Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Clin Dysmorphol ; 33(1): 43-49, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37865865

ABSTRACT

Argininosuccinate lyase (ASL) deficiency is an autosomal recessive disorder of the urea cycle with a diverse spectrum of clinical presentation that is detectable in newborn screening. We report an 8-year-old girl with ASL deficiency who was detected through newborn screening and was confirmed using biochemical and functional assay. She is compound heterozygous for a likely pathogenic variant NM_000048.4(ASL):c.283C>T (p.Arg95Cys) and a likely benign variant NM_000048.4(ASL): c.1319T>C (p.Leu440Pro). Functional characterisation of the likely benign genetic variant in ASL was performed. Genomic sequencing was performed on the index patient presenting with non-specific symptoms of poor feeding and lethargy and shown to have increased serum and urine argininosuccinic acid. Functional assay using HEK293T cell model was performed. ASL enzymatic activity was reduced for Leu440Pro. This study highlights the role of functional testing of a variant that may appear benign in a patient with a phenotype consistent with ASL deficiency, and reclassifies NM_000048.4(ASL): c.1319T>C (p.Leu440Pro) variant as likely pathogenic.


Subject(s)
Argininosuccinic Aciduria , Infant, Newborn , Female , Humans , Child , Argininosuccinic Aciduria/diagnosis , Argininosuccinic Aciduria/genetics , Argininosuccinate Lyase/genetics , Argininosuccinate Lyase/chemistry , Argininosuccinate Lyase/metabolism , Neonatal Screening , HEK293 Cells , Base Sequence
2.
Nat Commun ; 14(1): 563, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36732506

ABSTRACT

Engineered T cells transiently expressing tumor-targeting receptors are an attractive form of engineered T cell therapy as they carry no risk of insertional mutagenesis or long-term adverse side-effects. However, multiple rounds of treatment are often required, increasing patient discomfort and cost. To mitigate this, we sought to improve the antitumor activity of transient engineered T cells by screening a panel of small molecules targeting epigenetic regulators for their effect on T cell cytotoxicity. Using a model for engineered T cells targetting hepatocellular carcinoma, we find that short-term inhibition of G9a/GLP increases T cell antitumor activity in in vitro models and an orthotopic mouse model. G9a/GLP inhibition increases granzyme expression without terminal T cell differentiation or exhaustion and results in specific changes in expression of genes and proteins involved in pro-inflammatory pathways, T cell activation and cytotoxicity.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Mice , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/therapy , T-Lymphocytes , Liver Neoplasms/genetics , Liver Neoplasms/therapy , Disease Models, Animal
3.
Cell Mol Life Sci ; 80(1): 4, 2022 Dec 07.
Article in English | MEDLINE | ID: mdl-36477411

ABSTRACT

Excessive fat accumulation in the liver has become a major health threat worldwide. Unresolved fat deposition in the liver can go undetected until it develops into fatty liver disease, followed by steatohepatitis, fibrosis, cirrhosis, and eventually hepatocellular carcinoma. Lipid deposition in the liver is governed by complex communication, primarily between metabolic organs. This can be mediated by hormones, organokines, and also, as has been more recently discovered, metabolites. Although how metabolites from peripheral organs affect the liver is well documented, the effect of metabolic players released from the liver during the development of fatty liver disease or associated comorbidities needs further attention. Here we focus on interorgan crosstalk based on metabolites released from the liver and how these molecules act as signaling molecules in peripheral tissues. Due to the liver's specific role, we are covering lipid and bile mechanism-derived metabolites. We also discuss the high sucrose intake associated with uric acid release from the liver. Excessive fat deposition in the liver during fatty liver disease development reflects disrupted metabolic processes. As a response, the liver secretes a variety of signaling molecules as well as metabolites which act as a footprint of the metabolic disruption. In the coming years, the reciprocal exchange of metabolites between the liver and other metabolic organs will gain further importance and will help to better understand the development of fatty liver disease and associated diseases.


Subject(s)
Liver Diseases , Humans , Lipids
4.
APL Bioeng ; 5(4): 041502, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34632251

ABSTRACT

Despite diagnostic and therapeutic advances, liver cancer kills more than 18 million people every year worldwide, urging new strategies to model the disease and to improve the current therapeutic options. In vitro tumor models of human cancer continue to evolve, and they represent an important screening tool. However, there is a tremendous need to improve the physiological relevance and reliability of these in vitro models to fulfill today's research requirements for better understanding of cancer progression and treatment options at different stages of the disease. This review describes the hepatocellular carcinoma microenvironmental characteristics and illustrates the current immunotherapy strategy to fight the disease. Moreover, we present a recent collection of 2D and 3D in vitro liver cancer models and address the next generation of in vitro systems recapitulating the tumor microenvironment complexity in more detail.

5.
Elife ; 92020 12 21.
Article in English | MEDLINE | ID: mdl-33345777

ABSTRACT

Cell cycle progression and lipid metabolism are well-coordinated processes required for proper cell proliferation. In liver diseases that arise from dysregulated lipid metabolism, proliferation is diminished. To study the outcome of CDK1 loss and blocked hepatocyte proliferation on lipid metabolism and the consequent impact on whole-body physiology, we performed lipidomics, metabolomics, and RNA-seq analyses on a mouse model. We observed reduced triacylglycerides in liver of young mice, caused by oxidative stress that activated FOXO1 to promote expression of Pnpla2/ATGL. Additionally, we discovered that hepatocytes displayed malfunctioning ß-oxidation, reflected by increased acylcarnitines (ACs) and reduced ß-hydroxybutyrate. This led to elevated plasma free fatty acids (FFAs), which were transported to the adipose tissue for storage and triggered greater insulin secretion. Upon aging, chronic hyperinsulinemia resulted in insulin resistance and hepatic steatosis through activation of LXR. Here, we demonstrate that loss of hepatocyte proliferation is not only an outcome but also possibly a causative factor for liver pathology.


Subject(s)
CDC2 Protein Kinase/physiology , Cell Division/physiology , Hepatocytes/physiology , Hyperinsulinism/metabolism , Lipid Metabolism , Adipose Tissue/chemistry , Adipose Tissue/metabolism , Animals , CDC2 Protein Kinase/metabolism , Fatty Acids, Nonesterified/blood , Fatty Acids, Nonesterified/metabolism , Hepatocytes/metabolism , Insulin Resistance , Liver/chemistry , Liver/metabolism , Liver/physiology , Liver Diseases/etiology , Liver Diseases/metabolism , Male , Mice , Mice, Knockout , Oxidative Stress
6.
PLoS Genet ; 16(11): e1009084, 2020 11.
Article in English | MEDLINE | ID: mdl-33147210

ABSTRACT

The liver possesses a remarkable regenerative capacity based partly on the ability of hepatocytes to re-enter the cell cycle and divide to replace damaged cells. This capability is substantially reduced upon chronic damage, but it is not clear if this is a cause or consequence of liver disease. Here, we investigate whether blocking hepatocyte division using two different mouse models affects physiology as well as clinical liver manifestations like fibrosis and inflammation. We find that in P14 Cdk1Liv-/- mice, where the division of hepatocytes is abolished, polyploidy, DNA damage, and increased p53 signaling are prevalent. Cdk1Liv-/- mice display classical markers of liver damage two weeks after birth, including elevated ALT, ALP, and bilirubin levels, despite the lack of exogenous liver injury. Inflammation was further studied using cytokine arrays, unveiling elevated levels of CCL2, TIMP1, CXCL10, and IL1-Rn in Cdk1Liv-/- liver, which resulted in increased numbers of monocytes. Ablation of CDK2-dependent DNA re-replication and polyploidy in Cdk1Liv-/- mice reversed most of these phenotypes. Overall, our data indicate that blocking hepatocyte division induces biological processes driving the onset of the disease phenotype. It suggests that the decrease in hepatocyte division observed in liver disease may not only be a consequence of fibrosis and inflammation, but also a pathological cue.


Subject(s)
Cell Division/physiology , Hepatocytes/physiology , Liver Cirrhosis/metabolism , Animals , Apoptosis/physiology , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Cycle , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Fibrosis/physiopathology , Hepatitis/metabolism , Hepatitis/physiopathology , Hepatocytes/metabolism , Inflammation/pathology , Liver/metabolism , Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Male , Mice , Mice, Knockout , Signal Transduction
7.
Mol Omics ; 16(5): 436-447, 2020 10 12.
Article in English | MEDLINE | ID: mdl-32519713

ABSTRACT

We have developed MetaboKit, a comprehensive software package for compound identification and relative quantification in mass spectrometry-based untargeted metabolomics analysis. In data dependent acquisition (DDA) analysis, MetaboKit constructs a customized spectral library with compound identities from reference spectral libraries, adducts, dimers, in-source fragments (ISF), MS/MS fragmentation spectra, and more importantly the retention time information unique to the chromatography system used in the experiment. Using the customized library, the software performs targeted peak integration for precursor ions in DDA analysis and for precursor and product ions in data independent acquisition (DIA) analysis. With its stringent identification algorithm requiring matches by both MS and MS/MS data, MetaboKit provides identification results with significantly greater specificity than the competing software packages without loss in sensitivity. The proposed MS/MS-based screening of ISFs also reduces the chance of unverifiable identification of ISFs considerably. MetaboKit's quantification module produced peak area values highly correlated with known concentrations in a DIA analysis of the metabolite standards at both MS1 and MS2 levels. Moreover, the analysis of Cdk1Liv-/- mouse livers showed that MetaboKit can identify a wide range of lipid species and their ISFs, and quantitatively reconstitute the well-characterized fatty liver phenotype in these mice. In DIA data, the MS1-level and MS2-level peak area data produced similar fold change estimates in the differential abundance analysis, and the MS2-level peak area data allowed for quantitative comparisons in compounds whose precursor ion chromatogram was too noisy for peak integration.


Subject(s)
Data Mining , Metabolomics , Software , Animals , Liver/metabolism , Mice, Knockout , Reference Standards , Tandem Mass Spectrometry
8.
J Mol Cell Biol ; 10(2): 161-174, 2018 04 01.
Article in English | MEDLINE | ID: mdl-28992061

ABSTRACT

Myogenic differentiation is accompanied by alterations in the chromatin states, which permit or restrict the transcriptional machinery and thus impact distinctive gene expression profiles. The mechanisms by which higher-order chromatin remodeling is associated with gene activation and silencing during differentiation is not fully understood. In this study, we provide evidence that the euchromatic lysine methyltransferase GLP regulates heterochromatin organization and myogenic differentiation. Interestingly, GLP represses expression of the methyl-binding protein MeCP2 that induces heterochromatin clustering during differentiation. Consequently, MeCP2 and HP1γ localization at major satellites are altered upon modulation of GLP expression. In GLP knockdown cells, depletion of MeCP2 restored both chromatin organization and myogenic differentiation. These results identify a novel regulatory axis between a histone methylation writer and DNA methylation reader, which is important for heterochromatin organization during differentiation.


Subject(s)
Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Muscle Development , Animals , Ankyrin Repeat , Cell Differentiation , Cell Line , Heterochromatin/ultrastructure , Histone-Lysine N-Methyltransferase/analysis , Methyl-CpG-Binding Protein 2/analysis , Mice , Protein Domains
9.
Sci Rep ; 6: 34163, 2016 Sep 26.
Article in English | MEDLINE | ID: mdl-27667720

ABSTRACT

In this study, we demonstrate that the lysine methyltransferase G9a inhibits sarcomere organization through regulation of the MEF2C-HDAC5 regulatory axis. Sarcomeres are essential for muscle contractile function. Presently, skeletal muscle disease and dysfunction at the sarcomere level has been associated with mutations of sarcomere proteins. This study provides evidence that G9a represses expression of several sarcomere genes and its over-expression disrupts sarcomere integrity of skeletal muscle cells. G9a inhibits MEF2C transcriptional activity that is essential for expression of sarcomere genes. Through protein interaction assays, we demonstrate that G9a interacts with MEF2C and its co-repressor HDAC5. In the presence of G9a, calcium signaling-dependent phosphorylation and export of HDAC5 to the cytoplasm is blocked which likely results in enhanced MEF2C-HDAC5 association. Activation of calcium signaling or expression of constitutively active CaMK rescues G9a-mediated repression of HDAC5 shuttling as well as sarcomere gene expression. Our results demonstrate a novel epigenetic control of sarcomere assembly and identifies new therapeutic avenues to treat skeletal and cardiac myopathies arising from compromised muscle function.

10.
J Pathol ; 240(3): 269-281, 2016 11.
Article in English | MEDLINE | ID: mdl-27453350

ABSTRACT

Alveolar rhabdomyosarcoma (ARMS) is an aggressive paediatric cancer of skeletal muscle with poor prognosis. A PAX3-FOXO1 fusion protein acts as a driver of malignancy in ARMS by disrupting tightly coupled but mutually exclusive pathways of proliferation and differentiation. While PAX3-FOXO1 is an attractive therapeutic target, no current treatments are designed to block its oncogenic activity. The present work shows that the histone acetyltransferase P/CAF (KAT2B) is overexpressed in primary tumours from ARMS patients. Interestingly, in fusion-positive ARMS cell lines, P/CAF acetylates and stabilizes PAX3-FOXO1 rather than MyoD, a master regulator of muscle differentiation. Silencing P/CAF, or pharmacological inhibition of its acetyltransferase activity, down-regulates PAX3-FOXO1 levels concomitant with reduced proliferation and tumour burden in xenograft mouse models. Our studies identify a P/CAF-PAX3-FOXO1 signalling node that promotes oncogenesis and may contribute to MyoD dysfunction in ARMS. This work exemplifies the therapeutic potential of targeting chromatin-modifying enzymes to inhibit fusion oncoproteins that are a frequent event in sarcomas. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Oncogene Proteins, Fusion/metabolism , Paired Box Transcription Factors/metabolism , Protein Processing, Post-Translational , Rhabdomyosarcoma, Alveolar/genetics , p300-CBP Transcription Factors/metabolism , Animals , Carcinogenesis/pathology , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Epigenomics , Gene Silencing , Heterografts , Mice , Mice, Nude , Muscles/pathology , MyoD Protein/genetics , MyoD Protein/metabolism , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Fusion/genetics , Paired Box Transcription Factors/genetics , Rhabdomyosarcoma, Alveolar/pathology , Signal Transduction , p300-CBP Transcription Factors/genetics
11.
Nucleic Acids Res ; 44(17): 8129-43, 2016 09 30.
Article in English | MEDLINE | ID: mdl-27229136

ABSTRACT

Differentiation of skeletal muscle cells, like most other cell types, requires a permanent exit from the cell cycle. The epigenetic programming underlying these distinct cellular states is not fully understood. In this study, we provide evidence that the lysine methyltransferase G9a functions as a central axis to regulate proliferation and differentiation of skeletal muscle cells. Transcriptome analysis of G9a knockdown cells revealed deregulation of many cell cycle regulatory genes. We demonstrate that G9a enhances cellular proliferation by two distinct mechanisms. G9a blocks cell cycle exit via methylation-dependent transcriptional repression of the MyoD target genes p21(Cip/Waf1) and Rb1. In addition, it activates E2F1-target genes in a methyltransferase activity-independent manner. We show that G9a is present in the E2F1/PCAF complex, and enhances PCAF occupancy and histone acetylation marks at E2F1-target promoters. Interestingly, G9a preferentially associates with E2F1 at the G1/S phase and with MyoD at the G2/M phase. Our results provide evidence that G9a functions both as a co-activator and a co-repressor to enhance cellular proliferation and inhibit myogenic differentiation.


Subject(s)
Cell Cycle , Cell Differentiation , Histone-Lysine N-Methyltransferase/metabolism , Muscle Development , Animals , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , E2F1 Transcription Factor/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Lysine/metabolism , Methylation , Mice, Inbred C57BL , Muscle Development/genetics , MyoD Protein/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Promoter Regions, Genetic , p300-CBP Transcription Factors/metabolism
12.
Oxid Med Cell Longev ; 2016: 6842568, 2016.
Article in English | MEDLINE | ID: mdl-26798425

ABSTRACT

Oxidative stress is a loss of balance between the production of reactive oxygen species during cellular metabolism and the mechanisms that clear these species to maintain cellular redox homeostasis. Increased oxidative stress has been associated with muscular dystrophy, and many studies have proposed mechanisms that bridge these two pathological conditions at the molecular level. In this review, the evidence indicating a causal role of oxidative stress in the pathogenesis of various muscular dystrophies is revisited. In particular, the mediation of cellular redox status in dystrophic muscle by NF-κB pathway, autophagy, telomere shortening, and epigenetic regulation are discussed. Lastly, the current stance of targeting these pathways using antioxidant therapies in preclinical and clinical trials is examined.


Subject(s)
Muscle, Skeletal/pathology , Muscular Atrophy/therapy , Oxidative Stress , Animals , Humans , Models, Biological , Reactive Oxygen Species/metabolism , Signal Transduction
13.
Stem Cells Dev ; 25(1): 55-67, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26423361

ABSTRACT

Pluripotent cells are promising tools in the arena of regenerative medicine. For many years, research efforts have been directed toward uncovering the underlying mechanisms that govern the pluripotent state and this involves identifying new pluripotency-associated factors. Zinc finger protein 553 (Zfp553) has been hypothesized to be one such factor because of its predominant expression in inner cell mass of the mouse early embryo. In this study, we have identified Zfp553 as a regulator of pluripotency. Zfp553 knockdown downregulates pluripotency markers and triggers differentiation in mouse embryonic stem cells (mESCs). Further investigation revealed that Zfp553 regulates pluripotency in mESCs through the transcriptional activation of Pou5f1 and Nanog. Microarray results revealed that depletion of Zfp553 downregulates many pluripotency genes, as well as genes associated with metabolism-related processes. ChIP-sequencing (ChIP-seq) depicted the genomic binding sites of Zfp553 in mESCs and its binding motif. In addition, we found that depletion of Zfp553 could impair somatic cell reprogramming, evidenced by reduced reprogramming efficiency and cell viability. Together, our preliminary findings provide novel insights to a newly identified pluripotency factor Zfp553 and its role in pluripotency regulation.


Subject(s)
Cell Proliferation/genetics , Cellular Reprogramming/genetics , DNA-Binding Proteins/physiology , Induced Pluripotent Stem Cells/metabolism , Nuclear Proteins/physiology , Animals , Cell Differentiation/genetics , Cells, Cultured , Embryonic Stem Cells/physiology , Energy Metabolism/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Induced Pluripotent Stem Cells/physiology , Mice , Microarray Analysis , Zinc Fingers
14.
Sci Rep ; 4: 7519, 2014 Dec 17.
Article in English | MEDLINE | ID: mdl-25515777

ABSTRACT

The acquisition of pluripotent cells can be achieved by combined overexpression of transcription factors Oct4, Klf4, Sox2 and c-Myc in somatic cells. This cellular reprogramming process overcomes various barriers to re-activate pluripotency genes and re-acquire the highly dynamic pluripotent chromatin status. Many genetic and epigenetic factors are essentially involved in the reprogramming process. We previously reported that Patz1 is required for maintenance of ES cell identity. Here we report that Patz1 plays an inhibitory role in OKSM-induced reprogramming process since more iPS colonies can be induced from Patz1(+/-) MEFs than wild type MEFs; while the addition of Patz1 significantly repressed reprogramming efficiency. Patz1(+/-) MEFs can surpass the senescence barrier of Ink4a/Arf locus, thus enhancing iPS colonies formation. Moreover, Patz1(+/-) MEFs displayed higher levels of acetylated histone H3, H3K4me2, H3K4me3, H3K36me3 and lower levels of histone H3K9me3 and HP1α, indicating that heterozygous knockout of Patz1 results in a globally open chromatin which is more accessible for transcriptional activation. However, Patz1(-/-) MEFs gave the lowest reprogramming efficiency which may result from cell senescence trigged by up-regulated Ink4a/Arf locus. Together, we have demonstrated that the dosage of Patz1 modulates reprogramming process via significantly influencing cell senescence, proliferation and chromatin structure.


Subject(s)
Cellular Reprogramming/genetics , Kruppel-Like Transcription Factors/genetics , Animals , Cell Line , Cellular Reprogramming/physiology , Cellular Senescence/genetics , Cellular Senescence/physiology , Chromatin/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/physiology , Fibroblasts/metabolism , Fibroblasts/physiology , Histones/genetics , Kruppel-Like Factor 4 , Mice , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/physiology , Transcriptional Activation/genetics
15.
Curr Top Dev Biol ; 110: 317-38, 2014.
Article in English | MEDLINE | ID: mdl-25248481

ABSTRACT

Stra13 and Sharp-1 are transcriptional repressors that share domain structure similarity with members of the basic helix-loop-helix-Orange subfamily. In contrast to other members that include Hes and Hey proteins, transcriptional repression mediated by Stra13 and Sharp-1 does not involve recruitment of the corepressor Groucho. Both proteins undergo sumoylation at evolutionarily conserved sites, and this posttranslational modification serves as a platform for association with chromatin-modifying enzymes including histone deacetylases and histone methyltransferases. In addition to being widely expressed during embryonic development and in adult tissues, the expression of both genes is induced by a number of stimuli. Loss-of-function and gain-of-function studies have demonstrated their function in cellular differentiation and regeneration, in regulation of circadian rhythms, immune homeostasis, and metabolism. Given their diverse physiological functions in several tissues, it is not surprising that deregulated expression of Stra13 and Sharp-1 is apparent in human pathologies. Here, we review our current understanding of their cellular functions that suggest a requirement in maintenance of tissue homeostasis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Homeodomain Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Animals , Apoptosis/genetics , Cell Cycle Checkpoints , Cell Differentiation , DNA Repair/physiology , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Histone Deacetylases/metabolism , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Homeodomain Proteins/metabolism , Humans , Hypoxia/metabolism , Immune System/metabolism , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/genetics , Sumoylation , Transcription Factors/metabolism
16.
Stem Cells Dev ; 23(10): 1062-73, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24380431

ABSTRACT

Embryonic stem cells (ESCs) derived from the inner cell mass (ICM) of blastocysts are pluripotent. Pluripotency is maintained by a transcriptional network in which Oct4 and Nanog are master regulators. Notably, several zinc finger transcription factors have important roles in this network. Patz1, a BTB/POZ-domain-containing zinc finger protein, is expressed at higher levels in the ICM relative to the trophectoderm. However, its function in pluripotency has been poorly studied. Here, we show that Patz1 is an important regulator of pluripotency in ESCs. Patz1 RNAi, chromatin immunoprecipitation (ChIP), and reporter assays indicate that Patz1 directly regulates Pou5f1 and Nanog. Global transcriptome changes upon Patz1 knockdown largely involve upregulation of apoptotic genes and downregulation of cell cycle and cellular metabolism genes. Patz1 ChIP sequencing further identified more than 5,000 binding sites of Patz1 in mouse genome, from which two binding motifs were extracted. Further, gene ontology analysis of genes associated with the binding sites displays enrichment for proximity to developmental genes. In addition, embryoid body assays suggest that Patz1 represses developmental genes. Together, these results propose that Patz1 is important for ESC pluripotency.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Expression Regulation/physiology , Neoplasm Proteins/metabolism , Pluripotent Stem Cells/metabolism , Repressor Proteins/metabolism , Animals , Cell Line , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Embryonic Stem Cells/cytology , Homeodomain Proteins/biosynthesis , Mice , Nanog Homeobox Protein , Octamer Transcription Factor-3/biosynthesis , Pluripotent Stem Cells/cytology , Response Elements/physiology
17.
Epigenetics ; 8(1): 16-22, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23257913

ABSTRACT

Lysine methylation of histone and non-histone substrates by the methyltransferase G9a is mostly associated with transcriptional repression. Recent studies, however, have highlighted its role as an activator of gene expression through mechanisms that are independent of its methyltransferase activity. Here we review the growing repertoire of molecular mechanisms and substrates through which G9a regulates gene expression. We also discuss emerging evidence for its wide-ranging functions in development, pluripotency, cellular differentiation and cell cycle regulation that underscore the complexity of its functions. The deregulated expression of G9a in cancers and other human pathologies suggests that it may be a viable therapeutic target in various diseases.


Subject(s)
Gene Expression Regulation , Histone-Lysine N-Methyltransferase/metabolism , Animals , Cellular Senescence , Histone-Lysine N-Methyltransferase/chemistry , Humans , Neoplasms/enzymology , Neoplasms/pathology , Repressor Proteins/metabolism , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...