Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Eur J Histochem ; 68(1)2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38568207

ABSTRACT

During the aging process, cells can enter cellular senescence, a state in which cells leave the cell cycle but remain viable. This mechanism is thought to protect tissues from propagation of damaged cells and the number of senescent cells has been shown to increase with age. The speed of aging determines the lifespan of a species and it varies significantly in different species. To assess the progress of cellular senescence during lifetime, we performed a comparative longitudinal study using histochemical detection of the senescence-associated beta-galactosidase as senescence marker to map the staining patterns in organs of the long-lived zebrafish and the short-lived turquoise killifish using light- and electron microscopy. We compared age stages corresponding to human stages of newborn, childhood, adolescence, adult and old age. We found tissue-specific but conserved signal patterns with respect to organ distribution. However, we found dramatic differences in the onset of tissue staining. The stained zebrafish organs show little to no signal at newborn age followed by a gradual increase in signal intensity, whereas the organs of the short-lived killifish show an early onset of staining already at newborn stage, which remains conspicuous at all age stages. The most prominent signal was found in liver, intestine, kidney and heart, with the latter showing the most prominent interspecies divergence in onset of staining and in staining intensity. In addition, we found staining predominantly in epithelial cells, some of which are post-mitotic, such as the intestinal epithelial lining. We hypothesize that the association of the strong and early-onset signal pattern in the short-lived killifish is consistent with a protective mechanism in a fast growing species. Furthermore, we believe that staining in post-mitotic cells may play a role in maintaining tissue integrity, suggesting different roles for cellular senescence during life.


Subject(s)
Galactosidases , Killifishes , Longevity , Humans , Adolescent , Adult , Animals , Infant, Newborn , Child , Zebrafish , Longitudinal Studies , Fundulus heteroclitus
2.
Biology (Basel) ; 12(10)2023 Oct 03.
Article in English | MEDLINE | ID: mdl-37887015

ABSTRACT

INTRODUCTION: Osteoarthritis is a classical age-related disease, which affects millions of patients worldwide. To further understand the pathophysiology and to develop therapeutic strategies for this disease, animal models play a significant role. Nothobranchius furzeri is an established model for accelerated aging that spontaneously develops spinal deformities. Although the bone properties of N. furzeri are well described, characteristics of the intervertebral discs are still unknown. The aim of this study was to investigate the characteristics of the intervertebral discs of healthy and deformed N. furzeri. MATERIAL AND METHODS: Intervertebral properties of healthy and deformed N. furzeri were investigated in 8-, 12-, 18- and 21.5-week-old male fish of the GRZ strain. For histological evaluations the fish were decalcified, paraffin-embedded and stained with (1) hematoxylin and eosin, (2) toluidine blue and (3) alcian blue/picrosirius red. RESULTS: 8-week-old and deformed N. furzeri showed spongy-like tissue containing vacuolated notochord cells and a beginning formation of fibrous tissue in the central area. Older healthy fish showed fibrous tissue in the central region and a spongy-like tissue in the peripheral region. CONCLUSION: Our study revealed age- and disease-related alterations of the vertebral discs in N. furzeri. Further studies should investigate the utility of N. furzeri as a model for degenerative spine diseases.

3.
Gerontology ; 68(12): 1415-1427, 2022.
Article in English | MEDLINE | ID: mdl-35472763

ABSTRACT

INTRODUCTION: Osteoporosis is a frequent age-related disease, which affects millions of people worldwide. Despite significant progress in the treatment of the disease, a high number of patients still are underdiagnosed and undertreated. Therefore, novel animal models for the investigation of the disease are necessary. Nothobranchius furzeri is the shortest-lived vertebrate (with a lifespan of 3-7 months) that can be kept in captivity. Although it is an established model for aging research, studies on bone are lacking. The aim of this study was therefore to characterize N. furzeri as a potential model for age-related osteoporosis. MATERIALS AND METHODS: Bone properties of aging N. furzeri were investigated in male and female fish of the Gona Re Zhou strain, which were between 8 and 20 weeks old. Micro-computed tomography (Scanco Medical µCT35) was performed to determine the bone properties of the vertebral bodies. Bone structure and remodeling were investigated by different histological staining techniques and histomorphometry. The chemical composition of fish vertebrae and intervertebral discs was analyzed by Raman microspectroscopy. RESULTS: Osteoblasts, mono- and multinucleated osteoclasts but no osteocytes could be observed in the vertebral area of N. furzeri. Histomorphometric evaluations revealed a significant decrease of the number of osteoblasts/bone perimeter and for osteoid volume/bone volume (BV) a trend toward a decrease in old male N. furzeri. Comparing male and female fish, males showed higher BV densities and cortical thickness. The relative values of the bone volume density of 20-week-old male N. furzeri were significantly lower than 10-week-old ones. The mineral to matrix ratio increased with age in male and female fish. In the intervertebral discs, proteoglycans in relation to the organic matrix were significantly lower in older female fish. CONCLUSION: Our finding of a lack of osteocytes is in agreement with the fact that N. furzeri belongs to the evolutionarily advanced teleost fish. Furthermore, not only age-specific but also sex-specific differences were visible in the bone properties of N. furzeri, which can be taken into consideration for the study of gender aspects of age-related musculoskeletal diseases.


Subject(s)
Cyprinodontiformes , Fundulidae , Osteoporosis , Animals , Male , Female , X-Ray Microtomography , Longevity , Aging
4.
Front Mol Biosci ; 8: 627143, 2021.
Article in English | MEDLINE | ID: mdl-34222326

ABSTRACT

Erosion of the epigenetic DNA methylation landscape is a widely recognized hallmark of aging. Emerging advances in high throughput sequencing techniques, in particular DNA methylation data analysis, have resulted in the establishment of precise human and murine age prediction tools. In vertebrates, methylation of cytosine at the C5 position of CpG dinucleotides is executed by DNA methyltransferases (DNMTs) whereas the process of enzymatic demethylation is highly dependent on the activity of the ten-eleven translocation methylcytosine dioxygenase (TET) family of enzymes. Here, we report the identification of the key players constituting the DNA methylation machinery in the short-lived teleost aging model Nothobranchius furzeri. We present a comprehensive spatio-temporal expression profile of the methylation-associated enzymes from embryogenesis into late adulthood, thereby covering the complete killifish life cycle. Data mining of the N. furzeri genome produced five dnmt gene family orthologues corresponding to the mammalian DNMTs (DNMT1, 2, 3A, and 3B). Comparable to other teleost species, N. furzeri harbors multiple genomic copies of the de novo DNA methylation subfamily. A related search for the DNMT1 recruitment factor UHRF1 and TET family members resulted in the identification of N. furzeri uhrf1, tet1, tet2, and tet3. Phylogenetic analysis revealed high cross-species similarity on the amino acid level of all individual dnmts, tets, and uhrf1, emphasizing a high degree of functional conservation. During early killifish development all analyzed dnmts and tets showed a similar expression profile characterized by a strong increase in transcript levels after fertilization, peaking either at embryonic day 6 or at the black eye stage of embryonic development. In adult N. furzeri, DNA methylation regulating enzymes showed a ubiquitous tissue distribution. Specifically, we observed an age-dependent downregulation of dnmts, and to some extent uhrf1, which correlated with a significant decrease in global DNA methylation levels in the aging killifish liver and muscle. The age-dependent DNA methylation profile and spatio-temporal expression characteristics of its enzymatic machinery reported here may serve as an essential platform for the identification of an epigenetic aging clock in the new vertebrate model system N. furzeri.

5.
Proc Natl Acad Sci U S A ; 116(30): 14995-15000, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31289233

ABSTRACT

Patterns of gene expression are primarily determined by proteins that locally enhance or repress transcription. While many transcription factors target a restricted number of genes, others appear to modulate transcription levels globally. An example is MeCP2, an abundant methylated-DNA binding protein that is mutated in the neurological disorder Rett syndrome. Despite much research, the molecular mechanism by which MeCP2 regulates gene expression is not fully resolved. Here, we integrate quantitative, multidimensional experimental analysis and mathematical modeling to indicate that MeCP2 is a global transcriptional regulator whose binding to DNA creates "slow sites" in gene bodies. We hypothesize that waves of slowed-down RNA polymerase II formed behind these sites travel backward and indirectly affect initiation, reminiscent of defect-induced shockwaves in nonequilibrium physics transport models. This mechanism differs from conventional gene-regulation mechanisms, which often involve direct modulation of transcription initiation. Our findings point to a genome-wide function of DNA methylation that may account for the reversibility of Rett syndrome in mice. Moreover, our combined theoretical and experimental approach provides a general method for understanding how global gene-expression patterns are choreographed.


Subject(s)
DNA Methylation , Models, Theoretical , RNA Polymerase II/metabolism , Animals , Cell Line , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Mice , Protein Binding , Transcription Elongation, Genetic , Transcription Initiation, Genetic , Transcriptional Activation
6.
Gene Expr Patterns ; 33: 11-19, 2019 09.
Article in English | MEDLINE | ID: mdl-31075496

ABSTRACT

The founding member of the sirtuin family, yeast Sir2, was the first evolutionarily conserved gene to be identified as a regulator of longevity. Sirtuins constitute a protein family of metabolic sensors, translating changes in NAD + levels into adaptive responses, thereby acting as crucial regulators of the network that controls energy homeostasis and as such determines healthspan. In mammals the sirtuin family comprises seven proteins, SIRT1-SIRT7, which vary in tissue specificity, subcellular localization, enzymatic activity and targets. Here, we report the identification and a detailed spatio-temporal expression profile of sirtuin genes in the short-lived fish Nothobranchius furzeri, from embryogenesis to late adulthood, mapping its entire life cycle. Database exploration of the recently published N. furzeri genome revealed eight orthologues corresponding to the seven known mammalian sirtuins, including two copies of the sirt5 gene. Phylogenetic analysis showed high cross species similarity of individual sirtuins in both their overall amino acid sequence and catalytic domain, suggesting a high degree of functional conservation. Moreover, we show that N. furzeri sirtuins exhibit ubiquitous and wide tissue distribution with a unique spatial expression pattern for each individual member of this enzyme family. Specifically, we observed a transcriptional down-regulation of several sirtuin genes with age, most significantly sirt1, sirt5a, sirt6 and sirt7 in a wide range of functionally distinct tissues. Overall, this spatio-temporal expression analysis provides the foundation for future research, both into genetic and pharmacological manipulation of this important group of enzymes in Nothobranchius furzeri, an emerging model organism for aging research.


Subject(s)
Aging/genetics , Cyprinodontiformes/genetics , Fish Proteins/genetics , Sirtuins/genetics , Aging/metabolism , Animals , Conserved Sequence , Cyprinodontiformes/classification , Cyprinodontiformes/growth & development , Fish Proteins/chemistry , Fish Proteins/metabolism , Gene Expression Regulation, Developmental , Organ Specificity , Phylogeny , Sirtuins/chemistry , Sirtuins/metabolism
7.
Dev Genes Evol ; 228(6): 255-265, 2018 12.
Article in English | MEDLINE | ID: mdl-30178389

ABSTRACT

Members of the Klotho gene family have been identified as modulators of the aging process. Deletion of αklotho in the mouse results in a syndrome resembling rapid human aging. Conversely, overexpression of αklotho extends mammalian lifespan. Here, we identify klotho orthologs in the vertebrate aging model Nothobranchius furzeri and provide a detailed spatio-temporal expression profile of both paralogs, α and ßklotho, from embryogenesis until old age spanning the entire life cycle of the organism. Specifically, we observe low levels of expression of both paralogs during embryogenesis followed by a significant transcriptional induction as development proceeds. In adult killifish, αklotho is predominantly expressed in the liver, the kidney, and the developing pharyngeal teeth. Particularly high levels of αKlotho protein were identified in the kidney tubules, closely resembling mammalian expression patterns. Prominent ßklotho expression was detected in the killifish intestine and liver. Overall, qRT-PCR analysis of Klotho members as a function of age revealed steady transcript levels, except for ßklotho expression in the liver which was significantly downregulated with age. This spatio-temporal expression profiling may serve as a useful starting point to further investigate the distinct physiological roles of Klotho members during the aging process.


Subject(s)
Aging , Cyprinodontiformes/genetics , Fish Proteins/genetics , Glucuronidase/genetics , Animals , Cloning, Molecular , Cyprinodontiformes/growth & development , Klotho Proteins , Longevity , Transcriptome
8.
Histochem Cell Biol ; 150(3): 255-269, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29951776

ABSTRACT

Aging is associated with profound changes in the epigenome, resulting in alterations of gene expression, epigenetic landscape, and genome architecture. Class I Histone deacetylases (HDACs), consisting of HDAC1, HDAC2, HDAC3, and HDAC8, play a major role in epigenetic regulation of chromatin structure and transcriptional control, and have been implicated as key players in the pathogenesis of age-dependent diseases and disorders affecting health and longevity. Here, we report the identification of class I Hdac orthologs and their detailed spatio-temporal expression profile in the short-lived fish Nothobranchius furzeri from the onset of embryogenesis until old age covering the entire lifespan of the organism. Database search of the recently annotated N. furzeri genomes retrieved four distinct genes: two copies of hdac1 and one copy of each hdac3 and hdac8. However, no hdac2 ortholog could be identified. Phylogenetic analysis grouped the individual killifish class I Hdacs within the well-defined terminal clades. We find that upon aging, Hdac1 is significantly down-regulated in muscle, liver, and brain, and this age-dependent down-regulation in brain clearly correlates with increased mRNA levels of the cyclin-dependent kinase inhibitor cdkn1a (p21). Furthermore, this apparent reduction of class I HDACs in transcript and protein levels is mirrored in the mouse brain, highlighting an evolutionarily conserved role of class I HDACs during normal development and in the aging process.


Subject(s)
Aging , Fishes , Histone Deacetylase 1/genetics , Animals , Gene Expression Profiling , Histone Deacetylase 1/metabolism , Immunohistochemistry , Mice , Mice, Inbred C57BL , Survival Analysis
9.
Eur J Cell Biol ; 97(1): 63-74, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29254732

ABSTRACT

The human LEM-domain protein family is involved in fundamental aspects of nuclear biology. The LEM-domain interacts with the barrier-to-autointegration factor (BAF), which itself binds DNA. LEM-domain proteins LAP2, emerin and MAN1 are proteins of the inner nuclear membrane; they have important functions: maintaining the integrity of the nuclear lamina and regulating gene expression at the nuclear periphery. LEM4/ANKLE-2 has been proposed to participate in nuclear envelope reassembly after mitosis and to mediate dephosphorylation of BAF through binding to phosphatase PP2A. Here, we used CRISPR/Cas9 to create several cell lines deficient in LEM4/ANKLE-2. By using time-lapse video microscopy, we show that absence of this protein severely compromises the post mitotic re-association of the nuclear proteins BAF, LAP2α and LaminA to chromosomes. These defects give rise to a strong mechanical instability of the nuclear envelope in telophase and to a chromosomal instability leading to increased number of hyperploid cells. Reintroducing LEM4/ANKLE-2 in the cells by transfection could efficiently restore the telophase association of BAF and LAP2α to the chromosomes. This rescue phenotype was abolished for N- or C-terminally truncated mutants that had lost the capacity to bind PP2A. We demonstrate also that, in addition to binding to PP2A, LEM4/ANKLE-2 binds BAF through its LEM-domain, providing further evidence for a generic function of this domain as a principal interactor of BAF.


Subject(s)
Cell Nucleus/pathology , Chromosomal Instability , Membrane Proteins/metabolism , Mitosis , Nuclear Envelope/pathology , Nuclear Proteins/metabolism , Ploidies , Telophase , CRISPR-Cas Systems , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Lamin Type A/metabolism , Nuclear Envelope/metabolism
10.
Genome Biol ; 17(1): 220, 2016 10 25.
Article in English | MEDLINE | ID: mdl-27782844

ABSTRACT

BACKGROUND: Short interspersed elements (SINEs) represent the most abundant group of non-long-terminal repeat transposable elements in mammalian genomes. In primates, Alu elements are the most prominent and homogenous representatives of SINEs. Due to their frequent insertion within or close to coding regions, SINEs have been suggested to play a crucial role during genome evolution. Moreover, Alu elements within mRNAs have also been reported to control gene expression at different levels. RESULTS: Here, we undertake a genome-wide analysis of insertion patterns of human Alus within transcribed portions of the genome. Multiple, nearby insertions of SINEs within one transcript are more abundant in tandem orientation than in inverted orientation. Indeed, analysis of transcriptome-wide expression levels of 15 ENCODE cell lines suggests a cis-repressive effect of inverted Alu elements on gene expression. Using reporter assays, we show that the negative effect of inverted SINEs on gene expression is independent of known sensors of double-stranded RNAs. Instead, transcriptional elongation seems impaired, leading to reduced mRNA levels. CONCLUSIONS: Our study suggests that there is a bias against multiple SINE insertions that can promote intramolecular base pairing within a transcript. Moreover, at a genome-wide level, mRNAs harboring inverted SINEs are less expressed than mRNAs harboring single or tandemly arranged SINEs. Finally, we demonstrate a novel mechanism by which inverted SINEs can impact on gene expression by interfering with RNA polymerase II.


Subject(s)
RNA Polymerase II/genetics , Short Interspersed Nucleotide Elements/genetics , Transcription, Genetic , Transcriptome/genetics , Alu Elements/genetics , Cell Line , Evolution, Molecular , Gene Expression Regulation , Genome, Human , Humans , RNA, Double-Stranded/genetics , RNA, Messenger/genetics
11.
Genome Res ; 24(11): 1808-20, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25135956

ABSTRACT

The selectivity of transcriptional responses to extracellular cues is reflected by the deposition of stimulus-specific chromatin marks. Although histone H3 phosphorylation is a target of numerous signaling pathways, its role in transcriptional regulation remains poorly understood. Here, for the first time, we report a genome-wide analysis of H3S28 phosphorylation in a mammalian system in the context of stress signaling. We found that this mark targets as many as 50% of all stress-induced genes, underlining its importance in signal-induced transcription. By combining ChIP-seq, RNA-seq, and mass spectrometry we identified the factors involved in the biological interpretation of this histone modification. We found that MSK1/2-mediated phosphorylation of H3S28 at stress-responsive promoters contributes to the dissociation of HDAC corepressor complexes and thereby to enhanced local histone acetylation and subsequent transcriptional activation of stress-induced genes. Our data reveal a novel function of the H3S28ph mark in the activation of mammalian genes in response to MAP kinase pathway activation.


Subject(s)
Histones/metabolism , Serine/metabolism , Stress, Physiological/genetics , Transcriptional Activation , 3T3 Cells , Acetylation , Animals , Chromatin Immunoprecipitation , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling , Gene Ontology , Genome-Wide Association Study , HeLa Cells , High-Throughput Nucleotide Sequencing , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , MAP Kinase Signaling System/genetics , Mice , Oligonucleotide Array Sequence Analysis , Phosphorylation , Promoter Regions, Genetic/genetics , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
12.
Development ; 141(3): 604-616, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24449838

ABSTRACT

The histone deacetylases HDAC1 and HDAC2 are crucial regulators of chromatin structure and gene expression, thereby controlling important developmental processes. In the mouse brain, HDAC1 and HDAC2 exhibit different developmental stage- and lineage-specific expression patterns. To examine the individual contribution of these deacetylases during brain development, we deleted different combinations of Hdac1 and Hdac2 alleles in neural cells. Ablation of Hdac1 or Hdac2 by Nestin-Cre had no obvious consequences on brain development and architecture owing to compensation by the paralog. By contrast, combined deletion of Hdac1 and Hdac2 resulted in impaired chromatin structure, DNA damage, apoptosis and embryonic lethality. To dissect the individual roles of HDAC1 and HDAC2, we expressed single alleles of either Hdac1 or Hdac2 in the absence of the respective paralog in neural cells. The DNA-damage phenotype observed in double knockout brains was prevented by expression of a single allele of either Hdac1 or Hdac2. Strikingly, Hdac1(-/-)Hdac2(+/-) brains showed normal development and no obvious phenotype, whereas Hdac1(+/-)Hdac2(-/-) mice displayed impaired brain development and perinatal lethality. Hdac1(+/-)Hdac2(-/-) neural precursor cells showed reduced proliferation and premature differentiation mediated by overexpression of protein kinase C, delta, which is a direct target of HDAC2. Importantly, chemical inhibition or knockdown of protein kinase C delta was sufficient to rescue the phenotype of neural progenitor cells in vitro. Our data indicate that HDAC1 and HDAC2 have a common function in maintaining proper chromatin structures and show that HDAC2 has a unique role by controlling the fate of neural progenitors during normal brain development.


Subject(s)
Alleles , Brain/embryology , Brain/enzymology , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/genetics , Sequence Homology, Amino Acid , Acetophenones/pharmacology , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/genetics , Benzopyrans/pharmacology , Brain/metabolism , Brain/pathology , Co-Repressor Proteins/metabolism , DNA Damage/genetics , Embryo Loss/enzymology , Embryo Loss/pathology , Gene Deletion , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Histone Deacetylase 1/genetics , Histone Deacetylase 2/metabolism , Mice , Mice, Inbred C57BL , Phenotype , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
13.
Mol Pharmacol ; 85(3): 420-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24366667

ABSTRACT

The multispecific efflux transporter, P-glycoprotein, plays an important role in drug disposition. Substrate translocation occurs along the interface of its transmembrane domains. The rotational C2 symmetry of ATP-binding cassette transporters implies the existence of two symmetry-related sets of substrate-interacting amino acids. These sets are identical in homodimeric transporters, and remain evolutionary related in full transporters, such as P-glycoprotein, in which substrates bind preferentially, but nonexclusively, to one of two binding sites. We explored the role of pore-exposed tyrosines for hydrogen-bonding interactions with propafenone type ligands in their preferred binding site 2. Tyrosine 953 is shown to form hydrogen bonds not only with propafenone analogs, but also with the preferred site 1 substrate rhodamine123. Furthermore, an accessory role of tyrosine 950 for binding of selected propafenone analogs is demonstrated. The present study demonstrates the importance of domain interface tyrosine residues for interaction of small molecules with P-glycoprotein.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Hydrogen/metabolism , Propafenone/metabolism , Tyrosine/genetics , Tyrosine/metabolism , Binding Sites/genetics , Cell Line , HEK293 Cells , Humans , Hydrogen Bonding , Ligands , Mutation/genetics , Protein Structure, Tertiary/genetics
14.
RNA Biol ; 10(2): 192-204, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23353575

ABSTRACT

Adenosine to inosine deamination of RNA is widespread in metazoa. Inosines are recognized as guanosines and, therefore, this RNA-editing can influence the coding potential, localization and stability of RNAs. Therefore, RNA editing contributes to the diversification of the transcriptome in a flexible manner. The editing reaction is performed by adenosine deaminases that act on RNA (ADARs), which are essential for normal life and development in many organisms. Changes in editing levels are observed during development but also in neurological pathologies like schizophrenia, depression or tumors. Frequently, changes in editing levels are not reflected by changes in ADAR levels suggesting a regulation of enzyme activity. Until now, only a few factors are known that influence the activity of ADARs. Here we present a two-stage in vivo editing screen aimed to isolate enhancers of editing. A primary, high-throughput yeast-screen is combined with a more accurate secondary screen in mammalian cells that uses a fluorescent read-out to detect minor differences in RNA-editing. The screen was successfully employed to identify DSS1/SHFM1, the RNA binding protein hnRNP A2/B1 and a 3' UTR as enhancers of editing. By varying intracellular DSS1/SHFM1 levels, we can modulate A to I editing by up to 30%. Proteomic analysis indicates an interaction of DSS1/SHFM1 and hnRNP A2/B1 suggesting that both factors may act by altering the cellular RNP landscape. An extension of this screen to cDNAs from different tissues or developmental stages may prove useful for the identification of additional enhancers of RNA-editing.


Subject(s)
Adenosine Deaminase/metabolism , Enhancer Elements, Genetic , High-Throughput Nucleotide Sequencing/methods , RNA Editing , 3' Untranslated Regions , Adenosine Deaminase/genetics , Animals , DNA, Complementary/genetics , DNA, Complementary/metabolism , Genetic Vectors/genetics , Genetic Vectors/metabolism , HEK293 Cells , HeLa Cells , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Humans , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins , Rats , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
15.
Differentiation ; 85(1-2): 55-66, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23328540

ABSTRACT

Epigenetic mechanisms serve as key regulatory elements during vertebrate embryogenesis. Histone acetylation levels, controlled by the opposing action of histone acetyl transferases (HATs) and histone deacetylases (HDACs), influence the accessibility of DNA to transcription factors and thereby dynamically regulate transcriptional programs. HDACs execute important functions in the control of proliferation, differentiation, and the establishment of cell identities during embryonic development. To investigate the global role of the HDAC family during neural tube development, we employed Trichostatin A (TSA) to locally block enzymatic HDAC activity in chick embryos in ovo. We found that TSA treatment induces neural tube defects at the level of the posterior neuropore, ranging from slight undulations to a complete failure of neural tube closure. This phenotype is accompanied by morphological changes in neuroepithelial cells and induction of apoptosis. As a molecular consequence of HDAC inhibition, we observed a timely deregulated cadherin switching in the dorsal neural tube, illustrated by induction of Cadherin 6B as well as reciprocal downregulation of N-Cadherin expression. Concomitantly, several neural crest specific markers, including Bmp4, Pax3, Sox9 and Sox10 are induced, causing a premature loss of epithelial characteristics. Our findings provide evidence that HDAC function is crucial to control the regulatory circuits operating during trunk neural crest development and neural tube closure.


Subject(s)
Histone Deacetylase Inhibitors/toxicity , Hydroxamic Acids/toxicity , Neural Crest/drug effects , Neural Tube Defects/chemically induced , Animals , Apoptosis/drug effects , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Cadherins/genetics , Cadherins/metabolism , Chick Embryo , Neural Crest/embryology , Neural Tube/drug effects , Neural Tube/embryology , Neuroepithelial Cells/drug effects , Neuroepithelial Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Nucleic Acids Res ; 41(4): 2581-93, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23275536

ABSTRACT

RNA editing by adenosine deaminases that act on RNA (ADARs) diversifies the transcriptome by changing adenosines to inosines. In mammals, editing levels vary in different tissues, during development, and also in pathogenic conditions. From a screen for repressors of editing we have isolated three proteins that repress ADAR2-mediated RNA editing. The three proteins RPS14, SFRS9 and DDX15 interact with RNA. Overexpression or depletion of these proteins can decrease or increase editing levels by 15%, thus allowing a modulation of RNA editing up to 30%. Interestingly, the three proteins alter RNA editing in a substrate-specific manner that correlates with their RNA binding preferences. In mammalian cells, SFRS9 significantly affects editing of the two substrates CFLAR and cyFIP2, while the ribosomal protein RPS14 mostly inhibits editing of cyFIP2 messenger RNA. The helicase DDX15, in turn, has a strong effect on editing in Caenorhabditis elegans. Expression of the three factors decreases during mouse brain development. Moreover, expression levels of SFRS9 and DDX15 respond strongly to neuronal stimulation or repression, showing an inverse correlation with editing levels. Colocalization and immunoprecipitation studies demonstrate a direct interaction of SFRS9 and RPS14 with ADAR2, while DDX15 associates with other helicases and splicing factors. Our data show that different editing sites can be specifically altered in their editing pattern by changing the local RNP landscape.


Subject(s)
Adenosine Deaminase/metabolism , Caenorhabditis elegans Proteins/metabolism , RNA Editing , RNA-Binding Proteins/metabolism , Animals , Brain/embryology , Brain/growth & development , Brain/metabolism , Caenorhabditis elegans , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Cells, Cultured , HeLa Cells , Humans , Mice , Neurons/metabolism , Neurons/physiology , Nuclear Proteins/metabolism , RNA/metabolism , RNA Helicases/metabolism , RNA Interference , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Rats , Rats, Sprague-Dawley , Ribosomal Proteins/metabolism , Saccharomyces cerevisiae/genetics , Serine-Arginine Splicing Factors
17.
EMBO J ; 29(23): 3992-4007, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20967026

ABSTRACT

Histone deacetylase (HDAC) inhibitors induce cell cycle arrest, differentiation or apoptosis in tumour cells and are, therefore, promising anti-cancer reagents. However, the specific HDAC isoforms that mediate these effects are not yet identified. To explore the role of HDAC1 in tumourigenesis and tumour proliferation, we established an experimental teratoma model using wild-type and HDAC1-deficient embryonic stem cells. HDAC1-deficient teratomas showed no significant difference in size compared with wild-type teratomas. Surprisingly, loss of HDAC1 was not only linked to increased apoptosis, but also to significantly enhanced proliferation. Epithelial structures showed reduced differentiation as monitored by Oct3/4 expression and changed E-cadherin localization and displayed up-regulated expression of SNAIL1, a regulator of epithelial cell plasticity. Increased levels of the transcriptional regulator SNAIL1 are crucial for enhanced proliferation and reduced differentiation of HDAC1-deficient teratoma. Importantly, the analysis of human teratomas revealed a similar link between loss of HDAC1 and enhanced tumour malignancy. These findings reveal a novel role for HDAC1 in the control of tumour proliferation and identify HDAC1 as potential marker for benign teratomas.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Expression Regulation, Neoplastic , Histone Deacetylase 1/genetics , Teratoma/enzymology , Animals , Apoptosis , Cadherins/genetics , Carcinoma, Embryonal/enzymology , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/pathology , Cell Line , Cell Line, Tumor , Cell Proliferation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/pathology , Histone Deacetylase 1/metabolism , Humans , Mice , Octamer Transcription Factor-3/genetics , Phenotype , Snail Family Transcription Factors , Teratoma/genetics , Teratoma/pathology , Transcription Factors/genetics
18.
Int J Dev Biol ; 54(10): 1527-37, 2010.
Article in English | MEDLINE | ID: mdl-20979029

ABSTRACT

Histone deacetylases (HDACs) are a family of enzymes which regulate the acetylation state of nucleosomal histones, as well as non-histone proteins. By altering local chromatin architecture, HDACs play important roles in shaping cell differentiation and morphogenesis. Expression of class I HDACs during early chick development has so far not been analyzed. Here, we report the expression profile of chick class I HDACs from the onset of gastrulation (HH2) to day 4 of development and compare it to relevant stages during mouse development. Visualized by in situ hybridization to whole mount embryos and tissue sections, we found tissue-specific overlapping temporal and spatial expression domains for all four class I HDACs in chick and mouse, although species-specific differences could be identified. All class I HDACs in both species are highly expressed in the developing brain. In particular, HDAC1 is expressed at sites of anterior and posterior neural tube closure most obvious in the hot spot-like expression of HDAC1 in HH12 chicken embryos. A significant species-specific spatio-temporal expression pattern was observed for HDAC8. Whereas HDAC8 is exclusively found in fore- and midbrain regions during early mouse embryogenesis, the chick ortholog shows an expanded expression pattern, suggesting a more diversified role of HDAC8 in the chick system. Our results present a basis for further functional analysis of class I HDACs in chick development.


Subject(s)
Brain/embryology , Embryonic Development , Histone Deacetylases/genetics , Acetylation , Animals , Blotting, Western , Brain/enzymology , Cell Differentiation , Chick Embryo , Chromatin/chemistry , Embryo, Mammalian , Embryo, Nonmammalian , Gastrulation , Gene Expression Regulation, Developmental , Histone Deacetylases/metabolism , Histones/genetics , Histones/metabolism , In Situ Hybridization , Mice , Neural Tube/embryology , Nucleosomes/genetics , Nucleosomes/metabolism , Reverse Transcriptase Polymerase Chain Reaction
19.
Front Biosci ; 12: 3104-16, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17485285

ABSTRACT

RNAi refers to the sequence-specific degradation of RNA that follows the cellular introduction of homologous short interfering (si) RNA. RNAi has emerged as a powerful tool to probe the function of genes of known sequence in vitro and in vivo. Advances in vector design permit the effective expression of siRNA in human cells. Numerous recent investigations have described the ability of RNAi to decrease the replication of human immunodeficiency virus type 1 (HIV-1) in lymphocytic cells using siRNA targeting viral (e.g. tat, gag, rev) and host (e.g. CCR5, CD4) proteins. Can RNAi be used as a form of genetic therapy for HIV-1 infection? Recent data indicate that the dynamic replication kinetics of HIV-1 pose a considerable barrier to achieving durable virus suppression by RNAi with the rapid emergence of HIV-1 mutants resistant to siRNA. This review summarizes recent work on HIV-1 specific RNAi with a focus on potential strategies to overcome HIV-1 resistance to RNAi.


Subject(s)
HIV-1/genetics , RNA Interference , Base Sequence , Genes, tat , Humans , MicroRNAs/genetics , Promoter Regions, Genetic , RNA, Viral/genetics
20.
AIDS ; 20(15): 1917-22, 2006 Oct 03.
Article in English | MEDLINE | ID: mdl-16988512

ABSTRACT

OBJECTIVES: To engineer Lactobacillus spp. to secrete HIV-1 fusion inhibitors with potent neutralizing activity against primary HIV-1 isolates. METHODS: HIV-1 fusion inhibitors (FI-1, FI-2, and FI-3) were introduced into the previously developed shuttle vector pTSV2 and transformed in L. plantarum and L. gasseri. The signal peptide Usp45 from L. lactis was used to achieve high secretion efficiency of peptides into the bacterial supernatant. The antiviral activity of lactobacillus-derived HIV-1 fusion inhibitors was tested against a panel of primary HIV-1 isolates and a chimeric simian/HIV (SHIV-162P3) using the TZM infection assay. TZM-bl cells are engineered HeLa cells that express CD4, CCR5, and CXCR4 and contain integrated reporter genes for firefly luciferase and beta-galactosidase under the control of an HIV-1 long terminal repeat. The amount of secreted fusion inhibitor FI-3 was determined by Western blot analysis and the antiviral specificity verified by antibody-mediated depletion of peptide FI-3 and HIV-1 infection with VSV-G envelope pseudotyped virions. RESULTS: Viral infectivity of primary HIV-1 isolates and SHIV-162P3 was neutralized by up to 98% and 72%, respectively, by 10% (v/v) lactobacillus supernatant containing fusion inhibitor FI-3. The antiviral activity of the lactobacillus-derived fusion inhibitor FI-3 was clearly shown to be attributable to the secreted fusion inhibitor peptide. CONCLUSION: The development of recombinant lactobacilli expressing HIV-1 fusion inhibitors with potent neutralizing activity represents an important step toward the development of a live microbial (topical) microbicide against HIV-1 transmission.


Subject(s)
Anti-HIV Agents , Bioreactors , HIV Fusion Inhibitors , Lactobacillus/physiology , Animals , Blotting, Western , HIV Infections/drug therapy , HIV-1/drug effects , HeLa Cells , Humans , Lactobacillus plantarum/metabolism , Neutralization Tests , Organisms, Genetically Modified , Recombinant Proteins/metabolism , Simian Immunodeficiency Virus/drug effects , Transduction, Genetic/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...