Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Parasit Vectors ; 7: 428, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25190631

ABSTRACT

BACKGROUND: Parascaris univalens is an ascaridoid nematode of equids. Little is known about its epidemiology and population genetics in domestic and wild horse populations. PCR-based methods are suited to support studies in these areas, provided that reliable genetic markers are used. Recent studies have shown that mitochondrial (mt) genomic markers are applicable in such methods, but no such markers have been defined for P. univalens. METHODS: Mt genome regions were amplified from total genomic DNA isolated from P. univalens eggs by long-PCR and sequenced using Illumina technology. The mt genome was assembled and annotated using an established bioinformatic pipeline. Amino acid sequences inferred from all protein-encoding genes of the mt genomes were compared with those from other ascaridoid nematodes, and concatenated sequences were subjected to phylogenetic analysis by Bayesian inference. RESULTS: The circular mt genome was 13,920 bp in length and contained two ribosomal RNA, 12 protein-coding and 22 transfer RNA genes, consistent with those of other ascaridoids. Phylogenetic analysis of the concatenated amino acid sequence data for the 12 mt proteins showed that P. univalens was most closely related to Ascaris lumbricoides and A. suum, to the exclusion of other ascaridoids. CONCLUSIONS: This mt genome representing P. univalens now provides a rich source of genetic markers for future studies of the genetics and epidemiology of this parasite and its congener, P. equorum. This focus is significant, given that there is no published information on the specific prevalence and distribution of P. univalens infection in domestic and wild horse populations.


Subject(s)
Ascaridoidea/genetics , DNA, Helminth/genetics , Genome, Mitochondrial/genetics , Amino Acid Sequence , Animals , Phylogeny
2.
BMC Biol ; 12: 30, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24779377

ABSTRACT

BACKGROUND: Epimorphic regeneration of a missing appendage in fish and urodele amphibians involves the creation of a blastema, a heterogeneous pool of progenitor cells underneath the wound epidermis. Current evidence indicates that the blastema arises by dedifferentiation of stump tissues in the vicinity of the amputation. In response to tissue loss, silenced developmental programs are reactivated to form a near-perfect copy of the missing body part. However, the importance of chromatin regulation during epimorphic regeneration remains poorly understood. RESULTS: We found that specific components of the Nucleosome Remodeling and Deacetylase complex (NuRD) are required for fin regeneration in zebrafish. Transcripts of the chromatin remodeler chd4a/Mi-2, the histone deacetylase hdac1/HDAC1/2, the retinoblastoma-binding protein rbb4/RBBP4/7, and the metastasis-associated antigen mta2/MTA were specifically co-induced in the blastema during adult and embryonic fin regeneration, and these transcripts displayed a similar spatial and temporal expression patterns. In addition, chemical inhibition of Hdac1 and morpholino-mediated knockdown of chd4a, mta2, and rbb4 impaired regenerative outgrowth, resulting in reduction in blastema cell proliferation and in differentiation defects. CONCLUSION: Altogether, our data suggest that specialized NuRD components are induced in the blastema during fin regeneration and are involved in blastema cell proliferation and redifferentiation of osteoblast precursor cells. These results provide in vivo evidence for the involvement of key epigenetic factors in the cellular reprogramming processes occurring during epimorphic regeneration in zebrafish.


Subject(s)
Animal Fins/physiology , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Regeneration/physiology , Zebrafish Proteins/metabolism , Zebrafish/physiology , Animal Fins/drug effects , Animals , Benzamides/pharmacology , Biomarkers/metabolism , Body Patterning/drug effects , Body Patterning/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Gene Knockdown Techniques , Genome/genetics , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Morpholinos/pharmacology , Osteoblasts/cytology , Osteoblasts/drug effects , Pyrimidines/pharmacology , Regeneration/drug effects , Regeneration/genetics , Sequence Homology, Amino Acid , Up-Regulation/drug effects , Up-Regulation/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
3.
Stem Cell Reports ; 2(4): 547-59, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24749077

ABSTRACT

Throughout their journey to forming new individuals, germline stem cells must remain totipotent, particularly by maintaining a specific chromatin structure. However, the place epigenetic factors occupy in this process remains elusive. So far, "sensitization" of chromatin by modulation of histone arrangement and/or content was believed to facilitate transcription-factor-induced germ cell reprogramming. Here, we demonstrate that the combined reduction of two epigenetic factors suffices to reprogram C. elegans germ cells. The histone H3K4 demethylase SPR-5/LSD1 and the chromatin remodeler LET-418/Mi2 function together in an early process to maintain germ cell status and act as a barrier to block precocious differentiation. This epigenetic barrier is capable of limiting COMPASS-mediated H3K4 methylation, because elevated H3K4me3 levels correlate with germ cell reprogramming in spr-5; let-418 mutants. Interestingly, germ cells deficient for spr-5 and let-418 mainly reprogram as neurons, suggesting that neuronal fate might be the first to be derepressed in early embryogenesis.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Cellular Reprogramming , DNA-Binding Proteins/metabolism , Germ Cells/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Oxidoreductases, N-Demethylating/metabolism , Stem Cells/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Cell Differentiation/genetics , Cell Division/genetics , Cell Transformation, Neoplastic/genetics , Chromatin/metabolism , DNA-Binding Proteins/genetics , Down-Regulation , Female , Gene Expression Regulation , Germ Cells/cytology , Histones/metabolism , Male , Methylation , Mutation , Neoplasms, Germ Cell and Embryonal/genetics , Neurons/cytology , Neurons/metabolism , Oxidoreductases, N-Demethylating/genetics , Protein Binding , Stem Cells/cytology
4.
Aging Cell ; 12(6): 1012-20, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23815345

ABSTRACT

The evolutionarily conserved nucleosome-remodeling protein Mi2 is involved in transcriptional repression during development in various model systems, plays a role in embryonic patterning and germ line development, and participates in DNA repair and cell cycle progression. It is the catalytic subunit of the nucleosome remodeling and histone deacetylase (NuRD) complex, a key determinant of differentiation in mammalian embryonic stem cells. In addition, the Drosophila and C. elegans Mi2 homologs participate in another complex, the MEC complex, which also plays an important developmental role in these organisms. Here we show a new and unexpected feature of the C. elegans Mi2 homolog, LET-418/Mi2. Lack of LET-418/Mi2 results in longevity and enhanced stress resistance, a feature that we found to be conserved in Drosophila and in Arabidopsis. The fact that depletion of other components of the NuRD and the MEC complexes did not result in longevity suggests that LET-418 may regulate lifespan in a different molecular context. Genetic interaction studies suggest that let-418 could act in the germ-cell-loss pathway, downstream of kri-1 and tcer-1. On the basis of our data and on previous findings showing a role for let-418 during development, we propose that LET-418/Mi2 could be part of a system that drives development and reproduction with concomitant life-reducing effects later in life.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , DNA-Binding Proteins/metabolism , Longevity/physiology , Animals , Arabidopsis/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Conserved Sequence , Drosophila melanogaster , Environment , Evolution, Molecular , Insulin/metabolism , Oxidative Stress , Protein Binding , Sequence Analysis, Protein , Signal Transduction , Stress, Physiological/genetics , Up-Regulation
5.
Development ; 138(21): 4649-60, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21989912

ABSTRACT

Morphogenesis represents a phase of development during which cell fates are executed. The conserved hox genes are key cell fate determinants during metazoan development, but their role in controlling organ morphogenesis is less understood. Here, we show that the C. elegans hox gene lin-39 regulates epidermal morphogenesis via its novel target, the essential zinc finger protein VAB-23. During the development of the vulva, the egg-laying organ of the hermaphrodite, the EGFR/RAS/MAPK signaling pathway activates, together with LIN-39 HOX, the expression of VAB-23 in the primary cell lineage to control the formation of the seven vulval toroids. VAB-23 regulates the formation of homotypic contacts between contralateral pairs of cells with the same sub-fates at the vulval midline by inducing smp-1 (semaphorin) transcription. In addition, VAB-23 prevents ectopic vulval cell fusions by negatively regulating expression of the fusogen eff-1. Thus, LIN-39 and the EGFR/RAS/MAPK signaling pathway, which specify cell fates earlier during vulval induction, continue to act during the subsequent phase of cell fate execution by regulating various aspects of epidermal morphogenesis. Vulval cell fate specification and execution are, therefore, tightly coupled processes.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/anatomy & histology , Caenorhabditis elegans/embryology , Carrier Proteins/metabolism , ErbB Receptors/metabolism , Homeodomain Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Morphogenesis/physiology , Signal Transduction/physiology , Animals , Base Sequence , Biomarkers/metabolism , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Carrier Proteins/genetics , Cell Fusion , Cell Lineage , ErbB Receptors/genetics , Gene Expression Regulation, Developmental , Genes, Reporter , Homeodomain Proteins/genetics , Intracellular Signaling Peptides and Proteins , Mitogen-Activated Protein Kinases/genetics , Molecular Sequence Data , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Semaphorins/genetics , Semaphorins/metabolism , Sequence Alignment , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc Fingers
6.
PLoS One ; 5(10): e13681, 2010 Oct 27.
Article in English | MEDLINE | ID: mdl-21060680

ABSTRACT

Biochemical purifications from mammalian cells and Xenopus oocytes revealed that vertebrate Mi-2 proteins reside in multisubunit NuRD (Nucleosome Remodeling and Deacetylase) complexes. Since all NuRD subunits are highly conserved in the genomes of C. elegans and Drosophila, it was suggested that NuRD complexes also exist in invertebrates. Recently, a novel dMec complex, composed of dMi-2 and dMEP-1 was identified in Drosophila. The genome of C. elegans encodes two highly homologous Mi-2 orthologues, LET-418 and CHD-3. Here we demonstrate that these proteins define at least three different protein complexes, two distinct NuRD complexes and one MEC complex. The two canonical NuRD complexes share the same core subunits HDA-1/HDAC, LIN-53/RbAp and LIN-40/MTA, but differ in their Mi-2 orthologues LET-418 or CHD-3. LET-418 but not CHD-3, interacts with the Krüppel-like protein MEP-1 in a distinct complex, the MEC complex. Based on microarrays analyses, we propose that MEC constitutes an important LET-418 containing regulatory complex during C. elegans embryonic and early larval development. It is required for the repression of germline potential in somatic cells and acts when blastomeres are still dividing and differentiating. The two NuRD complexes may not be important for the early development, but may act later during postembryonic development. Altogether, our data suggest a considerable complexity in the composition, the developmental function and the tissue-specificity of the different C. elegans Mi-2 complexes.


Subject(s)
Adenosine Triphosphatases/physiology , Autoantigens/physiology , Caenorhabditis elegans/embryology , Drosophila Proteins/physiology , Animals , Caenorhabditis elegans/genetics , Germ Cells , Green Fluorescent Proteins/genetics
7.
BMC Dev Biol ; 10: 78, 2010 Jul 28.
Article in English | MEDLINE | ID: mdl-20667114

ABSTRACT

BACKGROUND: Hox genes play a central role in axial patterning during animal development. They are clustered in the genome and specify cell fate in sequential domains along the anteroposterior (A-P) body axis in a conserved order that is co-linear with their relative genomic position. In the soil worm Caenorhabditis elegans, this striking rule of co-linearity is broken by the anterior Hox gene ceh-13, which is located between the two middle Hox paralogs, lin-39 and mab-5, within the loosely organized nematode Hox cluster. Despite its evolutionary and developmental significance, the functional consequence of this unusual genomic organization remains unresolved. RESULTS: In this study we have investigated the role of ceh-13 in different developmental processes, and found that its expression and function are not restricted to the anterior body part. We show that ceh-13 affects cell migration and fusion as well as tissue patterning in the middle and posterior body regions too. These data reveal novel roles for ceh-13 in developmental processes known to be under the control of middle Hox paralogs. Consistently, enhanced activity of lin-39 and mab-5 can suppress developmental arrest and morphologic malformation in ceh-13 deficient animals. CONCLUSION: Our findings presented here show that, unlike other Hox genes in C. elegans which display region-specific accumulation and function along the A-P axis, the expression and functional domain of the anterior Hox paralog ceh-13 extends beyond the anterior region of the worm. Furthermore, ceh-13 and the middle Hox paralogs share several developmental functions. Together, these results suggest the emergence of the middle-group Hox genes from a ceh-13-like primordial Hox ancestor.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/embryology , Cell Movement , Evolution, Molecular , Genes, Homeobox , Homeodomain Proteins/metabolism , Animals , Caenorhabditis elegans/cytology , Embryo, Nonmammalian/metabolism
8.
Dev Biol ; 344(2): 593-602, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20478293

ABSTRACT

In the germ line of the Caenorhabditis elegans hermaphrodite, nuclei either proliferate through mitosis or initiate meiosis, finally differentiating as spermatids or oocytes. The production of oocytes requires repression of the fem-3 mRNA by cytoplasmic FBF and nuclear MOG proteins. Here we report the identification of the sex determining gene mog-3 and show that in addition to its role in gamete sex determination, it is necessary for meiosis by acting downstream of GLP-1/Notch. Furthermore, we found that MOG-3 binds both to the nuclear proteins MEP-1 and CIR-1. MEP-1 is necessary for oocyte production and somatic differentiation, while the mammalian CIR-1 homolog counters Notch signaling. We propose that MOG-3, MEP-1 and CIR-1 associate in a nuclear complex which regulates different aspects of germ cell development. While FBF triggers the sperm/oocyte switch by directly repressing the fem-3 mRNA in the cytoplasm, the MOG proteins play a more indirect role in the nucleus, perhaps by acting as epigenetic regulators or by controlling precise splicing events.


Subject(s)
Caenorhabditis elegans , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/physiology , Co-Repressor Proteins , Germ Cells/metabolism , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Male , Meiosis , Oocytes/metabolism , Oocytes/physiology , RNA Splicing , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spermatozoa/metabolism
9.
Dev Biol ; 306(2): 469-79, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17466968

ABSTRACT

The fate of the vulval cells in Caenorhabditis elegans is specified, at least in part, through a highly conserved RTK/Ras mediated signaling cascade that negatively regulates the activity of the ETS-like transcription factor LIN-1. The Hox gene lin-39 functions downstream of both, the LIN-3/RTK/Ras pathway and LIN-1 and plays a pivotal role in controlling vulva cell competence and induction. Here we show that LET-418, a C. elegans ortholog of the human NuRD component Mi-2, negatively modulates the activity of lin-39. LET-418 interacts in vivo with specific regions in the promoter of lin-39 and this interaction depends on LIN-1. Our data provide evidence for a model in which LIN-1 recruits LET-418/Mi-2 as co-repressor to the promoter of lin-39, thereby restricting its activity to the basal levels required in the vulva precursor cells (VPCs) for normal vulval development. Thus, our data suggest that the interaction between LIN-1 and LET-418/Mi-2 may link RTK/Ras signaling with chromatin remodeling and gene expression.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/metabolism , DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins/physiology , Nucleosomes/metabolism , Promoter Regions, Genetic , Transcription Factors/metabolism , Vulva/embryology , Animals , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , Chromatin/metabolism , DNA-Binding Proteins/chemistry , Female , Models, Genetic , Mutation , Phenotype , RNA Interference
10.
Dev Biol ; 302(2): 661-9, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17084835

ABSTRACT

The Notch signaling pathway controls growth, differentiation and patterning in divergent animal phyla; in humans, defective Notch signaling has been implicated in cancer, stroke and neurodegenerative disorders. Despite its developmental and medical significance, little is known about the factors that render cells to become competent for Notch signaling. Here we show that during vulval development in the nematode Caenorhabditis elegans the HOX protein LIN-39 and its EXD/PBX-like cofactor CEH-20 are required for LIN-12/Notch-mediated lateral signaling that specifies the 2 degrees vulval cell fate. Inactivation of either lin-39 or ceh-20 resulted in the misspecification of 2 degrees vulval cells and suppressed the multivulva phenotype of lin-12(n137) gain-of-function mutant animals. Furthermore, both LIN-39 and CEH-20 are required for the expression of basal levels of the genes encoding the LIN-12/Notch receptor and one of its ligands in the vulval precursor cells, LAG-2/Delta/Serrate, rendering them competent for the subsequent lin-12/Notch induction events. Our results suggest that the transcription factors LIN-39 and CEH-20, which function at the bottom of the RTK/Ras and Wnt pathways in vulval induction, serve as major integration sites in coordinating and transmitting signals to the LIN-12/Notch cascade to regulate vulval cell fates.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Homeodomain Proteins/physiology , Receptors, Notch/physiology , Transcription Factors/physiology , Transcription, Genetic , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/genetics , Female , Homeodomain Proteins/genetics , Larva , Membrane Proteins/genetics , Membrane Proteins/physiology , Signal Transduction , Transcription Factors/genetics , Vulva/growth & development , Vulva/physiology
11.
Nature ; 442(7105): 882, 2006 Aug 24.
Article in English | MEDLINE | ID: mdl-16929289

ABSTRACT

Small RNA molecules participate in a variety of activities in the cell: in a process known as RNA interference (RNAi), double-stranded RNA triggers the degradation of messenger RNA that has a matching sequence; the small RNA intermediates of this process can also modify gene expression in the nucleus. Here we show that a single episode of RNAi in the nematode Caenorhabditis elegans can induce transcriptional silencing effects that are inherited indefinitely in the absence of the original trigger. Our findings may prove useful in the ongoing development of RNAi to treat disease.


Subject(s)
Caenorhabditis elegans/genetics , Genes, Helminth/genetics , Heredity/genetics , RNA Interference , Animals , Caenorhabditis elegans Proteins/genetics , Genes, Dominant/genetics , Green Fluorescent Proteins/genetics , Homeodomain Proteins/genetics , Male , Phenotype , Transcription, Genetic/genetics , Transgenes/genetics
12.
Curr Biol ; 15(16): 1513-7, 2005 Aug 23.
Article in English | MEDLINE | ID: mdl-16111945

ABSTRACT

Programmed cell death (PCD) is an essential and highly orchestrated process that plays a major role in morphogenesis and tissue homeostasis during development. In humans, defects in regulation or execution of cell death lead to diabetes, neurodegenerative disorders, and cancer. Two major types of PCD have been distinguished: the caspase-mediated process of apoptosis and the caspase-independent process involving autophagy. Although apoptosis and autophagy are often activated together in response to stress, the molecular mechanisms underlying their interplay remain unclear. Here we show that BEC-1, the C. elegans ortholog of the yeast and mammalian autophagy proteins Atg6/Vps30 and Beclin 1, is essential for development. We demonstrate that BEC-1 is necessary for the function of the class III PI3 kinase LET-512/Vps34, an essential protein required for autophagy, membrane trafficking, and endocytosis. Furthermore, BEC-1 forms a complex with the antiapoptotic protein CED-9/Bcl-2, and its depletion triggers CED-3/Caspase-dependent PCD. Based on our results, we propose that bec-1 represents a link between autophagy and apoptosis, thus supporting the view that the two processes act in concerted manner in the cell death machinery.


Subject(s)
Apoptosis/genetics , Autophagy/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Gene Silencing , Animals , Blotting, Western , Immunoprecipitation , Phosphatidylinositol 3-Kinases/metabolism , RNA Interference , Vesicular Transport Proteins , bcl-Associated Death Protein/metabolism
13.
Mol Cell Biol ; 24(11): 5016-27, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15143192

ABSTRACT

Bloom's syndrome (BS) is an autosomal-recessive human disorder caused by mutations in the BS RecQ helicase and is associated with loss of genomic integrity and an increased incidence of cancer. We analyzed the mitotic and the meiotic roles of Caenorhabditis elegans him-6, which we show to encode the ortholog of the human BS gene. Mutations in him-6 result in an enhanced irradiation sensitivity, a partially defective S-phase checkpoint, and in reduced levels of DNA-damage induced apoptosis. Furthermore, him-6 mutants exhibit a decreased frequency of meiotic recombination that is probably due to a defect in the progression of crossover recombination. In mitotically proliferating germ cells, our genetic interaction studies, as well as the assessment of the number of double-strand breaks via RAD-51 foci, reveal a complex regulatory network that is different from the situation in yeast. Although the number of double-strand breaks in him-6 and top-3 single mutants is elevated, the combined depletion of him-6 and top-3 leads to mitotic catastrophe concomitant with a massive increase in the level of double-strand breaks, a phenotype that is completely suppressed by rad-51. him-6 and top-3 are thus needed to maintain low levels of double-strand breaks in normally proliferating germ cells, and both act in partial redundant pathways downstream of rad-51 to prevent mitotic catastrophy. Finally, we show that topoisomerase IIIalpha acts independently during a late stage of meiotic recombination.


Subject(s)
Bloom Syndrome/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , DNA Topoisomerases, Type I/genetics , DNA-Binding Proteins/genetics , Adenosine Triphosphatases/genetics , Animals , Bloom Syndrome/metabolism , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , DNA Damage , DNA Helicases/genetics , DNA Topoisomerases, Type I/metabolism , DNA-Binding Proteins/metabolism , Meiosis/physiology , Rad51 Recombinase , RecQ Helicases , Recombination, Genetic/physiology
14.
Mol Biochem Parasitol ; 134(1): 53-64, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14747143

ABSTRACT

Chromatin diminution in the parasitic nematodes Ascaris suum and Parascaris univalens represents a rather complex molecular phenomenon that includes chromosomal breakage, DNA degradation and new telomere formation. At a given elimination site, DNA breakage and new telomere addition does not take place at a single chromosomal locus but at many different places within a several kilobase long chromosomal region, referred to as chromosomal breakage region (CBR). Here we describe the cloning and the characterisation of seven CBRs from A. suum and P. univalens and we show that the process has been conserved between the two species. A detailed sequence comparison provides evidence that the sequences of the CBRs and their flanking regions are not directly important for the specification of the elimination sites. Six out of the seven CBRs are conserved between the two nematode species, suggesting that they have already existed in a common ancestor. We present a hypothesis stating that the elimination process ensures the maintenance of a functional somatic genome and concomitantly allows extremely rapid and profound changes in the germ line genome, thereby allowing the development of new germ line specific functions and thus providing a selective advantage for the chromatin eliminating nematodes during further evolution.


Subject(s)
Ascaridoidea/genetics , Chromatin/metabolism , Evolution, Molecular , Genome , Animals , Ascaris suum/genetics , Chromosome Breakage , Chromosomes/genetics , Chromosomes/metabolism , Conserved Sequence , DNA, Helminth/chemistry , DNA, Helminth/isolation & purification , DNA, Helminth/metabolism , Gene Rearrangement , Germ Cells/growth & development , Molecular Sequence Data , Sequence Analysis, DNA , Sequence Homology , Telomere/genetics , Telomere/metabolism
16.
Dev Biol ; 259(1): 137-49, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12812794

ABSTRACT

Hox genes are transcriptional regulators of metazoan body regionalization along the anteroposterior axis that act by specifying positional identity in differentiating cells. ceh-13, the labial orthologue in Caenorhabditis elegans, is expressed both during embryogenesis and post- embryonic development. Using GFP reporter analysis and immunocytochemistry, we discovered a spatiotemporal pattern of gene expression in the male tail during the L3 and L4 larval stages that is TGF-beta pathway-dependent. Analysis of reporter activity in transgenic animals identified a distinct promoter region driving male tail-specific ceh-13 expression. We also report the interspecies conservation of sequence motifs within this region and speculate that, in the course of evolutionary diversification, ceh-13 may have acquired new functionality while conserving its homeotic role.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/embryology , Homeodomain Proteins/physiology , Tail/embryology , Transforming Growth Factor beta , Animals , Base Sequence , Conserved Sequence , Enhancer Elements, Genetic , Male , Molecular Sequence Data , Morphogenesis , Neuropeptides/physiology , Promoter Regions, Genetic , Rectum/embryology
17.
Curr Biol ; 12(17): 1448-61, 2002 Sep 03.
Article in English | MEDLINE | ID: mdl-12225660

ABSTRACT

BACKGROUND: TOR is a phosphatidylinositol kinase (PIK)-related kinase that controls cell growth and proliferation in response to nutritional cues. We describe a C. elegans TOR homolog (CeTOR) and phenotypes associated with CeTOR deficiency. These phenotypes are compared with the response to starvation and the inactivation of a variety of putative TOR targets. RESULTS: Whether caused by mutation or RNA interference, TOR deficiency results in developmental arrest at mid-to-late L3, which is accompanied by marked gonadal degeneration and a pronounced intestinal cell phenotype. A population of refractile, autofluorescent intestinal vesicles, which take up the lysosomal dye Neutral Red, increases dramatically in size, while the number of normal intestinal vesicles and the intestinal cytoplasmic volume decrease progressively. This is accompanied by an increase in the gut lumen size and a compromise in the intestine's ability to digest and absorb nutrients. CeTOR-deficient larvae exhibit no significant dauer characteristics, but share some features with starved L3 larvae. Notably, however, starved larvae do not have severe intestinal atrophy. Inactivation of C. elegans p70S6K or TAP42 homologs does not reproduce CeTOR deficiency phenotypes, nor does inactivation of C. elegans TIP41, a putative negative regulator of CeTOR function, rescue CeTOR deficiency. In contrast, inactivating the C. elegans eIF-4G homolog and eIF-2 subunits results in developmental arrest accompanied by the appearance of large, refractile intestinal vesicles and severe intestinal atrophy resembling that of CeTOR deficiency. CONCLUSIONS: The developmental arrest and intestinal phenotypes of CeTOR deficiency are due to an inhibition of global mRNA translation. Thus, TOR is a major upstream regulator of overall mRNA translation in C. elegans, as in yeast.


Subject(s)
Caenorhabditis elegans Proteins/pharmacology , Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/growth & development , Gene Expression Regulation, Developmental , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Protein Biosynthesis , RNA, Messenger/genetics , Adaptation, Physiological/genetics , Alleles , Animals , Atrophy , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/isolation & purification , Eukaryotic Initiation Factor-2/deficiency , Eukaryotic Initiation Factor-2/physiology , Eukaryotic Initiation Factor-4G/deficiency , Eukaryotic Initiation Factor-4G/physiology , Gene Targeting , Genes, Lethal , Genetic Complementation Test , Intestines/growth & development , Intestines/pathology , Larva , Phenotype , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/isolation & purification , RNA Interference , Ribosomal Protein S6 Kinases, 70-kDa/deficiency , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/physiology , Sequence Homology, Amino Acid , Sirolimus/pharmacology , Species Specificity , Starvation
18.
EMBO J ; 21(7): 1673-83, 2002 Apr 02.
Article in English | MEDLINE | ID: mdl-11927551

ABSTRACT

Here we present a detailed genetic analysis of let-512/vps34 that encodes the Caenorhabditis elegans homologue of the yeast phosphatidylinositol 3-kinase Vps34p. LET-512/VPS34 has essential functions and is ubiquitously expressed in all tissues and developmental stages. It accumulates at a perinuclear region, and mutations in let-512/vps34 result in an expansion of the outer nuclear membrane as well as in a mislocalization and subsequent complete lack of expression of LRP-1, a C.elegans LDL receptor normally associated with the apical surface of hypodermal cells. Using a GFP::2xFYVE fusion protein we found that the phosphatidylinositol 3-phosphate (PtdIns 3-P) product of LET-512/VPS34 is associated with a multitude of intracellular membranes and vesicles located at the periphery, including endocytic vesicles. We propose that LET-512/VPS34 is required for membrane transport from the outer nuclear membrane towards the cell periphery. Thus, LET-512/VPS34 may regulate the secretory pathway in a much broader range of compartments than was previously suggested for the yeast orthologue.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , Nuclear Envelope/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Amino Acid Sequence , Animals , Biological Transport , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Cell Nucleus/metabolism , Endocytosis/physiology , Gene Expression , Molecular Sequence Data , Mutagenesis , Phosphatidylinositol 3-Kinases/genetics , Transport Vesicles/metabolism
19.
Int J Dev Biol ; 46(1): 143-8, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11902675

ABSTRACT

Chromatin diminution in the parasitic nematode Ascaris suum represents an interesting case of developmentally programmed DNA rearrangement in higher eukaryotes. At the molecular level, it is a rather complex event including chromosome breakage, new telomere formation and DNA degradation. Analysis of a cloned somatic telomere (pTel1) revealed that it has been newly created during the process of chromatin diminution by the addition of telomeric repeats (TTAGGC)n to a chromosomal breakage site (Müller et al., 1991). However, telomere addition does not occur at a single chromosomal locus, but at many different sites within a short chromosomal region, termed CBR1 (chromosomal breakage region 1). Here we present the cloning and the analysis of 83 different PCR amplified telomere addition sites from the region of CBR1. The lack of any obvious sequence homology shared among them argues for a telomerase-mediated healing process, rather than for a recombinational event. This hypothesis is strongly supported by the existence of 1-6 nucleotides corresponding to and being in frame with the newly added telomeric repeats at almost all of the telomere addition sites. Furthermore, we show that telomeres are not only added to the ends of the retained chromosomal portions, but also to the eliminated part of the chromosomes, which later on become degraded in the cytoplasm. This result suggests that de novo telomere formation during the process of chromatin diminution represents a non-specific process which can heal any broken DNA end.


Subject(s)
Ascaris suum/embryology , Chromatin/metabolism , DNA, Helminth/biosynthesis , DNA, Helminth/genetics , Gene Expression Regulation, Developmental , Telomere/ultrastructure , Animals , Binding Sites , Cloning, Molecular , DNA, Helminth/metabolism , Models, Genetic , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Analysis, DNA
20.
EMBO Rep ; 3(3): 235-41, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11850401

ABSTRACT

Proteins of the highly conserved heterochromatin protein 1 (HP1) family have been found to function in the dynamic organization of nuclear architecture and in gene regulation throughout the eukaryotic kingdom. In addition to being key players in heterochromatin-mediated gene silencing, HP1 proteins may also contribute to the transcriptional repression of euchromatic genes via the recruitment to specific promoters. To investigate the role played by these different activities in specific developmental pathways, we identified HP1 homologues in the genome of Caenorhabditis elegans and used RNA-mediated interference to study their function. We show that one of the homologues, HPL-2, is required for the formation of a functional germline and for the development of the vulva by acting in an Rb-related pathway. We suggest that, by acting as repressors of gene expression, HP1 proteins may fulfil specific functions in both somatic and germline differentiation processes throughout development.


Subject(s)
Caenorhabditis elegans/physiology , Chromosomal Proteins, Non-Histone/physiology , Amino Acid Sequence , Animals , Caenorhabditis elegans/embryology , Chromobox Protein Homolog 5 , Female , Germ Cells/growth & development , Humans , Molecular Sequence Data , Sequence Alignment , Vulva/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...