Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Neuron ; 112(2): 247-263.e6, 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-37924811

ABSTRACT

Oligodendrocytes are the primary producers of many extracellular matrix (ECM)-related proteins found in the CNS. Therefore, oligodendrocytes play a critical role in the determination of brain stiffness, node of Ranvier formation, perinodal ECM deposition, and perineuronal net formation, all of which depend on the ECM. Nevertheless, the transcription factors that control ECM-related gene expression in oligodendrocytes remain unknown. Here, we found that the transcription factor Osterix (also known as Sp7) binds in proximity to genes important for CNS ECM and node of Ranvier formation and mediates their expression. Oligodendrocyte-specific ablation of Sp7 changes ECM composition and brain stiffness and results in aberrant node of Ranvier formation. Sp7 is known to control osteoblast maturation and bone formation. Our comparative analyses suggest that Sp7 plays a conserved biological role in oligodendrocytes and in bone-forming cells, where it mediates brain and bone tissue stiffness by controlling expression of ECM components.


Subject(s)
Oligodendroglia , Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Oligodendroglia/physiology , Extracellular Matrix/metabolism , Bone and Bones/metabolism , Extracellular Matrix Proteins/metabolism , Gene Expression
2.
Cancers (Basel) ; 15(16)2023 Aug 18.
Article in English | MEDLINE | ID: mdl-37627196

ABSTRACT

Sarcoma classification is challenging and can lead to treatment delays. Previous studies used DNA aberrations and machine-learning classifiers based on methylation profiles for diagnosis. We aimed to classify sarcomas by analyzing methylation signatures obtained from low-coverage whole-genome sequencing, which also identifies copy-number alterations. DNA was extracted from 23 suspected sarcoma samples and sequenced on an Oxford Nanopore sequencer. The methylation-based classifier, applied in the nanoDx pipeline, was customized using a reference set based on processed Illumina-based methylation data. Classification analysis utilized the Random Forest algorithm and t-distributed stochastic neighbor embedding, while copy-number alterations were detected using a designated R package. Out of the 23 samples encompassing a restricted range of sarcoma types, 20 were successfully sequenced, but two did not contain tumor tissue, according to the pathologist. Among the 18 tumor samples, 14 were classified as reported in the pathology results. Four classifications were discordant with the pathological report, with one compatible and three showing discrepancies. Improving tissue handling, DNA extraction methods, and detecting point mutations and translocations could enhance accuracy. We envision that rapid, accurate, point-of-care sarcoma classification using nanopore sequencing could be achieved through additional validation in a diverse tumor cohort and the integration of methylation-based classification and other DNA aberrations.

3.
Genome Biol ; 23(1): 158, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35841107

ABSTRACT

The Oxford Nanopore (ONT) platform provides portable and rapid genome sequencing, and its ability to natively profile DNA methylation without complex sample processing is attractive for point-of-care real-time sequencing. We recently demonstrated ONT shallow whole-genome sequencing to detect copy number alterations (CNAs) from the circulating tumor DNA (ctDNA) of cancer patients. Here, we show that cell type and cancer-specific methylation changes can also be detected, as well as cancer-associated fragmentation signatures. This feasibility study suggests that ONT shallow WGS could be a powerful tool for liquid biopsy.


Subject(s)
Cell-Free Nucleic Acids , Circulating Tumor DNA , Nanopore Sequencing , Neoplasms , DNA Methylation , High-Throughput Nucleotide Sequencing , Humans , Neoplasms/genetics
4.
RNA Biol ; 19(1): 333-352, 2022.
Article in English | MEDLINE | ID: mdl-35220879

ABSTRACT

Latent 5' splice sites, not normally used, are highly abundant in human introns, but are activated under stress and in cancer, generating thousands of nonsense mRNAs. A previously proposed mechanism to suppress latent splicing was shown to be independent of NMD, with a pivotal role for initiator-tRNA independent of protein translation. To further elucidate this mechanism, we searched for nuclear proteins directly bound to initiator-tRNA. Starting with UV-crosslinking, we identified nucleolin (NCL) interacting directly and specifically with initiator-tRNA in the nucleus, but not in the cytoplasm. Next, we show the association of ini-tRNA and NCL with pre-mRNA. We further show that recovery of suppression of latent splicing by initiator-tRNA complementation is NCL dependent. Finally, upon nucleolin knockdown we show activation of latent splicing in hundreds of coding transcripts having important cellular functions. We thus propose nucleolin, a component of the endogenous spliceosome, through its direct binding to initiator-tRNA and its effect on latent splicing, as the first protein of a nuclear quality control mechanism regulating splice site selection to protect cells from latent splicing that can generate defective mRNAs.


Subject(s)
Binding Sites , Phosphoproteins/metabolism , RNA Splice Sites , RNA Splicing , RNA-Binding Proteins/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Gene Knockdown Techniques , Humans , Mass Spectrometry , Protein Binding , RNA Interference , RNA, Transfer/genetics , Nucleolin
6.
Sci Rep ; 9(1): 18795, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31827209

ABSTRACT

ERBB2 amplification is a prognostic marker for aggressive tumors and a predictive marker for prolonged survival following treatment with HER2 inhibitors. We attempt to sub-group HER2+ tumors based on amplicon structures and co-amplified genes. We examined five HER2+ cell lines, three HER2+ xenographs and 57 HER2+ tumor tissues. ERBB2 amplification was analyzed using digital droplet PCR and low coverage whole genome sequencing. In some HER2+ tumors PPM1D, that encodes WIP1, is co-amplified. Cell lines were treated with HER2 and WIP1 inhibitors. We find that inverted duplication is the amplicon structure in the majority of HER2+ tumors. In patients suffering from an early stage disease the ERBB2 amplicon is composed of a single segment while in patients suffering from advanced cancer the amplicon is composed of several different segments. We find robust WIP1 inhibition in some HER2+ PPM1D amplified cell lines. Sub-grouping HER2+ tumors using low coverage whole genome sequencing identifies inverted duplications as the main amplicon structure and based on the number of segments, differentiates between local and advanced tumors. In addition, we found that we could determine if a tumor is a recurrent tumor or second primary tumor and identify co-amplified oncogenes that may serve as targets for therapy.


Subject(s)
Gene Amplification , Neoplasms/classification , Receptor, ErbB-2/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Disease Progression , Enzyme Inhibitors/pharmacology , Female , Genes, erbB-2 , Humans , Male , Middle Aged , Neoplasms/genetics , Polymerase Chain Reaction , Protein Phosphatase 2C/antagonists & inhibitors , Protein Phosphatase 2C/genetics , Whole Genome Sequencing , Young Adult
7.
Cell Rep ; 23(8): 2254-2263, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29791837

ABSTRACT

Zinc finger protein ZFP24, formerly known as ZFP191, is essential for oligodendrocyte maturation and CNS myelination. Nevertheless, the mechanism by which ZFP24 controls these processes is unknown. We demonstrate that ZFP24 binds to a consensus DNA sequence in proximity to genes important for oligodendrocyte differentiation and CNS myelination, and we show that this binding enhances target gene expression. We also demonstrate that ZFP24 DNA binding is controlled by phosphorylation. Phosphorylated ZFP24, which does not bind DNA, is the predominant form in oligodendrocyte progenitor cells. As these cells mature into oligodendrocytes, the non-phosphorylated, DNA-binding form accumulates. Interestingly, ZFP24 displays overlapping genomic binding sites with the transcription factors MYRF, SOX10, and OLIG2, which are known to control oligodendrocyte differentiation. Our findings provide a mechanism by which dephosphorylation of ZFP24 mediates its binding to regulatory regions of genes important for oligodendrocyte maturation, controls their expression, and thereby regulates oligodendrocyte differentiation and CNS myelination.


Subject(s)
Carrier Proteins/metabolism , Cell Differentiation , Oligodendroglia/cytology , Oligodendroglia/metabolism , Alanine/genetics , Animals , Base Sequence , Cell Lineage , DNA/metabolism , HEK293 Cells , Humans , Mice, Inbred C57BL , Phosphorylation , Protein Binding , Rats
8.
Diabetes ; 65(7): 2081-93, 2016 07.
Article in English | MEDLINE | ID: mdl-26993067

ABSTRACT

The molecular program underlying infrequent replication of pancreatic ß-cells remains largely inaccessible. Using transgenic mice expressing green fluorescent protein in cycling cells, we sorted live, replicating ß-cells and determined their transcriptome. Replicating ß-cells upregulate hundreds of proliferation-related genes, along with many novel putative cell cycle components. Strikingly, genes involved in ß-cell functions, namely, glucose sensing and insulin secretion, were repressed. Further studies using single-molecule RNA in situ hybridization revealed that in fact, replicating ß-cells double the amount of RNA for most genes, but this upregulation excludes genes involved in ß-cell function. These data suggest that the quiescence-proliferation transition involves global amplification of gene expression, except for a subset of tissue-specific genes, which are "left behind" and whose relative mRNA amount decreases. Our work provides a unique resource for the study of replicating ß-cells in vivo.


Subject(s)
Cell Division/genetics , Cell Proliferation/genetics , Insulin-Secreting Cells/metabolism , Transcriptome , Animals , Flow Cytometry , Gene Expression Regulation , Insulin-Secreting Cells/cytology , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Transgenic
10.
Mol Cancer ; 14: 167, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26370283

ABSTRACT

BACKGROUND: The SWI/SNF ATP dependent chromatin remodeling complex is a multi-subunit complex, conserved in eukaryotic evolution that facilitates nucleosomal re-positioning relative to the DNA sequence. In recent years the SWI/SNF complex has emerged to play a role in cancer development as various sub-units of the complex are found to be mutated in a variety of tumors. One core-subunit of the complex, which has been well established as a tumor suppressor gene is SMARCB1 (SNF5/INI1/BAF47). Mutation and inactivation of SMARCB1 have been identified as the underlying mechanism leading to Malignant Rhabdoid Tumors (MRT) and Atypical Teratoid/Rhabdoid Tumors (AT/RT), two highly aggressive forms of pediatric neoplasms. METHODS: We present a phosphoproteomic study of Smarcb1 dependent changes in signaling networks. The SILAC (Stable Isotopic Labeling of Amino Acids in Cell Culture) protocol was used to quantify in an unbiased manner any changes in the phosphoproteomic profile of Smarcb1 deficient murine rhabdoid tumor cell lines following Smarcb1 stable re-expression and under different serum conditions. RESULTS: This study illustrates broad changes in the regulation of multiple biological networks including cell cycle progression, chromatin remodeling, cytoskeletal regulation and focal adhesion. Specifically, we identify Smarcb1 dependent changes in phosphorylation and expression of the EGF receptor, demonstrate downstream signaling and show that inhibition of EGFR signaling specifically hinders the proliferation of Smarcb1 deficient cells. CONCLUSIONS: These results support recent findings regarding the effectivity of EGFR inhibitors in hindering the proliferation of human MRT cells and demonstrate that activation of EGFR signaling in Rhabdoid tumors is SMARCB1 dependent.


Subject(s)
Brain Neoplasms/genetics , Chromosomal Proteins, Non-Histone/biosynthesis , ErbB Receptors/genetics , Kidney Neoplasms/genetics , Phosphoproteins/biosynthesis , Rhabdoid Tumor/genetics , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/genetics , Gene Expression Regulation, Neoplastic , Humans , Isotope Labeling , Kidney Neoplasms/pathology , Mice , Phosphoproteins/genetics , Proteomics , Rhabdoid Tumor/pathology , SMARCB1 Protein , Signal Transduction
11.
Mol Cell Endocrinol ; 408: 62-72, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25724481

ABSTRACT

High output of steroid hormone synthesis in steroidogenic cells of the adrenal cortex and the gonads requires the expression of the steroidogenic acute regulatory protein (StAR) that facilitates cholesterol mobilization to the mitochondrial inner membrane where the CYP11A1/P450scc enzyme complex converts the sterol to the first steroid. Earlier studies have shown that StAR is active while pausing on the cytosolic face of the outer mitochondrial membrane while subsequent import of the protein into the matrix terminates the cholesterol mobilization activity. Consequently, during repeated activity cycles, high level of post-active StAR accumulates in the mitochondrial matrix. To prevent functional damage due to such protein overload effect, StAR is degraded by a sequence of three to four ATP-dependent proteases of the mitochondria protein quality control system, including LON and the m-AAA membranous proteases AFG3L2 and SPG7/paraplegin. Furthermore, StAR expression in both peri-ovulatory ovarian cells, or under ectopic expression in cell line models, results in up to 3-fold enrichment of the mitochondrial proteases and their transcripts. We named this novel form of mitochondrial stress as StAR overload response (SOR). To better understand the SOR mechanism at the transcriptional level we analyzed first the unexplored properties of the proximal promoter of the LON gene. Our findings suggest that the human nuclear respiratory factor 2 (NRF-2), also known as GA binding protein (GABP), is responsible for 88% of the proximal promoter activity, including the observed increase of transcription in the presence of StAR. Further studies are expected to reveal if common transcriptional determinants coordinate the SOR induced transcription of all the genes encoding the SOR proteases.


Subject(s)
GA-Binding Protein Transcription Factor/metabolism , Mitochondria/metabolism , Phosphoproteins/metabolism , Protease La/genetics , Stress, Physiological/genetics , Transcriptional Activation/genetics , Animals , Base Sequence , Humans , Molecular Sequence Data
12.
Nucleic Acids Res ; 42(9): 5689-701, 2014 May.
Article in English | MEDLINE | ID: mdl-24682826

ABSTRACT

DNA double-strand breaks (DSBs) are the most severe type of DNA damage. DSBs are repaired by non-homologous end-joining or homology directed repair (HDR). Identifying novel small molecules that affect HDR is of great importance both for research use and therapy. Molecules that elevate HDR may improve gene targeting whereas inhibiting molecules can be used for chemotherapy, since some of the cancers are more sensitive to repair impairment. Here, we performed a high-throughput chemical screen for FDA approved drugs, which affect HDR in cancer cells. We found that HDR frequencies are increased by retinoic acid and Idoxuridine and reduced by the antihypertensive drug Spironolactone. We further revealed that Spironolactone impairs Rad51 foci formation, sensitizes cancer cells to DNA damaging agents, to Poly (ADP-ribose) polymerase (PARP) inhibitors and cross-linking agents and inhibits tumor growth in xenografts, in mice. This study suggests Spironolactone as a new candidate for chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Recombinational DNA Repair/drug effects , Spironolactone/pharmacology , Animals , Antihypertensive Agents/pharmacology , Cell Line, Tumor , DNA Breaks, Double-Stranded , Double-Blind Method , Drug Approval , High-Throughput Screening Assays , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Rad51 Recombinase/metabolism , United States , United States Food and Drug Administration , Xenograft Model Antitumor Assays
13.
Dev Cell ; 23(4): 681-90, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23000141

ABSTRACT

Most adult mammalian tissues are quiescent, with rare cell divisions serving to maintain homeostasis. At present, the isolation and study of replicating cells from their in vivo niche typically involves immunostaining for intracellular markers of proliferation, causing the loss of sensitive biological material. We describe a transgenic mouse strain, expressing a CyclinB1-GFP fusion reporter, that marks replicating cells in the S/G2/M phases of the cell cycle. Using flow cytometry, we isolate live replicating cells from the liver and compare their transcriptome to that of quiescent cells to reveal gene expression programs associated with cell proliferation in vivo. We find that replicating hepatocytes have reduced expression of genes characteristic of liver differentiation. This reporter system provides a powerful platform for gene expression and metabolic and functional studies of replicating cells in their in vivo niche.


Subject(s)
Cell Proliferation , Hepatocytes/cytology , Transcription, Genetic/genetics , Transcriptome , Animals , Biomarkers/analysis , Biomarkers/metabolism , Cell Cycle , Cell Differentiation , Cell Survival , Cyclin B1/genetics , Cyclin B1/metabolism , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hepatocytes/metabolism , Mice , Mice, Transgenic , NIH 3T3 Cells , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction
14.
Stem Cell Rev Rep ; 8(4): 1076-87, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22661117

ABSTRACT

Parthenotes have been proposed as a source of embryonic stem cells but they lack the centriole which is inherited through the sperm in all mammalian species, except for rodents. We investigated the centrosome of parthenotes and parthenogenetic embryonic stem cells using parthenogenetic and biparental pig pre-implantation embryos, human and pig parthenogenetic and biparental embryonic stem cells, sheep fibroblasts derived from post implantation parthenogenetic and biparental embryos developed in vivo. We also determined the level of aneuploidy in parthenogenetic cells. Oocytes of all species were activated using ionomycin and 6-dimethylaminopurine (6-DMAP). Over 60% of parthenogenetic blastomeres were affected by an excessive number of centrioles. Centrosome amplification, was observed by microscopical and ultrastructural analysis also in parthenogenetic cell lines of all three species. Over expression of PLK2 and down regulation of CCNF, respectively involved in the stimulation and inhibition of centrosome duplication, were present in all species. We also detected down regulation of spindle assembly checkpoint components such as BUB1, CENPE and MAD2. Centrosome amplification was accompanied by multipolar mitotic spindles and all cell lines were affected by a high rate of aneuploidy. These observations indicate a link between centrosome amplification and the high incidence of aneuploidy and suggest that parthenogenetic stem cells may be a useful model to investigate how aneuploidy can be compatible with cell proliferation and differentiation.


Subject(s)
Aneuploidy , Blastomeres/metabolism , Centrosome/metabolism , Chromosomal Instability , Parthenogenesis , Spindle Apparatus/metabolism , Animals , Blastomeres/pathology , Calcium Ionophores/adverse effects , Calcium Ionophores/pharmacology , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Centrosome/pathology , Chromosomal Proteins, Non-Histone/metabolism , Humans , Ionomycin/adverse effects , Ionomycin/pharmacology , Mad2 Proteins , Oocytes/metabolism , Oocytes/pathology , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/metabolism , Sheep , Spindle Apparatus/pathology , Swine
15.
Endocrinology ; 152(7): 2589-98, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21521747

ABSTRACT

Understanding the molecular triggers of pancreatic ß-cell proliferation may facilitate the development of regenerative therapies for diabetes. Genetic studies have demonstrated an important role for cyclin D2 in ß-cell proliferation and mass homeostasis, but its specific function in ß-cell division and mechanism of regulation remain unclear. Here, we report that cyclin D2 is present at high levels in the nucleus of quiescent ß-cells in vivo. The major regulator of cyclin D2 expression is glucose, acting via glycolysis and calcium channels in the ß-cell to control cyclin D2 mRNA levels. Furthermore, cyclin D2 mRNA is down-regulated during S-G(2)-M phases of each ß-cell division, via a mechanism that is also affected by glucose metabolism. Thus, glucose metabolism maintains high levels of nuclear cyclin D2 in quiescent ß-cells and modulates the down-regulation of cyclin D2 in replicating ß-cells. These data challenge the standard model for regulation of cyclin D2 during the cell division cycle and suggest cyclin D2 as a molecular link between glucose levels and ß-cell replication.


Subject(s)
Calcium Channels/metabolism , Cell Proliferation , Cyclin D2/metabolism , Glucose/metabolism , Glycolysis , Islets of Langerhans/metabolism , Animals , Calcium Channel Agonists/pharmacology , Cell Cycle , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cyclin D2/genetics , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Glycolysis/drug effects , Humans , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred ICR , Mice, Transgenic , Organ Culture Techniques , RNA, Messenger/metabolism
16.
PLoS One ; 6(2): e17262, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21364955

ABSTRACT

MacroH2A1 is a histone H2A variant which contains a large non-histone C-terminal region of largely unknown function. Within this region is a macro domain which can bind ADP-ribose and related molecules. Most studies of macroH2A1 focus on the involvement of this variant in transcriptional repression. Studies in mouse embryos and in embryonic stem cells suggested that during early development macroH2A can be found at the centrosome. Centrosomal localization of macroH2A was later reported in somatic cells. Here we provide data showing that macroH2A1 does not localize to the centrosome and that the centrosomal signal observed with antibodies directed against the macroH2A1 non-histone region may be the result of antibody cross-reactivity.


Subject(s)
Centrosome/metabolism , Histones/genetics , Histones/metabolism , Animals , Cells, Cultured , Chromatin/metabolism , Gene Knockdown Techniques , Genetic Variation/physiology , Green Fluorescent Proteins/metabolism , Histones/antagonists & inhibitors , Humans , Mice , Mutant Proteins/metabolism , Protein Transport/genetics , Protein Transport/physiology , Recombinant Fusion Proteins/metabolism , Tissue Distribution , Transfection
17.
Nucleic Acids Res ; 39(4): 1326-35, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21030442

ABSTRACT

Promoter hypermethylation and heterochromatinization is a frequent event leading to gene inactivation and tumorigenesis. At the molecular level, inactivation of tumor suppressor genes in cancer has many similarities to the inactive X chromosome in female cells and is defined and maintained by DNA methylation and characteristic histone modifications. In addition, the inactive-X is marked by the histone macroH2A, a variant of H2A with a large non-histone region of unknown function. Studying tumor suppressor genes (TSGs) silenced in cancer cell lines, we find that when active, these promoters are associated with H2A.Z but become enriched for macroH2A1 once silenced. Knockdown of macroH2A1 was not sufficient for reactivation of silenced genes. However, when combined with DNA demethylation, macroH2A1 deficiency significantly enhanced reactivation of the tumor suppressor genes p16, MLH1 and Timp3 and inhibited cell proliferation. Our findings link macroH2A1 to heterochromatin of epigenetically silenced cancer genes and indicate synergism between macroH2A1 and DNA methylation in maintenance of the silenced state.


Subject(s)
DNA Methylation , Gene Silencing , Genes, p16 , Histones/metabolism , Alleles , Cell Line , Cell Line, Tumor , Colonic Neoplasms/genetics , Gene Knockdown Techniques , Genes, Tumor Suppressor , Histones/genetics , Humans , Promoter Regions, Genetic
18.
Stem Cells ; 28(8): 1349-54, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20572015

ABSTRACT

Direct reprogramming procedures reset the epigenetic memory of cells and convert differentiated somatic cells into pluripotent stem cells. In addition to epigenetic memory of cell identity, which is established during development, somatic cells can accumulate abnormal epigenetic changes that can contribute to pathological conditions. Aberrant promoter hypermethylation and epigenetic silencing of tumor suppressor genes (TSGs) are now recognized as an important mechanism in tumor initiation and progression. Here, we have studied the fate of the silenced TSGs p16(CDKN2A) during direct reprogramming. We find that following reprogramming, p16 expression is restored and is stably maintained even when cells are induced to differentiate. Large-scale methylation profiling of donor cells identified aberrant methylation at hundreds of additional sites. Methylation at many, but not all these sites was reversed following reprogramming. Our results suggest that reprogramming approaches may be applied to repair the epigenetic lesions associated with cancer.


Subject(s)
Cellular Reprogramming/physiology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Epigenesis, Genetic/genetics , Cells, Cultured , Cellular Reprogramming/genetics , DNA Methylation/genetics , DNA Methylation/physiology , Gene Silencing/physiology , Humans
19.
Stem Cells ; 27(11): 2686-90, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19711451

ABSTRACT

Genomic imprinting is an epigenetic phenomenon whereby genes are expressed in a monoallelic manner, which is inherited either maternally or paternally. Expression of imprinted genes has been examined in human embryonic stem (ES) cells, and the cells show a substantial degree of genomic imprinting stability. Recently, human somatic cells were reprogrammed to a pluripotent state using various defined factors. These induced pluripotent stem (iPS) cells are thought to have a great potential for studying genetic diseases and to be a source of patient-specific stem cells. Thus, studying the expression of imprinted genes in these cells is important. We examined the allelic expression of various imprinted genes in several iPS cell lines and found polymorphisms in four genes. After analyzing parent-specific expression of these genes, we observed overall normal monoallelic expression in the iPS cell lines. However, we found biallelic expression of the H19 gene in one iPS cell line and biallelic expression of the KCNQ10T1 gene in another iPS cell line. We further analyzed the DNA methylation levels of the promoter region of the H19 gene and found that the cell line that showed biallelic expression had undergone extensive DNA demethylation. Additionally we studied the imprinting gene expression pattern of multiple human iPS cell lines via DNA microarray analyses and divided the pattern of expression into three groups: (a) genes that showed significantly stable levels of expression in iPS cells, (b) genes that showed a substantial degree of variability in expression in both human ES and iPS cells, and (c) genes that showed aberrant expression levels in some human iPS cell lines, as compared with human ES cells. In general, iPS cells have a rather stable expression of their imprinted genes. However, we found a significant number of cell lines with abnormal expression of imprinted genes, and thus we believe that imprinted genes should be examined for each cell line if it is to be used for studying genetic diseases or for the purpose of regenerative medicine.


Subject(s)
Genomic Imprinting/genetics , Induced Pluripotent Stem Cells/metabolism , DNA Methylation/genetics , DNA Methylation/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Models, Genetic , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics
20.
Cancer Cell ; 11(2): 133-46, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17292825

ABSTRACT

Myocardin is known as an important transcriptional regulator in smooth and cardiac muscle development. Here we found that myocardin is frequently repressed during human malignant transformation, contributing to a differentiation defect. We demonstrate that myocardin is a transcriptional target of TGFbeta required for TGFbeta-mediated differentiation of human fibroblasts. Serum deprivation, intact contact inhibition response, and the p16ink4a/Rb pathway contribute to myocardin induction and differentiation. Restoration of myocardin expression in sarcoma cells results in differentiation and inhibition of malignant growth, whereas inactivation of myocardin in normal fibroblasts increases their proliferative potential. Myocardin expression is reduced in multiple types of human tumors. Collectively, our results demonstrate that myocardin is an important suppressive modifier of the malignant transformation process.


Subject(s)
Cell Differentiation , Cell Transformation, Neoplastic , Cyclin-Dependent Kinase Inhibitor p16/antagonists & inhibitors , Fibroblasts/cytology , Nuclear Proteins/antagonists & inhibitors , Trans-Activators/antagonists & inhibitors , Blotting, Western , Cell Adhesion , Cell Proliferation , Cells, Cultured , Colony-Forming Units Assay , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Methylation , Fibroblasts/metabolism , Fluorescent Antibody Technique , Gene Expression Regulation , Humans , Lung/embryology , Mesoderm/cytology , Mesoderm/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Plasmids , Promoter Regions, Genetic , RNA, Small Interfering/pharmacology , Trans-Activators/genetics , Trans-Activators/metabolism , Transforming Growth Factor beta/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...