Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nat Genet ; 50(2): 193-198, 2018 02.
Article in English | MEDLINE | ID: mdl-29335548

ABSTRACT

The regulation of parental genome dosage is of fundamental importance in animals and plants, as exemplified by X-chromosome inactivation and dosage compensation. The 'triploid block' is a classic example of dosage regulation in plants that establishes a reproductive barrier between species differing in chromosome number1,2. This barrier acts in the embryo-nourishing endosperm tissue and induces the abortion of hybrid seeds through a yet unknown mechanism 3 . Here we show that depletion of paternal epigenetically activated small interfering RNAs (easiRNAs) bypasses the triploid block in response to increased paternal ploidy in Arabidopsis thaliana. Paternal loss of the plant-specific RNA polymerase IV suppressed easiRNA formation and rescued triploid seeds by restoring small-RNA-directed DNA methylation at transposable elements (TEs), correlating with reduced expression of paternally expressed imprinted genes (PEGs). Our data suggest that easiRNAs form a quantitative signal for paternal chromosome number and that their balanced dosage is required for post-fertilization genome stability and seed viability.


Subject(s)
Arabidopsis/genetics , Gene Dosage/genetics , Genome, Plant , RNA, Messenger, Stored/physiology , Gene Expression Regulation, Plant , Genes, Plant , Plants, Genetically Modified , Seeds/genetics
2.
Biol Reprod ; 96(6): 1197-1209, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28575288

ABSTRACT

During oocyte meiotic maturation, Aurora kinase C (AURKC) is required to accomplish many critical functions including destabilizing erroneous kinetochore-microtubule (K-MT)attachments and regulating bipolar spindle assembly. How localized activity of AURKC is regulated in mammalian oocytes, however, is not fully understood. Female gametes from many species, including mouse, contain stores of maternal transcripts that are required for downstream developmental events. We show here that depletion of maternal RNA in mouse oocytes resulted in impaired meiotic progression, increased incidence of chromosome misalignment and abnormal spindle formation at metaphase I (Met I), and cytokinesis defects. Importantly, depletion of maternal RNA perturbed the localization and activity of AURKC within the chromosomal passenger complex (CPC). These perturbations were not observed when translation was inhibited by cycloheximide (CHX) treatment. These results demonstrate a translation-independent function of maternal RNA to regulate AURKC-CPC function in mouse oocytes.


Subject(s)
Aurora Kinase C/metabolism , Oocytes/physiology , Protein Biosynthesis/physiology , RNA, Messenger, Stored/physiology , Animals , Aurora Kinase C/genetics , Cloning, Molecular , Female , Meiosis/physiology , Mice
3.
Biol Reprod ; 87(3): 74, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22786823

ABSTRACT

In most vertebrates, fully grown oocytes are arrested in meiotic prophase I and only resume the cell cycle upon external stimuli, such as hormones. The proper arrest and resumption of the meiotic cycle is critical for reproduction. A Galpha(S) signaling pathway essential for the arrest is conserved in organisms from Xenopus to mouse and human. A previous gene association study implicated that mutations of human ACSL6 may be related to premature ovarian failure. However, functional roles of ACSL6 in human infertility have yet to be reported. In the present study, we found that triacsin C, a potent and specific inhibitor for ACSL, triggers maturation in Xenopus and mouse oocytes in the absence of hormone, suggesting ACSL activity is required for the oocyte arrest. In Xenopus, acsl1b may fulfill a major role in the process, because inhibition of acsl1b by knocking down its RNA results in abnormal acceleration of oocyte maturation. Such abnormally matured eggs cannot support early embryonic development. Moreover, direct inhibition of protein palmitoylation, which lies downstream of ACSLs, also causes oocyte maturation. Furthermore, palmitoylation of Galpha(s), which is essential for its function, is inhibited when the ACSL activity is blocked by triacsin C in Xenopus. Thus, disruption of ACSL activity causes inhibition of the Galpha(s) signaling pathway in the oocytes, which may result in premature ovarian failure in human.


Subject(s)
Acyl Coenzyme A/metabolism , Coenzyme A Ligases/physiology , Meiosis , Xenopus laevis , Animals , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/physiology , Cells, Cultured , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Embryo, Nonmammalian , Enzyme Activation , Female , In Vitro Oocyte Maturation Techniques , Meiosis/genetics , Meiosis/physiology , Mice , Mice, Inbred ICR , Oocytes/metabolism , Oogenesis/genetics , Oogenesis/physiology , RNA, Messenger, Stored/metabolism , RNA, Messenger, Stored/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis/genetics , Xenopus laevis/metabolism , Xenopus laevis/physiology
5.
Development ; 138(18): 3989-4000, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21813572

ABSTRACT

The Xenopus oocyte contains components of both the planar cell polarity and apical-basal polarity pathways, but their roles are not known. Here, we examine the distribution, interactions and functions of the maternal planar cell polarity core protein Vangl2 and the apical-basal complex component aPKC. We show that Vangl2 is distributed in animally enriched islands in the subcortical cytoplasm in full-grown oocytes, where it interacts with a post-Golgi v-SNARE protein, VAMP1, and acetylated microtubules. We find that Vangl2 is required for the stability of VAMP1 as well as for the maintenance of the stable microtubule architecture of the oocyte. We show that Vangl2 interacts with atypical PKC, and that both the acetylated microtubule cytoskeleton and the Vangl2-VAMP1 distribution are dependent on the presence of aPKC. We also demonstrate that aPKC and Vangl2 are required for the cell membrane asymmetry that is established during oocyte maturation, and for the asymmetrical distribution of maternal transcripts for the germ layer and dorsal/ventral determinants VegT and Wnt11. This study demonstrates the interaction and interdependence of Vangl2, VAMP1, aPKC and the stable microtubule cytoskeleton in the oocyte, shows that maternal Vangl2 and aPKC are required for specific oocyte asymmetries and vertebrate embryonic patterning, and points to the usefulness of the oocyte as a model to study the polarity problem.


Subject(s)
Body Patterning/genetics , Membrane Proteins/physiology , Oocytes/metabolism , Protein Kinase C/physiology , RNA, Messenger, Stored/physiology , Xenopus Proteins/physiology , Xenopus , Animals , Animals, Genetically Modified , Embryo, Nonmammalian , Female , Golgi Apparatus/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Biological , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Oocytes/physiology , Protein Binding , Protein Kinase C/genetics , Protein Kinase C/metabolism , RNA, Messenger, Stored/genetics , RNA, Messenger, Stored/metabolism , Tissue Distribution , Vesicle-Associated Membrane Protein 1/metabolism , Xenopus/embryology , Xenopus/genetics , Xenopus/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
6.
Reprod Biomed Online ; 22(6): 512-5, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21498124

ABSTRACT

Genetic knockout studies in the mouse model first revealed the essential role of zygotically derived Cdx2 transcription factor during the later stages of blastocyst formation, characterized by a lack of functioning trophectoderm. However, the extent to which the potential provision of maternally derived Cdx2 affects preimplantation development has proved much less simple to address. Within the last year, two reports have been published arguing for and against a distinct functional role for maternal Cdx2. This commentary aims to discuss the approaches, results and interpretations of both studies in an attempt to resolve the apparent conflict and to constructively advance collective understanding.


Subject(s)
Embryonic Development/genetics , Homeodomain Proteins/physiology , RNA, Messenger, Stored/genetics , Transcription Factors/physiology , Animals , CDX2 Transcription Factor , Cell Lineage/genetics , Embryonic Development/physiology , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , RNA, Messenger , RNA, Messenger, Stored/physiology
7.
Development ; 137(10): 1709-19, 2010 May.
Article in English | MEDLINE | ID: mdl-20430746

ABSTRACT

The metameric organization of the insect body plan is initiated with the activation of gap genes, a set of transcription-factor-encoding genes that are zygotically expressed in broad and partially overlapping domains along the anteroposterior (AP) axis of the early embryo. The spatial pattern of gap gene expression domains along the AP axis is generally conserved, but the maternal genes that regulate their expression are not. Building on the comprehensive knowledge of maternal gap gene activation in Drosophila, we used loss- and gain-of-function experiments in the hover fly Episyrphus balteatus (Syrphidae) to address the question of how the maternal regulation of gap genes evolved. We find that, in Episyrphus, a highly diverged bicoid ortholog is solely responsible for the AP polarity of the embryo. Episyrphus bicoid represses anterior zygotic expression of caudal and activates the anterior and central gap genes orthodenticle, hunchback and Krüppel. In bicoid-deficient Episyrphus embryos, nanos is insufficient to generate morphological asymmetry along the AP axis. Furthermore, we find that torso transiently regulates anterior repression of caudal and is required for the activation of orthodenticle, whereas all posterior gap gene domains of knirps, giant, hunchback, tailless and huckebein depend on caudal. We conclude that all maternal coordinate genes have altered their specific functions during the radiation of higher flies (Cyclorrhapha).


Subject(s)
Diptera/genetics , Gene Expression Regulation, Developmental , Genes, Insect , RNA, Messenger, Stored/physiology , Transcription Factors/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Body Patterning/genetics , Cell Polarity/genetics , Diptera/embryology , Embryo, Nonmammalian , Female , Genes, Insect/physiology , Molecular Sequence Data , Phylogeny , Sequence Homology , Transcription Factors/metabolism , Transcriptional Activation
8.
Development ; 137(6): 859-70, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20179092

ABSTRACT

The hiatus between oocyte and embryonic gene transcription dictates a role for stored maternal factors in early mammalian development. Encoded by maternal-effect genes, these factors accumulate during oogenesis and enable the activation of the embryonic genome, the subsequent cleavage stages of embryogenesis and the initial establishment of embryonic cell lineages. Recent studies in mice have yielded new findings on the role of maternally provided proteins and multi-component complexes in preimplantation development. Nevertheless, significant gaps remain in our mechanistic understanding of the networks that regulate early mammalian embryogenesis, which provide an impetus and opportunities for future investigations.


Subject(s)
Embryonic Development/genetics , Embryonic Development/physiology , Maternal-Fetal Exchange/physiology , Mice/embryology , Animals , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Maternal-Fetal Exchange/genetics , Mice/genetics , Models, Biological , Oocytes/growth & development , Oocytes/physiology , Pregnancy , Protein Processing, Post-Translational/physiology , RNA, Messenger, Stored/genetics , RNA, Messenger, Stored/metabolism , RNA, Messenger, Stored/physiology
9.
Development ; 137(6): 923-33, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20150281

ABSTRACT

In amphibian and teleost embryos, the dorsal determinants (DDs) are believed to be initially localized to the vegetal pole and then transported to the prospective dorsal side of the embryo along a microtubule array. The DDs are known to activate the canonical Wnt pathway and thereby promote the expression of genes that induce the dorsal organizer. Here, by identifying the locus of the maternal-effect ventralized mutant tokkaebi, we show that Syntabulin, a linker of the kinesin I motor protein, is essential for dorsal determination in zebrafish. We found that syntabulin mRNA is transported to the vegetal pole during oogenesis through the Bucky ball (Buc)-mediated Balbiani body-dependent pathway, which is necessary for establishment of animal-vegetal (AV) oocyte polarity. We demonstrate that Syntabulin is translocated from the vegetal pole in a microtubule-dependent manner. Our findings suggest that Syntabulin regulates the microtubule-dependent transport of the DDs, and provide evidence for the link between AV and dorsoventral axis formation.


Subject(s)
Body Patterning/genetics , Microtubule-Associated Proteins/physiology , Zebrafish Proteins/physiology , Animals , Animals, Genetically Modified , Cell Polarity/genetics , Cells, Cultured , Cloning, Molecular , Embryo, Nonmammalian , Female , Humans , Male , Mice , Mice, Inbred BALB C , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Molecular Motor Proteins/genetics , Molecular Motor Proteins/physiology , Protein Transport , RNA, Messenger, Stored/genetics , RNA, Messenger, Stored/physiology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
10.
BMC Dev Biol ; 8: 97, 2008 Oct 06.
Article in English | MEDLINE | ID: mdl-18837968

ABSTRACT

BACKGROUND: The maternal contribution of transcripts and proteins supplied to the zygote is crucial for the progression from a gametic to an embryonic control of preimplantation development. Here we compared the transcriptional profiles of two types of mouse MII oocytes, one which is developmentally competent (MIISN oocyte), the other that ceases development at the 2-cell stage (MIINSN oocyte), with the aim of identifying genes and gene expression networks whose misregulated expression would contribute to a reduced developmental competence. RESULTS: We report that: 1) the transcription factor Oct-4 is absent in MIINSN oocytes, accounting for 2) the down-regulation of Stella, a maternal-effect factor required for the oocyte-to-embryo transition and of which Oct-4 is a positive regulator; 3) eighteen Oct-4-regulated genes are up-regulated in MIINSN oocytes and are part of gene expression networks implicated in the activation of adverse biochemical pathways such as oxidative phosphorylation, mitochondrial dysfunction and apoptosis. CONCLUSION: The down-regulation of Oct-4 plays a crucial function in a sequence of molecular processes that leads to the developmental arrest of MIINSN oocytes. The use of a model study in which the MII oocyte ceases development consistently at the 2-cell stage has allowed to attribute a role to the maternal Oct-4 that has never been described before. Oct-4 emerges as a key regulator of the molecular events that govern the establishment of the developmental competence of mouse oocytes.


Subject(s)
Embryonic Development/genetics , Octamer Transcription Factor-3/physiology , Oocytes/growth & development , RNA, Messenger, Stored/physiology , Animals , Chromosomal Proteins, Non-Histone , Cleavage Stage, Ovum/metabolism , Cleavage Stage, Ovum/physiology , Cluster Analysis , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Metaphase/genetics , Metaphase/physiology , Mice , Mice, Inbred C57BL , Octamer Transcription Factor-3/genetics , Oligonucleotide Array Sequence Analysis , Oocytes/metabolism , Repressor Proteins/genetics , Repressor Proteins/physiology
11.
Genes Dev ; 22(12): 1607-16, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18559477

ABSTRACT

Parental origin-specific DNA methylation regulates the monoallelic expression of the mammalian imprinted genes. The methylation marks or imprints are established in the parental germline and maintained throughout embryonic development. However, it is unclear how the methylation imprints are maintained through extensive demethylation in cleavage-stage preimplantation embryos. Previous reports suggested that DNA methyltransferase(s) other than Dnmt1 is involved in the maintenance of the imprints during cleavage. Here we demonstrate, by using conditional knockout mice, that the other known DNA methyltransferases Dnmt3a and Dnmt3b are dispensable for the maintenance of the methylation marks at most imprinted loci. We further demonstrate that a lack of both maternal and zygotic Dnmt1 results in complete demethylation of all imprinted loci examined in blastocysts. Consistent with these results we find that zygotic Dnmt1 is expressed in the preimplantation embryo. Thus, contrary to the previous reports, Dnmt1 alone is sufficient to maintain the methylation marks of the imprinted genes.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/physiology , DNA Methylation , Embryonic Development/genetics , Genomic Imprinting , Zygote/metabolism , Animals , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Genomic Imprinting/genetics , Mice , Mice, Transgenic , Models, Biological , Oocytes/metabolism , Pregnancy , RNA, Messenger, Stored/physiology , DNA Methyltransferase 3B
12.
Development ; 134(22): 4023-32, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17965050

ABSTRACT

Our understanding of the maternal factors that initiate early chordate development, and of their direct zygotic targets, is still fragmentary. A molecular cascade is emerging for the mesendoderm, but less is known about the ectoderm, giving rise to epidermis and nervous tissue. Our cis-regulatory analysis surprisingly places the maternal transcription factor Ci-GATAa (GATA4/5/6) at the top of the ectodermal regulatory network in ascidians. Initially distributed throughout the embryo, Ci-GATAa activity is progressively repressed in vegetal territories by accumulating maternal beta-catenin. Once restricted to the animal hemisphere, Ci-GATAa directly activates two types of zygotic ectodermal genes. First, Ci-fog is activated from the 8-cell stage throughout the ectoderm, then Ci-otx is turned on from the 32-cell stage in neural precursors only. Whereas the enhancers of both genes contain critical and interchangeable GATA sites, their distinct patterns of activation stem from the additional presence of two Ets sites in the Ci-otx enhancer. Initially characterized as activating elements in the neural lineages, these Ets sites additionally act as repressors in non-neural lineages, and restrict GATA-mediated activation of Ci-otx. We thus identify a precise combinatorial code of maternal factors responsible for zygotic onset of a chordate ectodermal genetic program.


Subject(s)
Ciona intestinalis/embryology , Ectoderm/embryology , GATA Transcription Factors/physiology , Mothers , Proto-Oncogene Proteins c-ets/physiology , TCF Transcription Factors/physiology , beta Catenin/physiology , Animals , Animals, Genetically Modified , Body Patterning/genetics , Cell Differentiation/genetics , Ciona intestinalis/genetics , Ectoderm/metabolism , Embryo, Nonmammalian , GATA Transcription Factors/genetics , Gene Expression Regulation, Developmental , Models, Biological , Proto-Oncogene Proteins c-ets/genetics , RNA, Messenger, Stored/physiology , TCF Transcription Factors/genetics , Urochordata/embryology , Urochordata/genetics , beta Catenin/genetics
13.
Theriogenology ; 68 Suppl 1: S147-55, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17583784

ABSTRACT

Recently it has been demonstrated that, along with sperm, some of its RNA can be introduced into the oocyte during fertilization, which stays stable until the activation of the embryonic genome. Originally it was thought that RNA present in semen relates to contamination from somatic cells and/or immature sperm both containing substantially higher amounts of RNA than the fertilizing sperm. However, RNA is still found after stringent washing through density gradients resulting in a sperm fraction that is translational silenced and devoid of cytosolic rRNA and thus of potential RNA contamination-which is not transferable to the oocyte. Sperm only delivers a relatively small amount of paternal RNA (5-10 fg) into the fertilized oocyte when compared to the amount of maternal RNA (approximately 1 ng). Pooled human sperm contains about 5000 different mRNA sequences of which half are common between ejaculates. Besides mRNA sperm also contains small sperm RNA molecules that might interfere in gene expression (iRNA). In human sperm already more than 68 putative iRNAs have been identified and 15 of them may specifically inhibit genes that are only active during early embryonic development. The composition and quantity of sperm RNA is considered to be a valuable diagnostic tool for male fertility. However, only a subpopulation of the purified mature sperm fraction (with a yet unknown composition and quantity of RNA) will appropriately respond to capacitation media to become competent to fertilize the oocyte. In this review the origin and function of sperm borne RNA transferred into the oocyte is discussed along with their putative role in early embryogenesis, which still needs to be experimentally proven.


Subject(s)
Embryonic Development/physiology , RNA, Messenger, Stored/physiology , Spermatozoa/metabolism , Animals , Embryonic Development/genetics , Gene Transfer Techniques , Genome , Haploidy , Male , RNA Interference/physiology , Sperm Maturation/genetics , Time Factors
14.
Theriogenology ; 68 Suppl 1: S77-83, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17524469

ABSTRACT

Messenger RNA (mRNA) expression techniques have become a powerful tool to analyze the relative abundance of transcripts related to oocyte and/or embryo quality. Numerous efforts to identify candidate genes for the developmental competence of bovine oocytes and embryos have been made employing different strategies. The preimplantation bovine embryo is initially under the control of maternal genomic information that is accumulated during oogenesis. Soon, the genetic program of development becomes dependent upon new transcripts derived from activation of the embryonic genome. The early steps in development including maturation, fertilization, timing of first cleavage, activation of the embryonic genome, compaction, and blastocyst formation can be affected by the culture media and conditions as well as the production procedure itself. These perturbations can possibly result in a dramatic decrease of the quality of the resulting blastocysts, and may even affect the viability of offspring born after transfer.


Subject(s)
Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Oocytes/metabolism , RNA, Messenger, Stored/physiology , Animals , Biomarkers/analysis , Blastocyst/metabolism , Cattle , Cell Survival , Gene Expression Profiling , Genes, Developmental , RNA, Messenger, Stored/metabolism
15.
Genes Dev ; 20(13): 1744-54, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16818606

ABSTRACT

Zygotic genome activation (ZGA) is a nuclear reprogramming event that transforms the genome from transcriptional quiescence at fertilization to robust transcriptional activity shortly thereafter. The ensuing gene expression profile in the cleavage-stage embryo establishes totipotency and is required for further development. Although little is known about the molecular basis of ZGA, oocyte-derived mRNAs and proteins that alter chromatin structure are likely crucial. To test this hypothesis, we generated a maternal-effect mutation of Brg1, which encodes a catalytic subunit of SWI/SNF-related complexes, utilizing Cre-loxP gene targeting. In conditional-mutant females, BRG1-depleted oocytes completed meiosis and were fertilized. However, embryos conceived from BRG1-depleted eggs exhibited a ZGA phenotype including two-cell arrest and reduced transcription for approximately 30% of expressed genes. Genes involved in transcription, RNA processing, and cell cycle regulation were particularly affected. The early embryonic arrest is not a consequence of a defective oocyte because depleting maternal BRG1 after oocyte development is complete by RNA interference (RNAi) also resulted in two-cell arrest. To our knowledge, Brg1 is the first gene required for ZGA in mammals. Depletion of maternal BRG1 did not affect global levels of histone acetylation, whereas dimethyl-H3K4 levels were reduced. These data provide a framework for understanding the mechanism of ZGA.


Subject(s)
Genome , Nuclear Proteins/physiology , Transcription Factors/physiology , Zygote/physiology , Acetylation , Animals , Cell Cycle/physiology , Chromatin Assembly and Disassembly , DNA Helicases , Embryo, Mammalian/physiology , Female , Fertility , Gene Expression Regulation, Developmental , Histones/metabolism , In Vitro Techniques , Mice , Mice, Transgenic , Mutation , Nuclear Proteins/genetics , Oocytes/physiology , RNA Interference , RNA, Messenger, Stored/genetics , RNA, Messenger, Stored/physiology , Recombination, Genetic , Transcription Factors/genetics , Transcription, Genetic
16.
Dev Genes Evol ; 216(2): 69-80, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16369806

ABSTRACT

Maternal factors, such as a muscle determinant macho-1 mRNA that is localized to the posterior-vegetal cortex (PVC) of fertilized ascidian eggs, are crucial for embryonic axis formation and cell fate specification. Maternal mRNAs that show an identical posterior localization pattern to that of macho-1 in eggs and embryos are called Type I postplasmic/PEM mRNAs. We investigated the functions of five of the nine Type I mRNAs so far known in Halocynthia roretzi: Hr-Wnt-5, Hr-GLUT, Hr-PEM3, Hr-PEN1, and Hr-PEN2. Suppression of their functions with specific antisense morpholino oligonucleotides (MOs) had effects on the formation of various tissues: Hr-Wnt-5 on notochord, muscle, and mesenchyme, although zygotic function of Hr-Wnt-5 is responsible for notochord formation; Hr-GLUT on notochord, mesenchyme, and endoderm; and Hr-PEN2 on muscle, mesenchyme, and endoderm. On the other hand, Hr-PEM3 and Hr-PEN1 MOs seemed to have no effect. We conclude that the functions of at least some localized maternal Type I postplasmic/PEM mRNAs are necessary for early embryonic patterning in ascidians.


Subject(s)
Body Patterning/genetics , RNA, Messenger, Stored/analysis , RNA, Messenger, Stored/physiology , Urochordata/chemistry , Urochordata/embryology , Animals , Egg Proteins/antagonists & inhibitors , Egg Proteins/genetics , Egg Proteins/physiology , Embryo, Nonmammalian/chemistry , Embryo, Nonmammalian/metabolism , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/physiology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/physiology , Oligoribonucleotides, Antisense/genetics , Oligoribonucleotides, Antisense/pharmacology , RNA, Messenger, Stored/antagonists & inhibitors , Urochordata/genetics , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/genetics , Wnt Proteins/physiology
17.
Curr Biol ; 15(24): R1002-5, 2005 Dec 20.
Article in English | MEDLINE | ID: mdl-16360671

ABSTRACT

Gene products provided by the mother to the embryo determine the body axes in most animals. A recent study in zebrafish proposes that the TGFss signal Squint is one such factor.


Subject(s)
Body Patterning/physiology , Gene Expression Regulation, Developmental , RNA, Messenger, Stored/physiology , Signal Transduction/physiology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Blastomeres/metabolism , Cell Polarity/physiology , Nodal Signaling Ligands , RNA, Messenger, Stored/genetics , Zebrafish Proteins/physiology
18.
Development ; 132(21): 4845-55, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16207750

ABSTRACT

During the development of Xenopus laevis, maternal mRNAs and proteins stored in the egg direct early patterning events such as the specification of the dorsoventral axis and primary germ layers. In an expression screen to identify maternal factors important for early development, we isolated a truncated cDNA for maternal Zic2 (tZic2), encoding a zinc-finger transcription factor. The predicted tZic2 protein lacked the N-terminal region, but retained the zinc-finger domain. When expressed in embryos, tZic2 inhibited head and axial development, and blocked the ability of full-length Zic2 to induce neural crest genes. Depletion of maternal Zic2 from oocytes, using antisense oligonucleotides, caused exogastrulation, anterior truncations and axial defects. We show that loss of maternal Zic2 results in persistent and increased expression of Xenopus nodal-related (Xnr) genes, except for Xnr4, and overall increased Nodal signaling. Injection of a Nodal antagonist, Cerberus-short, reduced the severity of head and axial defects in Zic2-depleted embryos. Depletion of Zic2 could not restore Xnr expression to embryos additionally depleted of VegT, a T-domain transcription factor and an activator of Xnr gene transcription. Taken together, our results suggest a role for maternal Zic2 in the suppression of Xnr genes in early development. ZIC2 is mutated in human holoprosencephaly (HPE), a severe defect in brain hemisphere separation, and these results strengthen the suggestion that increased Nodal-related activity is a cause of HPE.


Subject(s)
Neural Crest , RNA, Messenger, Stored/physiology , Transcription Factors/genetics , Transforming Growth Factor beta/genetics , Xenopus Proteins/genetics , Xenopus laevis/genetics , Animals , Body Patterning , Down-Regulation , Embryonic Development/genetics , Head/abnormalities , Mutation , Oocytes/growth & development , Transcription Factors/deficiency , Transcription Factors/physiology , Xenopus Proteins/deficiency , Xenopus Proteins/physiology , Xenopus laevis/embryology , Xenopus laevis/growth & development
19.
Exp Cell Res ; 307(2): 354-66, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15894314

ABSTRACT

We report on the essential Drosophila mRpL55 gene conserved exclusively in metazoans. Null mRpL55 mutants did not grow after hatching, moved slowly and died as first instar larvae. MrpL55 is similar to mammalian MRPL55, a protein that, in a large-scale mass spectrometry study, has been found as a mitoribosome-specific large subunit protein. We showed that MrpL55 was localised to the mitochondrion in S2 cells and tissues and was enriched in cells with a higher protein synthesis activity. The MrpL55 protein contains a KOW-like motif present in proteins with a role in transcriptional anti-termination and regulation of translation. Modulation of mRpL55 expression level is critical for development. Somatic clonal analysis showed that MrpL55 was not required in larval eye imaginal discs but required in pupal discs apparently during the second mitotic wave. Therefore, our results showed that the MrpL55 protein acts dynamically in the cell during development. We propose that MrpL55 is involved in Drosophila mitochondrial biogenesis and G2/M phase cell cycle progression.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/growth & development , RNA-Binding Proteins/physiology , Amino Acid Motifs/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Cell Line , Cloning, Molecular , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/physiology , Eye/cytology , Eye/growth & development , Female , Gene Deletion , Gene Expression/genetics , Gene Expression Regulation, Developmental , Humans , Immunohistochemistry , Larva/genetics , Larva/growth & development , Mitochondria/chemistry , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/physiology , Molecular Sequence Data , Mutation , Nematoda/genetics , Oogenesis/physiology , Phenotype , Protein Structure, Secondary , RNA, Messenger, Stored/analysis , RNA, Messenger, Stored/physiology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Recombination, Genetic/genetics , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomal Proteins/physiology , Salivary Glands/cytology , Salivary Glands/metabolism , Sequence Homology, Amino Acid , Subcellular Fractions/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...