the only one that was able to suppress this transgene-mediated insertional transcriptional activation. cHS4 had a strong suppressive effect on the activation expression of Map17 but has little or no effect on expression of Tal1. The suppressive activity of cHS4 is therefore promoter specific. Importantly, the observed suppressive effect of cHS4 on Map17 activation did not depend on its intercalation between the LCR and the Map 17 promoter. Rather, presence of one or two copies of cHS4 anywhere within the transgene was sufficient to almost completely block the activation of Map17. Therefore, at this complex locus, suppression of transgene-mediated insertional transcriptional activation by cHS4 could not be adequately explained by models that predict that cHS4 can only suppress expression through an enhancer-blocking activity that requires intercalation between an enhancer and a promoter. This has important implications for our theoretical understanding of the possible effects of the insertion of cHS4 on gene therapy vectors. We also show that cHS4 decreased the level of expression of the globin transgene. Therefore, the benefits of partially preventing insertional gene activation are in part negated by the lower expression level of the transgene. A cost/benefit analysis of the utility of incorporation of insulators in gene therapy vectors will require further studies in which the effects of insulators on both the therapeutic gene and the flanking genes are determined at a large number of integration sites. Identification of insulators with minimal promoter specificity would also be of great value.


Subject(s)
Gene Expression Regulation , Mutagenesis, Insertional , Transcriptional Activation , Transgenes , beta-Globins/genetics , Animals , Cell Separation , Chickens , Flow Cytometry , Gene Silencing , Genetic Techniques , Genetic Therapy/methods , Hemoglobinopathies/genetics , Hemoglobinopathies/therapy , Mutagenesis , Reverse Transcriptase Polymerase Chain Reaction , beta-Globins/metabolism
20.
J Cell Biol ; 180(5): 897-904, 2008 Mar 10.
Article in English | MEDLINE | ID: mdl-18332216

ABSTRACT

Human embryonic stem cells (hESCs) are potential therapeutic tools and models of human development. With a growing interest in primary cilia in signal transduction pathways that are crucial for embryological development and tissue differentiation and interest in mechanisms regulating human hESC differentiation, demonstrating the existence of primary cilia and the localization of signaling components in undifferentiated hESCs establishes a mechanistic basis for the regulation of hESC differentiation. Using electron microscopy (EM), immunofluorescence, and confocal microscopies, we show that primary cilia are present in three undifferentiated hESC lines. EM reveals the characteristic 9 + 0 axoneme. The number and length of cilia increase after serum starvation. Important components of the hedgehog (Hh) pathway, including smoothened, patched 1 (Ptc1), and Gli1 and 2, are present in the cilia. Stimulation of the pathway results in the concerted movement of Ptc1 out of, and smoothened into, the primary cilium as well as up-regulation of GLI1 and PTC1. These findings show that hESCs contain primary cilia associated with working Hh machinery.


Subject(s)
Cell Differentiation/genetics , Cilia/ultrastructure , Embryonic Stem Cells/ultrastructure , Hedgehog Proteins/metabolism , Signal Transduction/genetics , Axoneme/genetics , Axoneme/metabolism , Axoneme/ultrastructure , Cell Line , Cell Lineage/genetics , Cilia/genetics , Cilia/metabolism , Culture Media, Serum-Free/pharmacology , Embryonic Stem Cells/metabolism , Female , Fluorescent Antibody Technique , Hedgehog Proteins/genetics , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Microscopy, Confocal , Microscopy, Electron, Transmission , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc Finger Protein GLI1 , Zinc Finger Protein Gli2
SELECTION OF CITATIONS
SEARCH DETAIL
...