Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
3.
J Med Genet ; 51(6): 407-12, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24721835

ABSTRACT

BACKGROUND: In a subset of imprinting disorders caused by epimutations, multiple imprinted loci are affected. Familial occurrence of multilocus imprinting disorders is rare. PURPOSE/OBJECTIVE: We have investigated the clinical and molecular features of a familial DNA-methylation disorder. METHODS: Tissues of affected individuals and blood samples of family members were investigated by conventional and molecular karyotyping. Sanger sequencing and RT-PCR of imprinting-associated genes (NLRP2, NLRP7, ZFP57, KHDC3L, DNMT1o), exome sequencing and locus-specific, array-based and genome-wide technologies to determine DNA-methylation were performed. RESULTS: In three offspring of a healthy couple, we observed prenatal onset of severe growth retardation and dysmorphism associated with altered DNA-methylation at paternally and maternally imprinted loci. Array-based analyses in various tissues of the offspring identified the DNA-methylation of 2.1% of the genes in the genome to be recurrently altered. Despite significant enrichment of imprinted genes (OR 9.49), altered DNA-methylation predominately (90.2%) affected genes not known to be imprinted. Sequencing of genes known to cause comparable conditions and exome sequencing in affected individuals and their ancestors did not unambiguously point to a causative gene. CONCLUSIONS: The family presented herein suggests the existence of a familial disorder of DNA-methylation affecting imprinted but also not imprinted gene loci potentially caused by a maternal effect mutation in a hitherto not identified gene.


Subject(s)
DNA Methylation/genetics , Genetic Diseases, Inborn/genetics , Alleles , DNA Mutational Analysis , Epigenomics , Female , Humans , Infant, Newborn , Male , Pedigree
4.
PLoS One ; 8(9): e73288, 2013.
Article in English | MEDLINE | ID: mdl-24023855

ABSTRACT

Sex differences are well known to be determinants of development, health and disease. Epigenetic mechanisms are also known to differ between men and women through X-inactivation in females. We hypothesized that epigenetic sex differences may also result from sex hormone functions, in particular from long-lasting androgen programming. We aimed at investigating whether inactivation of the androgen receptor, the key regulator of normal male sex development, is associated with differences of the patterns of DNA methylation marks in genital tissues. To this end, we performed large scale array-based analysis of gene methylation profiles on genomic DNA from labioscrotal skin fibroblasts of 8 males and 26 individuals with androgen insensitivity syndrome (AIS) due to inactivating androgen receptor gene mutations. By this approach we identified differential methylation of 167 CpG loci representing 162 unique human genes. These were significantly enriched for androgen target genes and low CpG content promoter genes. Additional 75 genes showed a significant increase of heterogeneity of methylation in AIS compared to a high homogeneity in normal male controls. Our data show that normal and aberrant androgen receptor function is associated with distinct patterns of DNA-methylation marks in genital tissues. These findings support the concept that transcription factor binding to the DNA has an impact on the shape of the DNA methylome. These data which derived from a rare human model suggest that androgen programming of methylation marks contributes to sexual dimorphism in the human which might have considerable impact on the manifestation of sex-associated phenotypes and diseases.


Subject(s)
DNA Methylation/physiology , Receptors, Androgen/metabolism , Sex Characteristics , Androgen-Insensitivity Syndrome/genetics , Androgen-Insensitivity Syndrome/physiopathology , Androgens/metabolism , Apolipoproteins D/genetics , CpG Islands/genetics , Epigenomics , Female , Fibroblasts/metabolism , Gene Expression Regulation/genetics , Genitalia, Female/cytology , Genitalia, Male/cytology , Genomic Imprinting/genetics , Humans , Male , Mutation , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Receptors, Androgen/genetics , Skin/cytology
5.
Circ Res ; 111(12): 1504-16, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-22972877

ABSTRACT

RATIONALE: The emerging role of the ubiquitin-proteasome system in cardiomyocyte function and homeostasis implies the necessity of tight regulation of protein degradation. However, little is known about cardiac components of this machinery. OBJECTIVE: We sought to determine whether molecules exist that control turnover of cardiac-specific proteins. METHODS AND RESULTS: Using a bioinformatic approach to identify novel cardiac-enriched sarcomere proteins, we identified F-box and leucine-rich repeat protein 22 (Fbxl22). Tissue-specific expression was confirmed by multiple tissue Northern and Western Blot analyses as well as quantitative reverse-transcriptase polymerase chain reaction on a human cDNA library. Immunocolocalization experiments in neonatal and adult rat ventricular cardiomyocytes as well as murine heart tissue located Fbxl22 to the sarcomeric z-disc. To detect cardiac protein interaction partners, we performed a yeast 2-hybrid screen using Fbxl22 as bait. Coimmunoprecipitation confirmed the identified interactions of Fbxl22 with S-phase kinase-associated protein 1 and Cullin1, 2 critical components of SCF (Skp1/Cul1/F-box) E3- ligases. Moreover, we identified several potential substrates, including the z-disc proteins α-actinin and filamin C. Consistently, in vitro overexpression of Fbxl22-mediated degradation of both substrates in a dose-dependent fashion, whereas proteasome inhibition with MG-132 markedly attenuated degradation of both α-actinin and filamin C. Finally, targeted knockdown of Fbxl22 in rat cardiomyocytes as well as zebrafish embryos results in the accumulation of α-actinin associated with severely impaired contractile function and cardiomyopathy in vivo. CONCLUSIONS: These findings reveal the previously uncharacterized cardiac-specific F-box protein Fbxl22 as a component of a novel cardiac E3 ligase. Fbxl22 promotes the proteasome-dependent degradation of key sarcomeric proteins, such as α-actinin and filamin C, and is essential for maintenance of normal contractile function in vivo.


Subject(s)
F-Box Proteins/physiology , Myocardial Contraction/physiology , Myocytes, Cardiac/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Sarcomeres/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Cells, Cultured , HEK293 Cells , Humans , Molecular Sequence Data , Myocytes, Cardiac/physiology , Protein Transport/physiology , Rats , Sarcomeres/physiology
6.
J Mol Med (Berl) ; 90(12): 1439-47, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22669512

ABSTRACT

Genetic factors contribute to progression and modulation of hepatic fibrosis. High throughput genomics/transcriptomics approaches aiming at identifying key regulators of fibrosis development are tainted with the difficulty of separating essential biological "driver" from modifier genes. We applied a comparative transcriptomics approach and investigated fibrosis development in different organs to identify overlapping expression changes, since these genes may be part of core pathways in fibrosis development. Gene expression was analysed on publicly available microarray data from liver, lung and kidney fibrosis. RARRES1, AGER and S100A2 were differentially regulated in all fibrosis experiments. RARRES1 was extensively analysed by means of advanced bioinformatics analyses and functional studies. Microarray and Western Blot analysis of a standard liver fibrosis model (CCl(4)) demonstrated an early induction of RARRES1 mRNA and protein expression. In addition, quantitative RT-PCR in tissue samples from patients with advanced liver fibrosis showed higher expression as compared to non-fibrotic biopsies. Microarray analysis of RARRES1 overexpressing cells identified an enrichment of a major signature associated with fibrosis. Furthermore, RARRES1 expression increased during in vitro activation of hepatic stellate cells. To further verify the pro-fibrogenic role across organs, we demonstrated an increase in RARRES1 expression in a rat lung fibrosis model induced by adenoviral TGF-ß1 induction. We have performed a comparative transcriptomics analysis in order to identify core pathways of liver fibrogenesis, confirmed a candidate gene and enlightened the up- and downstream mechanisms of its action leading to fibrosis across organs and species.


Subject(s)
Hepatic Stellate Cells/metabolism , Liver Cirrhosis/metabolism , Liver/metabolism , Membrane Proteins/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Humans , In Vitro Techniques , Kidney/metabolism , Liver/pathology , Lung/metabolism , Male , Mice , Rats , Reverse Transcriptase Polymerase Chain Reaction
7.
Methods ; 57(3): 376-82, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22326879

ABSTRACT

The precise detection of pharmaceutical drug uptake and knowledge of a drug's efficacy at the single-cell level is crucial for understanding a compound's performance. Many pharmaceutical drugs, like the model substances Doxorubicin, Mitoxantrone or Irinotecan, have a distinctive natural fluorescence that can be readily exploited for research purposes. Utilizing this respective natural fluorescence, we propose a method analyzing simultaneously in real-time the efficiency, effects and the associated kinetics of compound-uptake and efflux in mammalian cells by flow cytometry. We show that real-time flow cytometric quantification of compound-uptake is reliably measured and that analyzing their respective uptake kinetic provides additional valuable information which can be used for improving drug dosage and delivery. Exploiting the native fluorescence of natural compounds is clearly advantageous compared to the usage of non-related fluorescent uptake-reporter substances, possibly yielding in unphysiological data. Flow cytometric analysis allows live-dye based multi-parametric high-throughput screening of pharmaceutical compound activity, improving cytotoxicity testing by combining several assays into a single, high resolution readout. This approach can be a useful tool identifying potential inhibitors for multiple drug resistance (MDR), representing a major challenge to the targeted treatment of various diseases.


Subject(s)
Antineoplastic Agents/metabolism , Camptothecin/analogs & derivatives , Doxorubicin/metabolism , Mitoxantrone/metabolism , Single-Cell Analysis/methods , Antineoplastic Agents/pharmacology , Biological Transport , Camptothecin/metabolism , Camptothecin/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Flow Cytometry , Fluoresceins , Fluorescence , Humans , Irinotecan , Kinetics , Mitoxantrone/pharmacology , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism
8.
Cancer Genomics Proteomics ; 9(1): 15-25, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22210045

ABSTRACT

BACKGROUND: The phenotypic expression of any given heterogeneous tissue type is the composite of interactions between individual cell types governed principally by inherent, cell-specific molecular mechanisms. MATERIALS AND METHODS: Using a combination of laser-capture microdissection, oligonucleotide microarrays, bioinformatic and statistical tools, we analyzed colorectal cancer tissues in order to spot the significant pathways that were differentially deregulated between the epithelial and stromal compartments and compared these alongside the ones of whole tissue dissection. RESULTS: The stromal pathway profiles were very similar to the ones of whole tissue, in contrast to the epithelial input, with stroma emerging as the major determinant of the cancer phenotype. Differentially expressed genes in the epithelial compartment correlated significantly with the carbohydrate antigen 19-9 tumor marker. CONCLUSION: The accurate interpretation of data arising from the analysis of heterogeneous tissue structures lends itself to inherent biases of its constitutive components with each component presenting explorable analytical advantages.


Subject(s)
Colorectal Neoplasms/genetics , Signal Transduction , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/metabolism , Epithelial Cells/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Male , Microdissection , Middle Aged , Neoplasm Staging , Stromal Cells/metabolism
9.
Am J Hum Genet ; 88(4): 450-7, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21457905

ABSTRACT

Tobacco smoking is responsible for substantial morbidity and mortality worldwide, in particular through cardiovascular, pulmonary, and malignant pathology. CpG methylation might plausibly play a role in a variety of smoking-related phenomena, as suggested by candidate gene promoter or global methylation studies. Arrays allowing hypothesis-free searches on a scale resembling genome-wide studies of SNPs have become available only very recently. Methylation extents in peripheral-blood DNA were assessed at 27,578 sites in more than 14,000 gene promoter regions in 177 current smokers, former smokers, and those who had never smoked, with the use of the Illumina HumanMethylation 27K BeadChip. This revealed a single locus, cg03636183, located in F2RL3, with genome-wide significance for lower methylation in smokers (p = 2.68 × 10(-31)). This was similarly significant in 316 independent replication samples analyzed by mass spectrometry and Sequenom EpiTyper (p = 6.33 × 10(-34)). Our results, which were based on a rigorous replication approach, show that the gene coding for a potential drug target of cardiovascular importance features altered methylation patterns in smokers. To date, this gene had not attracted attention in the literature on smoking.


Subject(s)
DNA Methylation , Smoking/adverse effects , Smoking/genetics , CpG Islands , DNA Primers/genetics , Female , Humans , Male , Middle Aged , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Receptors, Thrombin/genetics , Reproducibility of Results , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
10.
Mol Ther ; 19(4): 703-10, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21364536

ABSTRACT

Lentiviral vectors with self-inactivating (SIN) long terminal repeats (LTRs) are promising for safe and sustained transgene expression in dividing as well as quiescent cells. As genome organization and transcription substantially differs between actively dividing and postmitotic cells in vivo, we hypothesized that genomic vector integration preferences might be distinct between these biological states. We performed integration site (IS) analyses on mouse dividing cells (fibroblasts and hematopoietic progenitor cells (HPCs)) transduced ex vivo and postmitotic cells (eye and brain) transduced in vivo. As expected, integration in dividing cells occurred preferably into gene coding regions. In contrast, postmitotic cells showed a close to random frequency of integration into genes and gene spare long interspersed nuclear elements (LINE). Our studies on the potential mechanisms responsible for the detected differences of lentiviral integration suggest that the lowered expression level of Psip1 reduce the integration frequency in vivo into gene coding regions in postmitotic cells. The motif TGGAA might represent one of the factors for preferred lentiviral integration into mouse and rat Satellite DNA. These observations are highly relevant for the correct assessment of preclinical biosafety studies, indicating that lentiviral vectors are well suited for safe and effective clinical gene transfer into postmitotic tissues.


Subject(s)
Genetic Vectors/genetics , Lentivirus/genetics , Mitosis/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , DNA, Satellite/genetics , Female , Mice , Mice, Inbred BALB C , Polymerase Chain Reaction , Rats , Terminal Repeat Sequences/genetics , Transcription Factors/genetics , Virus Integration/genetics
11.
Circ Cardiovasc Genet ; 4(2): 110-22, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21252143

ABSTRACT

BACKGROUND: Today, mutations in more than 30 different genes have been found to cause inherited cardiomyopathies, some associated with very poor prognosis. However, because of the genetic heterogeneity and limitations in throughput and scalability of current diagnostic tools up until now, it is hardly possible to genetically characterize patients with cardiomyopathy in a fast, comprehensive, and cost-efficient manner. METHODS AND RESULTS: We established an array-based subgenomic enrichment followed by next-generation sequencing to detect mutations in patients with hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM). With this approach, we show that the genomic region of interest can be enriched by a mean factor of 2169 compared with the coverage of the whole genome, resulting in high sequence coverage of selected disease genes and allowing us to define the genetic pathogenesis of cardiomyopathies in a single sequencing run. In 6 patients, we detected disease-causing mutations, 2 microdeletions, and 4 point mutations. Furthermore, we identified several novel nonsynonymous variants, which are predicted to be harmful, and hence, might be potential disease mutations or modifiers for DCM or HCM. CONCLUSIONS: The approach presented here allows for the first time a comprehensive genetic screening in patients with hereditary DCM or HCM in a fast and cost-efficient manner.


Subject(s)
Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Hypertrophic/genetics , Sequence Analysis, DNA/methods , Adult , Base Sequence , Cardiac Myosins/genetics , Cardiomyopathy, Dilated/diagnosis , Cardiomyopathy, Hypertrophic/diagnosis , Carrier Proteins/genetics , Child , Codon, Nonsense , Female , Frameshift Mutation , Gene Deletion , Genetic Heterogeneity , Humans , Male , Middle Aged , Mutation, Missense , Myosin Heavy Chains/genetics , Point Mutation , Protein Serine-Threonine Kinases/genetics
12.
Exp Dermatol ; 20(4): 373-5, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20812966

ABSTRACT

Vitamin A is a pivotal regulator of differentiation and growth of developing and adult skin. Retinoic acid is the major physiologically active form of vitamin A regulating the expression of different genes through retinoic acid nuclear receptors. Here, we present evidence that other vitamin A derivates - retinol and retinal - are also capable of functioning as regulators of gene expression in the keratinocyte cell line HaCaT. We have shown that all-trans retinol (ATRol) and all-trans retinal (ATRal) are capable of modulating gene expression in keratinocytes, which is not because of vitamin A metabolism in the cells, and retinol and retinal modulate gene expression through nuclear receptors: retinoic acid receptors (RARs) and retinoid X receptors (RXRs). Based on the data, we propose that ATRol and all-trans retinal, in addition to all-trans retinoic acid, can function as important regulators of gene expression manifesting their effect through the nuclear receptors RARs and RXRs.


Subject(s)
Gene Expression Regulation , Retinaldehyde/genetics , Vitamin A/genetics , Humans , Keratinocytes/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinaldehyde/metabolism , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Transcription, Genetic , Vitamin A/metabolism
13.
Circ Res ; 107(10): 1253-64, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20847312

ABSTRACT

RATIONALE AND OBJECTIVE: The M-band represents a transverse structure in the center of the sarcomeric A-band and provides an anchor for the myosin-containing thick filaments. In contrast to other sarcomeric structures, eg, the Z-disc, only few M-band-specific proteins have been identified to date, and its exact molecular composition remains unclear. METHODS AND RESULTS: Using a bioinformatic approach to identify novel heart- and muscle-specific genes, we found a leucine rich protein, myomasp (Myosin-interacting, M-band-associated stress-responsive protein)/LRRC39. RT-PCR and Northern and Western blot analyses confirmed a cardiac-enriched expression pattern, and immunolocalization of myomasp revealed a strong and specific signal at the sarcomeric M-band. Yeast 2-hybrid screens, as well as coimmunoprecipitation experiments, identified the C terminus of myosin heavy chain (MYH)7 as an interaction partner for myomasp. Knockdown of myomasp in neonatal rat ventricular myocytes (NRVCMs) led to a significant upregulation of the stretch-sensitive genes GDF-15 and BNP. Conversely, the expression of MYH7 and the M-band proteins myomesin-1 and -2 was found to be markedly reduced. Mechanistically, knockdown of myomasp in NRVCM led to a dose-dependent suppression of serum response factor-dependent gene expression, consistent with earlier observations linking the M-band to serum response factor-mediated signaling. Finally, downregulation of myomasp/LRRC39 in spontaneously beating engineered heart tissue constructs resulted in significantly lower force generation and reduced fractional shortening. Likewise, knockdown of the myomasp/LRRC39 ortholog in zebrafish resulted in severely impaired heart function and cardiomyopathy in vivo. CONCLUSIONS: These findings reveal myomasp as a previously unrecognized component of an M-band-associated signaling pathway that regulates cardiomyocyte gene expression in response to biomechanical stress.


Subject(s)
Carrier Proteins/metabolism , Mechanotransduction, Cellular , Muscle Proteins/metabolism , Myocardial Contraction , Myocytes, Cardiac/metabolism , Proteins/metabolism , Sarcomeres/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Blotting, Northern , Blotting, Western , Cardiac Myosins/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/physiopathology , Carrier Proteins/genetics , Cells, Cultured , Cloning, Molecular , Connectin , Embryo, Nonmammalian/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation , Growth Differentiation Factor 15/metabolism , Humans , Immunohistochemistry , Immunoprecipitation , Leucine-Rich Repeat Proteins , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Myosin Heavy Chains/metabolism , Natriuretic Peptide, Brain/metabolism , Oligonucleotide Array Sequence Analysis , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Proteins/genetics , RNA Interference , Rats , Rats, Sprague-Dawley , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Serum Response Factor/metabolism , Stress, Mechanical , Transfection , Two-Hybrid System Techniques , Zebrafish
15.
PLoS Genet ; 6(5): e1000971, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20523906

ABSTRACT

Epigenetic changes are widely considered to play an important role in aging, but experimental evidence to support this hypothesis has been scarce. We have used array-based analysis to determine genome-scale DNA methylation patterns from human skin samples and to investigate the effects of aging, chronic sun exposure, and tissue variation. Our results reveal a high degree of tissue specificity in the methylation patterns and also showed very little interindividual variation within tissues. Data stratification by age revealed that DNA from older individuals was characterized by a specific hypermethylation pattern affecting less than 1% of the markers analyzed. Interestingly, stratification by sun exposure produced a fundamentally different pattern with a significant trend towards hypomethylation. Our results thus identify defined age-related DNA methylation changes and suggest that these alterations might contribute to the phenotypic changes associated with skin aging.


Subject(s)
Aging/genetics , Epigenesis, Genetic , Skin/radiation effects , Sunlight , Adult , DNA Methylation , Humans , Skin/metabolism
16.
Mol Cell Proteomics ; 9(1): 1-10, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19674966

ABSTRACT

Protein affinity reagents (PARs), most commonly antibodies, are essential reagents for protein characterization in basic research, biotechnology, and diagnostics as well as the fastest growing class of therapeutics. Large numbers of PARs are available commercially; however, their quality is often uncertain. In addition, currently available PARs cover only a fraction of the human proteome, and their cost is prohibitive for proteome scale applications. This situation has triggered several initiatives involving large scale generation and validation of antibodies, for example the Swedish Human Protein Atlas and the German Antibody Factory. Antibodies targeting specific subproteomes are being pursued by members of Human Proteome Organisation (plasma and liver proteome projects) and the United States National Cancer Institute (cancer-associated antigens). ProteomeBinders, a European consortium, aims to set up a resource of consistently quality-controlled protein-binding reagents for the whole human proteome. An ultimate PAR database resource would allow consumers to visit one on-line warehouse and find all available affinity reagents from different providers together with documentation that facilitates easy comparison of their cost and quality. However, in contrast to, for example, nucleotide databases among which data are synchronized between the major data providers, current PAR producers, quality control centers, and commercial companies all use incompatible formats, hindering data exchange. Here we propose Proteomics Standards Initiative (PSI)-PAR as a global community standard format for the representation and exchange of protein affinity reagent data. The PSI-PAR format is maintained by the Human Proteome Organisation PSI and was developed within the context of ProteomeBinders by building on a mature proteomics standard format, PSI-molecular interaction, which is a widely accepted and established community standard for molecular interaction data. Further information and documentation are available on the PSI-PAR web site.


Subject(s)
Databases, Protein/standards , Proteome/analysis , Database Management Systems/standards , Humans , International Cooperation , Proteomics/methods , Terminology as Topic
17.
Aging Cell ; 9(1): 54-63, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19895632

ABSTRACT

Within 2-3 months of in vitro culture-expansion, mesenchymal stromal cells (MSC) undergo replicative senescence characterized by cell enlargement, loss of differentiation potential and ultimate growth arrest. In this study, we have analyzed DNA methylation changes upon long-term culture of MSC by using the HumanMethylation27 BeadChip microarray assessing 27,578 unique CpG sites. Furthermore, we have compared MSC from young and elderly donors. Overall, methylation patterns were maintained throughout both long-term culture and aging but highly significant differences were observed at specific CpG sites. Many of these differences were observed in homeobox genes and genes involved in cell differentiation. Methylation changes were verified by pyrosequencing after bisulfite conversion and compared to gene expression data. Notably, methylation changes in MSC were overlapping in long-term culture and aging in vivo. This supports the notion that replicative senescence and aging represent developmental processes that are regulated by specific epigenetic modifications.


Subject(s)
Aging , Cellular Senescence , CpG Islands , DNA Methylation , Epigenesis, Genetic , Stromal Cells/metabolism , Adult , Aged , Aged, 80 and over , Cells, Cultured , Gene Expression Regulation , Humans , Middle Aged , Time Factors , Young Adult
18.
Mol Biosyst ; 5(9): 973-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19668862

ABSTRACT

In vitro systems for monitoring safety of nutritional additives are desirable for high-throughput screenings and as a substitute for animal models. Carrageenan (CGN) is a sulfated polysaccharide widely used as a thickener and texturizer in human nutrition and is intensely discussed regarding its pathophysiological potential. Low molecular weight (lm) variants of CGN are considered to exert more profound pathophysiological effects in vivo than high molecular weight (hm) variants. We used a systematic approach to construct reporter systems allowing distinction between CGN-variants with different pathophysiological potential. Reporter systems utilizing segments of the CGN-responsive DMBT1 promoter did not display substantial activity in SW620 cells of intestinal epithelial origin. Genome-wide profiling revealed stronger qualitative and quantitative changes in global gene activities for hm-CGN than for lm-CGN (824 versus 91 genes; -6.64 to 22.33-fold for hm-CGN versus the range of -2.65 to 2.96-fold for lm-CGN). Reporter systems with segments of the IL-8 promoter showed a specific activation in response to hm-sulfated polysaccharides with lower pathophysiological potential in vivo and provided a better classification of CGN-variants than cytotoxicity assays in vitro. IL-8 reporter systems can be used for discerning between the effects of sulfated polysaccharides in vivo. Our data further provide initial insights into the molecular mechanisms that may play a role in the different effects of CGN-variants.


Subject(s)
Biosensing Techniques/methods , Carrageenan/toxicity , Systems Biology/methods , Calcium-Binding Proteins , Carrageenan/chemistry , Cell Line, Tumor , Cell Survival/physiology , DNA-Binding Proteins , Gene Expression , Genes, Reporter , HCT116 Cells , HT29 Cells , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Intestinal Mucosa/cytology , Molecular Weight , NF-kappa B/genetics , NF-kappa B/metabolism , Promoter Regions, Genetic , Receptors, Cell Surface/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Proteins
19.
Clin Cancer Res ; 15(17): 5457-65, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19706813

ABSTRACT

PURPOSE: To identify and characterize epigenetically regulated genes able to predict sensitivity or resistance to currently tested chemotherapeutic agents in glioma therapy. EXPERIMENTAL DESIGN: We used methylation-sensitive BeadArray technology to identify novel epigenetically regulated genes associated with apoptosis and with potential therapeutic targets in glioma therapy. To elucidate the functional consequences of promoter methylation in the identified target death receptor 4 (DR4), we investigated tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated and anti-DR4-mediated apoptosis in glioma cell lines (U373 and A172) with loss of DR4 and one glioma cell line (LN18) with robust DR4 expression. RESULTS: In human astrocytic tumors, we detected DR4 promoter hypermethylation in 60% (n = 5) of diffuse astrocytomas WHO grade 2, in 75% (n = 8) of anaplastic astrocytomas WHO grade 3, and in 70% of glioblastomas WHO grade 4 (n = 33). DR4 is a cell surface protein restricted to glioma cells and is targeted by TRAIL. Glioma cell lines U373 and A172 harbored heavily methylated DR4 promoters, and 5-aza-2-deoxycytidine-mediated demethylation reconstituted DR4 expression in these cell lines. Functional knockdown of DR4 by DR4-specific small interfering RNA in TRAIL-sensitive glioma cell line LN18 significantly mitigated apoptosis induced by an agonistic anti-DR4 antibody. 5-Aza-2-deoxycytidine-mediated demethylation resulted in a functional reconstitution of DR4 on the cell surface of TRAIL-resistant glioma cell line U373 and sensitized U373 to TRAIL-mediated apoptosis. Suppression of DR4 by small interfering RNA in demethylated U373 successfully reestablished the TRAIL-resistant phenotype of U373. CONCLUSIONS: DR4 promoter methylation is frequent in human astrocytic gliomas, and epigenetic silencing of DR4 mediates resistance to TRAIL/DR4-based glioma therapies.


Subject(s)
Apoptosis , Drug Resistance, Neoplasm/genetics , Gene Silencing , Glioma/drug therapy , Receptors, Tumor Necrosis Factor/genetics , TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , DNA Methylation/drug effects , DNA Methylation/physiology , Decitabine , Humans , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/physiology , RNA, Small Interfering/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand
20.
Int J Med Sci ; 6(1): 18-27, 2009.
Article in English | MEDLINE | ID: mdl-19214198

ABSTRACT

Progresses in biology and pharmacology led to highly specific bioactive substances, but their poor bioavailability at the site of action is a result of their physico-chemical properties. Various design approaches for transport carrier molecules facilitating the cellular entry of bioactive substances could help to reach their molecular target in cells and tissues. The transfer efficacy and the subsequent pharmacological effects of the cargo molecules are well investigated, but the investigations of effects of the carrier molecules themselves on the target cells or tissues remain necessary. A special attention should be paid to the differential gene expression, particularly in the interpretation of the data achieved by highly specific active pharmaceutical products. After application of transmembrane transport peptides, particularly the pAnt and also the HIV-1 Tat, cells respond with a conspicuous altered gene expression of at least three genes. The PKN1 gene was induced and two genes (ZCD1 and BSG) were slightly repressed. The genes and the chromosomes are described, the moderate differential gene expression graphed, and the ontology is listed.


Subject(s)
Acrylamides/pharmacology , Drug Carriers/pharmacology , Gene Expression/drug effects , Peptide Fragments/pharmacology , Amino Acid Sequence , Basigin/genetics , Carrier Proteins/pharmacology , Cell-Penetrating Peptides , Computational Biology , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Expression Profiling , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Mitochondrial Proteins/genetics , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Protein Kinase C/genetics , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/drug effects , Up-Regulation/genetics , tat Gene Products, Human Immunodeficiency Virus/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...