Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Int J Androl ; 35(5): 731-40, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22435752

ABSTRACT

Recent societal acceptance of cannabinoids as recreational and therapeutic drugs has posed a potential hazard to male reproductive health. Mammals have a highly sophisticated endogenous cannabinoid (ECS) system that regulates male (and female) reproduction and exo-cannabinoids may influence it adversely. Therefore it is imperative to determine their effects on male reproduction so that men can make informed choices as to their use. Here, an animal model was used to administer HU210, a synthetic analogue of Δ9-tetrahydrocannabinol (THC) and potent cannabinoid receptor (CB) agonist to determine its effects on reproductive organ weights, spermatogenesis, testicular histology and sperm motility. Its effects on the physiological endocannabinoid system were also investigated. Spermatogenesis was markedly impaired with reductions in total sperm count after 2 weeks of exposure. Spermatogenic efficiency was depleted, and Sertoli cell number decreased as exposure time increased with seminiferous tubules showing germ cell depletion developing into atrophy in some cases. Sperm motility was also adversely affected with marked reductions from 2 weeks on. HU210 also acted on the sperm's endocannabinoid system. Long-term use of exo-cannabinoids has adverse effects on both spermatogenesis and sperm function. These findings highlight the urgent need for studies evaluating the fertility potential of male recreational drug users. HU210, a selective agonist for CB1 and CB2 cannabinoid receptors impairs spermatogenesis and sperm motility and deregulates the endocannabinoid system.


Subject(s)
Cannabinoid Receptor Agonists/toxicity , Dronabinol/analogs & derivatives , Spermatogenesis/drug effects , Animals , Dronabinol/toxicity , Endocannabinoids/physiology , Male , Organ Size/drug effects , Rats , Sertoli Cells , Sperm Motility/drug effects
2.
Curr Top Microbiol Immunol ; 310: 23-44, 2006.
Article in English | MEDLINE | ID: mdl-16909905

ABSTRACT

Epigenetic regulation of gene transcription relies on molecular marks like DNA methylation or histone modifications. Here we review recent advances in our understanding of epigenetic regulation in the fruit fly Drosophila melanogaster. In the past, DNA methylation research has primarily utilized mammalian model systems. However, several recent landmark discoveries have been made in other organisms. For example, the interaction between DNA methylation and histone methylation was first described in the filamentous fungus Neurospora crassa. Another example is provided by the interaction between epigenetic modifications and the RNA interference (RNAi) machinery that was first reported in the fission yeast Schizosaccharomyces pombe. Another organism with great experimental power is the fruit fly Drosophila. Epigenetic regulation by chromatin has been extensively analyzed in the fly and several of the key components have been discovered in this organism. In this chapter, we will focus on three aspects that represent the complexity of epigenetic gene regulation. (1) We will discuss the available data about the DNA methylation system, (2) we will illuminate the interaction between DNA methylation and chromatin regulation, and (3) we will provide an overview over the Polycomb system of epigenetic chromatin modifiers that has proved to be an important paradigm for a chromatin system regulating epigenetic programming.


Subject(s)
DNA Methylation , Drosophila/genetics , Epigenesis, Genetic , Acetylation , Animals , Chromatin/chemistry , Chromatin/physiology , DNA (Cytosine-5-)-Methyltransferases/physiology , DNA-Binding Proteins/physiology , Drosophila Proteins/physiology , Gene Expression , Histones/metabolism
4.
Genome Biol ; 5(1): R3, 2003.
Article in English | MEDLINE | ID: mdl-14709175

ABSTRACT

BACKGROUND: While the genome sequences for a variety of organisms are now available, the precise number of the genes encoded is still a matter of debate. For the human genome several stringent annotation approaches have resulted in the same number of potential genes, but a careful comparison revealed only limited overlap. This indicates that only the combination of different computational prediction methods and experimental evaluation of such in silico data will provide more complete genome annotations. In order to get a more complete gene content of the Drosophila melanogaster genome, we based our new D. melanogaster whole-transcriptome microarray, the Heidelberg FlyArray, on the combination of the Berkeley Drosophila Genome Project (BDGP) annotation and a novel ab initio gene prediction of lower stringency using the Fgenesh software. RESULTS: Here we provide evidence for the transcription of approximately 2,600 additional genes predicted by Fgenesh. Validation of the developmental profiling data by RT-PCR and in situ hybridization indicates a lower limit of 2,000 novel annotations, thus substantially raising the number of genes that make a fly. CONCLUSIONS: The successful design and application of this novel Drosophila microarray on the basis of our integrated in silico/wet biology approach confirms our expectation that in silico approaches alone will always tend to be incomplete. The identification of at least 2,000 novel genes highlights the importance of gathering experimental evidence to discover all genes within a genome. Moreover, as such an approach is independent of homology criteria, it will allow the discovery of novel genes unrelated to known protein families or those that have not been strictly conserved between species.


Subject(s)
Drosophila melanogaster/genetics , Gene Expression Profiling/methods , Genes, Insect/physiology , Genome , Oligonucleotide Array Sequence Analysis/methods , Animals , Cluster Analysis , Computational Biology/methods , Computational Biology/statistics & numerical data , Gene Expression Profiling/statistics & numerical data , In Situ Hybridization/methods , Models, Genetic , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis/statistics & numerical data , Predictive Value of Tests , Pseudogenes/genetics , RNA Interference/physiology , Reverse Transcriptase Polymerase Chain Reaction/methods
6.
Bioessays ; 23(7): 566-70, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11462210

ABSTRACT

Polycomb response elements (PREs) are regulatory switch elements that can direct the genes that they control to be either active or silenced. Once decided, this on or off state is maintained through subsequent cell divisions. We do not know how the switching works, or how it is copied to newly replicated chromosomes. Experiments that switch a silenced PRE to an active state have provided insights into both questions. A PRE switched experimentally can remember its previously silenced state and return to it after several cell divisions. In the most recent study of this phenomen on, the data show that several distinct variables affect the ability of PREs to "remember" and restore their previous state. The authors' interpretation of these results is discussed here.


Subject(s)
Drosophila Proteins , Drosophila melanogaster/genetics , Gene Expression Regulation , Gene Silencing , Genes, Insect , Insect Proteins/genetics , Response Elements , Animals , Polycomb Repressive Complex 1
7.
Biochim Biophys Acta ; 1552(1): 15-26, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11781112

ABSTRACT

The past several years have seen a tremendous advance in the understanding of the basic mechanisms of epigenetic regulation. A large number of studies have not only linked epigenetics with cell cycle regulation but also partially unravelled how epigenetics may regulate gene expression. The aim of this review is to provide an overview of the latest findings and current ideas on epigenetics with a focus on emphasizing the emerging influence epigenetics has on the onset and progression of cancer.


Subject(s)
DNA-Binding Proteins/physiology , Drosophila Proteins , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Repressor Proteins/physiology , Transcription Factors , Animals , Cell Cycle Proteins/physiology , DNA-Binding Proteins/genetics , Epithelial Cells/pathology , Humans , Models, Chemical , Neoplasms/pathology , Polycomb-Group Proteins , Repressor Proteins/genetics
9.
Mol Cell Biol ; 19(12): 8451-60, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10567570

ABSTRACT

The proteins of the Polycomb group (PcG) are required for maintaining regulator genes, such as the homeotic selectors, stably and heritably repressed in appropriate developmental domains. It has been suggested that PcG proteins silence genes by creating higher-order chromatin structures at their chromosomal targets, thus preventing the interaction of components of the transcriptional machinery with their cis-regulatory elements. An unresolved issue is how higher order-structures are anchored at the chromatin base, the nucleosomal fiber. Here we show a direct biochemical interaction of a PcG protein-the Polycomb (PC) protein-with nucleosomal core particles in vitro. The main nucleosome-binding domain coincides with a region in the C-terminal part of PC previously identified as the repression domain. Our results suggest that PC, by binding to the core particle, recruits other PcG proteins to chromatin. This interaction could provide a key step in the establishment or regulation of higher-order chromatin structures.


Subject(s)
Drosophila Proteins , Histones/metabolism , Insect Proteins/metabolism , Nucleosomes/metabolism , Repressor Proteins/metabolism , Animals , Base Sequence , Binding Sites , DNA/metabolism , Drosophila , Molecular Sequence Data , Polycomb Repressive Complex 1 , Protein Structure, Tertiary , Trypsin
10.
Science ; 286(5441): 955-8, 1999 Oct 29.
Article in English | MEDLINE | ID: mdl-10542150

ABSTRACT

The Drosophila Polycomb and trithorax group proteins act through chromosomal elements such as Fab-7 to maintain repressed or active gene expression, respectively. A Fab-7 element is switched from a silenced to a mitotically heritable active state by an embryonic pulse of transcription. Here, histone H4 hyperacetylation was found to be associated with Fab-7 after activation, suggesting that H4 hyperacetylation may be a heritable epigenetic tag of the activated element. Activated Fab-7 enables transcription of a gene even after withdrawal of the primary transcription factor. This feature may allow epigenetic maintenance of active states of developmental genes after decay of their early embryonic regulators.


Subject(s)
Chromatin/genetics , DNA-Binding Proteins/physiology , Drosophila Proteins , Drosophila/genetics , Insect Proteins/physiology , Saccharomyces cerevisiae Proteins , Trans-Activators/physiology , Acetylation , Animals , Animals, Genetically Modified , Drosophila/embryology , Fungal Proteins/metabolism , Gene Expression Regulation, Developmental , Histones/metabolism , Lac Operon , Polycomb Repressive Complex 1 , Regulatory Sequences, Nucleic Acid , Transcription Factors/metabolism
11.
Bioessays ; 21(10): 824-32, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10497332

ABSTRACT

Epigenetic regulation of transcription can lead to a stable differential expression of identical genetic information in the same cell or cell population. There is increasing evidence that higher order chromatin structures, involving specific multiprotein complexes, constitute one device to establish and maintain epigenetic marks. In addition, defined chromosomal elements conferring epigenetic inheritance of transcriptional expression states have recently been identified. During the period where the difference in expression of identical genes is established, these sequences appear to be used as switch elements by both negative and positive regulators. Once the epigenetic mark is "set", the elements maintain either the silenced or the activated expression state over many cell generations. Here we review recent data obtained from analyzing epigenetic gene regulation in different organisms and show that similarities in the underlying mechanisms appear to exist.


Subject(s)
Chromosomes/genetics , Alleles , Animals , Drosophila/genetics , Female , Gene Expression Regulation, Developmental , Genes, Switch , Genomic Imprinting , Heterochromatin/genetics , Male , Models, Genetic , Saccharomyces cerevisiae/genetics
12.
Chromosoma ; 108(2): 83-94, 1999 May.
Article in English | MEDLINE | ID: mdl-10382070

ABSTRACT

The chromatin protein Polycomb (PC) is necessary for keeping homeotic genes repressed in a permanent and heritable manner. PC is part of a large multimeric complex (PcG proteins) involved in generating silenced chromatin domains at target genes, thus preventing their inappropriate expression. In order to assess the intranuclear distribution of PC during mitosis in different developmental stages as well as in the germ line we generated transgenic fly lines expressing a PC-GFP (Green Fluorescent Protein) fusion protein. Rapidly dividing nuclei were found to display a rather homogeneous PC-GFP distribution. However, with increasing differentiation a pronounced subnuclear pattern was observed. In all investigated diploid somatic tissues the bulk of PC-GFP fusion protein is depleted from the chromosomes during mitosis: however, a detectable fraction remains associated. In the male germ line in early spermatogenesis, PC-GFP was closely associated with the chromosomal bivalents and gradually lost at later stages. Interestingly, we found that PC is associated with the nucleolus in spermatocytes, unlike somatic nuclei. In contrast to mature sperm showing no PC-GFP signal the female germ line retains PC in the germinal vesicle.


Subject(s)
Cell Nucleus/metabolism , Drosophila Proteins , Drosophila melanogaster/metabolism , Insect Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Animals , Animals, Genetically Modified , Drosophila melanogaster/embryology , Female , Germ Cells , Green Fluorescent Proteins , Insect Proteins/genetics , Interphase , Luminescent Proteins/genetics , Male , Microscopy, Fluorescence , Polycomb Repressive Complex 1 , Recombinant Fusion Proteins/genetics
14.
Proc Natl Acad Sci U S A ; 95(23): 13703-8, 1998 Nov 10.
Article in English | MEDLINE | ID: mdl-9811864

ABSTRACT

The importance of the amyloid precursor protein (APP) in the pathogenesis of Alzheimer's disease (AD) became apparent through the identification of distinct mutations in the APP gene, causing early onset familial AD with the accumulation of a 4-kDa peptide fragment (betaA4) in amyloid plaques and vascular deposits. However, the physiological role of APP is still unclear. In this work, Drosophila melanogaster is used as a model system to analyze the function of APP by expressing wild-type and various mutant forms of human APP in fly tissue culture cells as well as in transgenic fly lines. After expression of full-length APP forms, secretion of APP but not of betaA4 was observed in both systems. By using SPA4CT, a short APP form in which the signal peptide was fused directly to the betaA4 region, transmembrane domain, and cytoplasmic tail, we observed betaA4 release in flies and fly-tissue culture cells. Consequently, we showed a gamma-secretase activity in flies. Interestingly, transgenic flies expressing full-length forms of APP have a blistered-wing phenotype. As the wing is composed of interacting dorsal and ventral epithelial cell layers, this phenotype suggests that human APP expression interferes with cell adhesion/signaling pathways in Drosophila, independently of betaA4 generation.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Drosophila melanogaster/genetics , Endopeptidases/genetics , Gene Expression Regulation , Genes, Insect , Amyloid Precursor Protein Secretases , Animals , Animals, Genetically Modified , Aspartic Acid Endopeptidases , Cell Adhesion/genetics , Humans , Phenotype
15.
EMBO J ; 17(17): 5141-50, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9724650

ABSTRACT

In Drosophila, the maintenance of developmentally important transcription patterns is controlled at the level of chromatin structure. The Polycomb group (PcG) and trithorax group (trxG) genes encode proteins involved in chromatin remodelling. PcG genes have been proposed to act by packaging transcriptional repressed chromosomal domains into condensed heterochromatin-like structures. Some of the trxG proteins characterized so far are members of chromatin opening complexes (e.g. SWI/SNF and GAGA/NURF) which facilitate binding of transcription factors and components of the basal transcriptional machinery. Genetic and biochemical data suggest that these two groups of regulatory factors may act through a common set of DNA elements. In the present study, we have investigated the binding of Trithorax (TRX) and Polycomb (PC) protein in the bithorax complex (BX-C) during embryogenesis. In addition, we have identified the minimal fragments from the Ultrabithorax (Ubx) regulatory region that are capable of recruiting TRX to chromosomal sites containing them. Comparative analysis of the binding of the two proteins shows that TRX and PC bind target sequences (PcG-regulated elements, PREs) by cellular blastoderm, when BX-C transcription begins. At the same stage, TRX but not PC is strongly associated with core promoters. Later, at germ band extension, the time of derepression in Polycomb mutants, PC binding is also detected outside core PREs and additionally binds to the fragments containing promoters.


Subject(s)
Chromatin/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins , Drosophila/embryology , Homeodomain Proteins/metabolism , Insect Proteins/metabolism , Transcription Factors , Animals , Blastoderm , Polycomb Repressive Complex 1 , Protein Binding , Regulatory Sequences, Nucleic Acid , Time Factors , Transcription, Genetic
16.
Curr Opin Cell Biol ; 10(3): 354-60, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9640536

ABSTRACT

Chromo-domain proteins appear to be a central component in the epigenetic regulation of heterochromatin function and euchromatic gene expression. The recent discovery of a variety of interacting partners of chromo-domain proteins is yielding new molecular insights into epigenetic regulatory processes acting at the level of higher order chromatin structure.


Subject(s)
Chromatin/genetics , Chromosomal Proteins, Non-Histone , Gene Expression Regulation/physiology , Repressor Proteins , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data
17.
Cell ; 93(4): 505-18, 1998 May 15.
Article in English | MEDLINE | ID: mdl-9604927

ABSTRACT

Polycomb group (PcG) and trithorax group (trxG) gene products are responsible for the maintenance of repressed and active expression patterns of many developmentally important regulatory genes including the homeotic genes. In Drosophila embryos, Polycomb protein and the trxG protein GAGA factor colocalize at the Fab-7 DNA element of the bithorax complex. In transgenic lines, the Fab-7 element induces extensive silencing on a flanking GAL4-driven lacZ reporter and mini-white genes. However, a short single pulse of GAL4 during embryogenesis is sufficient to release PcG-dependent silencing from the transgene. Such an activated state of Fab-7 is mitotically inheritable through development and can be transmitted in a GAL4-independent manner to the subsequent generations through female meiosis. Thus, Fab-7 is a switchable chromosomal element, which can convey memory of epigenetically determined active and repressed chromatin states.


Subject(s)
ATP-Binding Cassette Transporters , Drosophila Proteins , Drosophila/genetics , Meiosis/genetics , Mitosis/genetics , Nuclear Proteins , Regulatory Sequences, Nucleic Acid/genetics , Saccharomyces cerevisiae Proteins , Animals , Animals, Genetically Modified , Chromatin/genetics , Crosses, Genetic , DNA/metabolism , DNA-Binding Proteins/genetics , Drosophila/embryology , Eye Color/genetics , Eye Proteins/genetics , Female , Fungal Proteins/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insect Proteins/genetics , Insect Proteins/metabolism , Larva , Molecular Sequence Data , Polycomb Repressive Complex 1 , Recombinant Fusion Proteins , Repressor Proteins/metabolism , Transcription Factors/metabolism
18.
Novartis Found Symp ; 214: 51-61; discussion 61-6, 104-13, 1998.
Article in English | MEDLINE | ID: mdl-9601011

ABSTRACT

In Drosophila the Polycomb group (PcG) and trithorax group (trxG) genes are required to maintain differential expression patterns of many important developmental regulatory genes. The PcG is responsible for heritable silencing throughout development. At target genes PcG response elements (PREs) attract PcG protein complexes and induce the formation of higher-order chromatin structures. We have mapped the distribution of Polycomb and other PcG members at various target genes by using an improved formaldehyde cross-linking and chromatin immunoprecipitation technique. We find that Polycomb spreads locally from PREs over several kilobases, thereby probably stabilizing the silencing complexes. Members of the trxG co-localize at PREs. GAGA factor was found to be constitutively bound to PREs independently of gene activity. PREs associated with active genes appear to have increased amounts of bound GAGA. We have developed a system capable of switching a PRE between the on/off modes. PREs and trxG-regulated elements are common chromosomal elements through which the proteins of the PcG/trxG exert their maintenance function on adjacent chromatin structures.


Subject(s)
Chromatin/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins , Insect Proteins/genetics , Animals , DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Humans , Insect Proteins/metabolism , Polycomb Repressive Complex 1 , Transcription Factors/metabolism
19.
Novartis Found Symp ; 214: 233-44; discussion 244-50, 1998.
Article in English | MEDLINE | ID: mdl-9601021

ABSTRACT

H19 and Igf2 are located within a large imprinting domain that confers monoallelic silencing of parental alleles. The silent paternal allele of H19 is hypermethylated and relatively resistant to nucleases. Using a 130 kb yeast artificial chromosome clone, appropriate imprinting of both H19 and Igf2 was observed at single insert loci in transgenic mice. Imprinting was also observed for H19-lacZ transgenes containing 4 kb of upstream sequence, but only at multicopy loci. The H19 RNA is therefore not essential for imprinting. When the H19-lacZ transgene was introduced into Drosophila, a 1.2 kb region was identified within the 4 kb upstream flank that functioned as a bi-directional silencer. This cis element is located within a region that is apparently necessary for imprinting in mice. These studies suggest an evolutionarily conserved mechanism for gene silencing in Drosophila and imprinting in mice. We propose a new model for imprinting of H19 and Igf2 in mice in which silencing of H19 is the default state, and activation of the maternal allele requires a specific activator element.


Subject(s)
Gene Expression , Genomic Imprinting , RNA, Untranslated , Animals , Chromosomes , Chromosomes, Artificial, Yeast , Drosophila/genetics , Genes, Reporter , Insulin-Like Growth Factor II/genetics , Lac Operon , Mice , Muscle Proteins/genetics , RNA, Long Noncoding
20.
Proc Natl Acad Sci U S A ; 95(4): 1698-702, 1998 Feb 17.
Article in English | MEDLINE | ID: mdl-9465079

ABSTRACT

Prader-Willi syndrome (PWS) and Angelman syndrome are neurogenetic disorders caused by the lack of a paternal or a maternal contribution from human chromosome 15q11-q13, respectively. Deletions in the transcription unit of the imprinted SNRPN gene have been found in patients who have PWS or Angelman syndrome because of a parental imprint switch failure in this chromosomal domain. It has been suggested that the SNRPN exon 1 region, which is deleted in the PWS patients, contains an imprint switch element from which the maternal and paternal epigenotypes of the 15q11-q13 domain originate. Using the model organism Drosophila, we show here that a fragment from this region can function as a silencer in transgenic flies. Repression was detected specifically from this element and could not be observed with control human sequences. Additional experiments allowed the delineation of the silencer to a fragment of 215 bp containing the SNRPN promoter region. These results provide an additional link between genomic imprinting and an evolutionary conserved silencing mechanism. We suggest that the identified element participates in the long range regulation of the imprinted 15q11-q13 domain or locally represses SNRPN expression from the maternal allele.


Subject(s)
Autoantigens/genetics , Chromosomes, Human, Pair 15 , Gene Expression Regulation , Genomic Imprinting , Ribonucleoproteins, Small Nuclear , Alleles , Animals , Animals, Genetically Modified , Biological Evolution , Drosophila melanogaster/genetics , Humans , Prader-Willi Syndrome/genetics , snRNP Core Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...