Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Dev Dyn ; 252(4): 483-494, 2023 04.
Article in English | MEDLINE | ID: mdl-36495293

ABSTRACT

BACKGROUND: Frem1 has been linked to human face shape variation, dysmorphology, and malformation, but little is known about its regulation and biological role in facial development. RESULTS: During midfacial morphogenesis in mice, we observed Frem1 expression in the embryonic growth centers that form the median upper lip, nose, and palate. Expansive spatial gradients of Frem1 expression in the cranial neural crest cell (cNCC) mesenchyme of these tissues suggested transcriptional regulation by a secreted morphogen. Accordingly, Frem1 expression paralleled that of the conserved Sonic Hedgehog (Shh) target gene Gli1 in the cNCC mesenchyme. Suggesting direct transcriptional regulation by Shh signaling, we found that Frem1 expression is induced by SHH ligand stimulation or downstream pathway activation in cNCCs and observed GLI transcription factor binding at the Frem1 transcriptional start site during midfacial morphogenesis. Finally, we found that FREM1 is sufficient to induce cNCC proliferation in a concentration-dependent manner and that Shh pathway antagonism reduces Frem1 expression during pathogenesis of midfacial hypoplasia. CONCLUSIONS: By demonstrating that the Shh signaling pathway regulates Frem1 expression in cNCCs, these findings provide novel insight into the mechanisms underlying variation in midfacial morphogenesis.


Subject(s)
Hedgehog Proteins , Neural Crest , Mice , Animals , Humans , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Morphogenesis/genetics , Signal Transduction/physiology , Mesoderm/metabolism , Extracellular Matrix Proteins/metabolism
2.
Dev Biol ; 492: 133-138, 2022 12.
Article in English | MEDLINE | ID: mdl-36270327

ABSTRACT

BioID is a proximity labeling strategy whose goal is to identify in vivo protein-protein interactions. The central components of this strategy are modified biotin ligase enzymes that promiscuously add biotin groups to proteins in close proximity. The transferred biotin group provides a powerful tag for purification and thus identification of interacting proteins. While a variety of modified biotin ligases were created for BioID, the original enzymes were inefficient, required long incubation times, and high intracellular biotin concentrations for protein labeling. These limitations hinder the application of BioID in contexts such as developing embryos where processes such as cell division and cell fate decisions occur rapidly. Recently, a new biotin ligase called TurboID was developed that addressed many of the deficiencies of previous enzymes. In this paper we compare TurboID to the BioID2 biotin ligase in developing Xenopus embryos. We find that the TurboID enzyme has several advantages over the BioID2 enzyme. TurboID labels proteins efficiently without the addition of additional biotin and occurs at a range of temperatures compatible with the culturing of Xenopus embryos. Biotinylation events occurred rapidly and were limited by TurboID expression and not its activity. Thus, TurboID is an efficient tool for BioID applications in Xenopus embryos and its use should facilitate the identification of interacting proteins in specific networks and complexes during Xenopus development.


Subject(s)
Biotin , Ligases , Animals , Xenopus laevis , Biotinylation
3.
Development ; 146(10)2019 05 15.
Article in English | MEDLINE | ID: mdl-31023875

ABSTRACT

Bicaudal-C (Bicc1) is a conserved RNA-binding protein that represses the translation of selected mRNAs to control development. In Xenopus embryos, Bicc1 binds and represses specific maternal mRNAs to control anterior-posterior cell fates. However, it is not known how Bicc1 binds its RNA targets or how binding affects Bicc1-dependent embryogenesis. Focusing on the KH domains, we analyzed Bicc1 mutants for their ability to bind RNA substrates in vivo and in vitro Analyses of these Bicc1 mutants demonstrated that a single KH domain, KH2, was crucial for RNA binding in vivo and in vitro, while the KH1 and KH3 domains contributed minimally. The Bicc1 mutants were also assayed for their ability to repress translation, and results mirrored the RNA-binding data, with KH2 being the only domain essential for repression. Finally, maternal knockdown and rescue experiments indicated that the KH domains were essential for the regulation of embryogenesis by Bicc1. These data advance our understanding of how Bicc1 selects target mRNAs and provide the first direct evidence that the RNA binding functions of Bicc1 are essential for both Bicc1-dependent translational repression and maternal vertebrate development.


Subject(s)
RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Xenopus Proteins/metabolism , 3' Untranslated Regions/genetics , 3' Untranslated Regions/physiology , Animals , Embryonic Development/genetics , Embryonic Development/physiology , Female , Immunoblotting , Immunoprecipitation , Protein Binding , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Xenopus Proteins/genetics , Xenopus laevis
4.
Development ; 143(5): 864-71, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26811381

ABSTRACT

Vertebrate Bicaudal-C (Bicc1) has important biological roles in the formation and homeostasis of multiple organs, but direct experiments to address the role of maternal Bicc1 in early vertebrate embryogenesis have not been reported. Here, we use antisense phosphorothioate-modified oligonucleotides and the host-transfer technique to eliminate specifically maternal stores of both bicc1 mRNA and Bicc1 protein from Xenopus laevis eggs. Fertilization of these Bicc1-depleted eggs produced embryos with an excess of dorsal-anterior structures and overexpressed organizer-specific genes, indicating that maternal Bicc1 is crucial for normal embryonic patterning of the vertebrate embryo. Bicc1 is an RNA-binding protein with robust translational repression function. Here, we show that the maternal mRNA encoding the cell-fate regulatory protein Wnt11b is a direct target of Bicc1-mediated repression. It is well established that the Wnt signaling pathway is crucial to vertebrate embryogenesis. Thus, the work presented here links the molecular function of Bicc1 in mRNA target-specific translation repression to its biological role in the maternally controlled stages of vertebrate embryogenesis.


Subject(s)
Cell Lineage , Gene Expression Regulation, Developmental , RNA-Binding Proteins/metabolism , Wnt Proteins/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Animals , Embryonic Development , Female , MicroRNAs/metabolism , Mutation , Oligonucleotides, Antisense/genetics , Oocytes/metabolism , Phenotype , RNA, Messenger/metabolism , RNA, Messenger, Stored/genetics , Signal Transduction , Transcription, Genetic
5.
Genes Dev ; 25(11): 1121-31, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21576259

ABSTRACT

We show that, in Xenopus laevis oocytes and early embryos, double-stranded exogenous siRNAs cannot function as microRNA (miRNA) mimics in either deadenylation or guided mRNA cleavage (RNAi). Instead, siRNAs saturate and inactivate maternal Argonaute (Ago) proteins, which are present in low amounts but are needed for Dicer processing of pre-miRNAs at the midblastula transition (MBT). Consequently, siRNAs impair accumulation of newly made miRNAs, such as the abundant embryonic pre-miR-427, but inhibition dissipates upon synthesis of zygotic Ago proteins after MBT. These effects of siRNAs, which are independent of sequence, result in morphological defects at later stages of development. The expression of any of several exogenous human Ago proteins, including catalytically inactive Ago2 (Ago2mut), can overcome the siRNA-mediated inhibition of miR-427 biogenesis and function. However, expression of wild-type, catalytically active hAgo2 is required to elicit RNAi in both early embryos and oocytes using either siRNA or endogenous miRNAs as guides. The lack of endogenous Ago2 endonuclease activity explains why these cells normally are unable to support RNAi. Expression of catalytically active exogenous Ago2, which appears not to perturb normal Xenopus embryonic development, can now be exploited for RNAi in this vertebrate model organism.


Subject(s)
Eukaryotic Initiation Factors/metabolism , Gene Expression Regulation, Developmental , MicroRNAs/biosynthesis , RNA Interference , Xenopus laevis/embryology , Animals , Argonaute Proteins , Embryo, Nonmammalian , Eukaryotic Initiation Factor-2/metabolism , Humans , Oocytes/metabolism , RNA Stability , RNA, Small Interfering , Ribonuclease III/antagonists & inhibitors , Ribonuclease III/metabolism , Xenopus laevis/genetics
6.
Adv Exp Med Biol ; 953: 49-82, 2017.
Article in English | MEDLINE | ID: mdl-27975270

ABSTRACT

The selective translation of maternal mRNAs encoding cell-fate determinants drives the earliest decisions of embryogenesis that establish the vertebrate body plan. This chapter will discuss studies in Xenopus laevis that provide insights into mechanisms underlying this translational control. Xenopus has been a powerful model organism for many discoveries relevant to the translational control of maternal mRNAs because of the large size of its oocytes and eggs that allow for microinjection of molecules and the relative ease of manipulating the oocyte to egg transition (maturation) and fertilization in culture. Consequently, many key studies have focused on the expression of maternal mRNAs during the oocyte to egg transition (the meiotic cell cycle) and the rapid cell divisions immediately following fertilization. This research has made seminal contributions to our understanding of translational regulatory mechanisms, but while some of the mRNAs under consideration at these stages encode cell-fate determinants, many encode cell cycle regulatory proteins that drive these early cell cycles. In contrast, while maternal mRNAs encoding key developmental (i.e., cell-fate) regulators that function after the first cleavage stages may exploit aspects of these foundational mechanisms, studies reveal that these mRNAs must also rely on distinct and, as of yet, incompletely understood mechanisms. These findings are logical because the functions of such developmental regulatory proteins have requirements distinct from cell cycle regulators, including becoming relevant only after fertilization and then only in specific cells of the embryo. Indeed, key maternal cell-fate determinants must be made available in exquisitely precise amounts (usually low), only at specific times and in specific cells during embryogenesis. To provide an appreciation for the regulation of maternal cell-fate determinant expression, an overview of the maternal phase of Xenopus embryogenesis will be presented. This section will be followed by a review of translational mechanisms operating in oocytes, eggs, and early cleavage-stage embryos and conclude with a discussion of how the regulation of key maternal cell-fate determinants at the level of translation functions in Xenopus embryogenesis. A key theme is that the molecular asymmetries critical for forming the body axes are established and further elaborated upon by the selective temporal and spatial regulation of maternal mRNA translation.


Subject(s)
Embryonic Development/genetics , Protein Biosynthesis , RNA, Messenger/biosynthesis , Xenopus laevis/growth & development , Animals , Cell Cycle/genetics , Female , Gene Expression Regulation, Developmental , Oocytes/growth & development , Oocytes/metabolism , RNA, Messenger/genetics , Transcription, Genetic , Xenopus laevis/genetics
7.
J Biol Chem ; 289(11): 7497-504, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24478311

ABSTRACT

Bicaudal-C (Bic-C) RNA binding proteins function as important translational repressors in multiple biological contexts within metazoans. However, their RNA binding sites are unknown. We recently demonstrated that Bic-C functions in spatially regulated translational repression of the xCR1 mRNA during Xenopus development. This repression contributes to normal development by confining the xCR1 protein, a regulator of key signaling pathways, to specific cells of the embryo. In this report, we combined biochemical approaches with in vivo mRNA reporter assays to define the minimal Bic-C target site within the xCR1 mRNA. This 32-nucleotide Bic-C target site is predicted to fold into a stem-loop secondary structure. Mutational analyses provided evidence that this stem-loop structure is important for Bic-C binding. The Bic-C target site was sufficient for Bic-C mediated repression in vivo. Thus, we describe the first RNA binding site for a Bic-C protein. This identification provides an important step toward understanding the mechanisms by which evolutionarily conserved Bic-C proteins control cellular function in metazoans.


Subject(s)
Gene Expression Regulation, Developmental , Protein Biosynthesis , RNA-Binding Proteins/metabolism , RNA/chemistry , Xenopus Proteins/metabolism , 3' Untranslated Regions , Animals , Base Sequence , Binding Sites , Luciferases/metabolism , Molecular Sequence Data , Nucleic Acid Conformation , Protein Binding , RNA, Messenger/metabolism , Ribonucleases/metabolism , Xenopus laevis
8.
RNA ; 19(11): 1575-82, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24062572

ABSTRACT

The Xenopus Cripto-1 protein is confined to the cells of the animal hemisphere during early embryogenesis where it regulates the formation of anterior structures. Cripto-1 protein accumulates only in animal cells because cripto-1 mRNA in cells of the vegetal hemisphere is translationally repressed. Here, we show that the RNA binding protein, Bicaudal-C (Bic-C), functioned directly in this vegetal cell-specific repression. While Bic-C protein is normally confined to vegetal cells, ectopic expression of Bic-C in animal cells repressed a cripto-1 mRNA reporter and associated with endogenous cripto-1 mRNA. Repression by Bic-C required its N-terminal domain, comprised of multiple KH motifs, for specific binding to relevant control elements within the cripto-1 mRNA and a functionally separable C-terminal translation repression domain. Bic-C-mediated repression required the 5' CAP and translation initiation factors, but not a poly(A) tail or the conserved SAM domain within Bic-C. Bic-C-directed immunoprecipitation followed by deep sequencing of associated mRNAs identified multiple Bic-C-regulated mRNA targets, including cripto-1 mRNA, providing new insights and tools for understanding the role of Bic-C in vertebrate development.


Subject(s)
GPI-Linked Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Xenopus Proteins/biosynthesis , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis/genetics , 3' Untranslated Regions , Animals , Base Sequence , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins , Protein Biosynthesis , Protein Structure, Tertiary , RNA, Messenger, Stored/genetics , RNA, Messenger, Stored/metabolism , RNA-Binding Proteins/chemistry , Sequence Analysis, RNA , Xenopus Proteins/chemistry , Xenopus laevis/metabolism
9.
Proc Natl Acad Sci U S A ; 108(19): 7844-9, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21518916

ABSTRACT

Translational control of many mRNAs in developing metazoan embryos is achieved by alterations in their poly(A) tail length. A family of cytoplasmic poly(A)-binding proteins (PABPs) bind the poly(A) tail and can regulate mRNA translation and stability. However, despite the extensive biochemical characterization of one family member (PABP1), surprisingly little is known about their in vivo roles or functional relatedness. Because no information is available in vertebrates, we address their biological roles, establishing that each of the cytoplasmic PABPs conserved in Xenopus laevis [PABP1, embryonic PABP (ePABP), and PABP4] is essential for normal development. Morpholino-mediated knockdown of PABP1 or ePABP causes both anterior and posterior phenotypes and embryonic lethality. In contrast, depletion of PABP4 results mainly in anterior defects and lethality at later stages. Unexpectedly, cross-rescue experiments reveal that neither ePABP nor PABP4 can fully rescue PABP1 depletion, establishing that PABPs have distinct functions. Comparative analysis of the uncharacterized PABP4 with PABP1 and ePABP shows that it shares a mechanistically conserved core role in promoting global translation. Consistent with this analysis, each morphant displays protein synthesis defects, suggesting that their roles in mRNA-specific translational regulation and/or mRNA decay, rather than global translation, underlie the functional differences between PABPs. Domain-swap experiments reveal that the basis of the functional specificity is complex, involving multiple domains of PABPs, and is conferred, at least in part, by protein-protein interactions.


Subject(s)
Poly(A)-Binding Proteins/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Xenopus laevis/metabolism , Animals , Base Sequence , Female , Male , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/genetics , Poly(A)-Binding Protein I/antagonists & inhibitors , Poly(A)-Binding Protein I/genetics , Poly(A)-Binding Protein I/metabolism , Poly(A)-Binding Proteins/antagonists & inhibitors , Poly(A)-Binding Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vertebrates/embryology , Vertebrates/genetics , Vertebrates/metabolism , Xenopus Proteins/antagonists & inhibitors , Xenopus Proteins/genetics , Xenopus laevis/genetics
10.
Dev Biol ; 368(2): 231-41, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22627292

ABSTRACT

Signaling inputs from multiple pathways are essential for the establishment of distinct cell and tissue types in the embryo. Therefore, multiple signals must be integrated to activate gene expression and confer cell fate, but little is known about how this occurs at the level of target gene promoters. During early embryogenesis, Wnt and Nodal signals are required for formation of the Spemann organizer, which is essential for germ layer patterning and axis formation. Signaling by both Wnt and Nodal pathways is required for the expression of multiple organizer genes, suggesting that integration of these signals is required for organizer formation. Here, we demonstrate transcriptional cooperation between the Wnt and Nodal pathways in the activation of the organizer genes Goosecoid (Gsc), Cerberus (Cer), and Chordin (Chd). Combined Wnt and Nodal signaling synergistically activates transcription of these organizer genes. Effectors of both pathways occupy the Gsc, Cer and Chd promoters and effector occupancy is enhanced with active Wnt and Nodal signaling. This suggests that, at organizer gene promoters, a stable transcriptional complex containing effectors of both pathways forms in response to combined Wnt and Nodal signaling. Consistent with this idea, the histone acetyltransferase p300 is recruited to organizer promoters in a Wnt and Nodal effector-dependent manner. Taken together, these results offer a mechanism for spatial and temporal restriction of organizer gene transcription by the integration of two major signaling pathways, thus establishing the Spemann organizer domain.


Subject(s)
Nodal Protein/metabolism , Organizers, Embryonic/metabolism , Wnt Proteins/metabolism , Xenopus Proteins/metabolism , Animals , Body Patterning/genetics , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Goosecoid Protein/genetics , Immunohistochemistry , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/genetics , Nodal Protein/genetics , Organizers, Embryonic/embryology , Promoter Regions, Genetic/genetics , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Time Factors , Transcriptional Activation , Wnt Proteins/genetics , Xenopus Proteins/genetics , Xenopus laevis/embryology , Xenopus laevis/metabolism , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
11.
Biochem J ; 445(1): 93-100, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22497250

ABSTRACT

Oocyte maturation and early embryonic development require the cytoplasmic polyadenylation and concomitant translational activation of stored maternal mRNAs. ePAB [embryonic poly(A)-binding protein, also known as ePABP and PABPc1-like] is a multifunctional post-transcriptional regulator that binds to poly(A) tails. In the present study we find that ePAB is a dynamically modified phosphoprotein in Xenopus laevis oocytes and show by mutation that phosphorylation at a four residue cluster is required for oocyte maturation. We further demonstrate that these phosphorylations are critical for cytoplasmic polyadenylation, but not for ePAB's inherent ability to promote translation. Our results provide the first insight into the role of post-translational modifications in regulating PABP protein activity in vivo.


Subject(s)
Gene Expression Regulation, Developmental , Oocytes/cytology , Poly A/metabolism , Poly(A)-Binding Proteins/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Xenopus laevis/metabolism , Animals , Blotting, Western , Cytoplasm/metabolism , Female , Fluorescent Antibody Technique , Immunoprecipitation , Mutation/genetics , Oocytes/metabolism , Oogenesis/physiology , Phosphorylation , Poly(A)-Binding Proteins/genetics , Polyadenylation , Protein Biosynthesis , Protein Processing, Post-Translational , RNA, Messenger/genetics , Xenopus Proteins/genetics
12.
Front Cell Dev Biol ; 10: 981696, 2022.
Article in English | MEDLINE | ID: mdl-36158189

ABSTRACT

Bicaudal-C (Bicc1) is an evolutionarily conserved RNA binding protein that functions in a regulatory capacity in a variety of contexts. It was originally identified as a genetic locus in Drosophila that when disrupted resulted in radical changes in early development. In the most extreme phenotypes embryos carrying mutations developed with mirror image duplications of posterior structures and it was this striking phenotype that was responsible for the name Bicaudal. These seminal studies established Bicc1 as an important regulator of Drosophila development. What was not anticipated from the early work, but was revealed subsequently in many different organisms was the broad fundamental impact that Bicc1 proteins have on developmental biology; from regulating cell fates in vertebrate embryos to defects associated with several human disease states. In the following review we present a perspective of Bicc1 focusing primarily on the molecular aspects of its RNA metabolism functions in vertebrate embryos.

13.
RNA ; 15(12): 2351-63, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19854872

ABSTRACT

We show that microRNA-427 (miR-427) mediates the rapid deadenylation of maternal mRNAs after the midblastula transition (MBT) of Xenopus laevis embryogenesis. By MBT, the stage when the embryonic cell cycle is remodeled and zygotic transcription of mRNAs is initiated, each embryo has accumulated approximately 10(9) molecules of miR-427 processed from multimeric pri-miR-427 transcripts synthesized after fertilization. We demonstrate that the maternal mRNAs for cyclins A1 and B2 each contain a single miR-427 target sequence, spanning less than 30 nucleotides, that is both necessary and sufficient for deadenylation, and that inactivation of miR-427 leads to stabilization of the mRNAs. Although this deadenylation normally takes place after MBT, exogenous miRNAs produced prematurely in vivo can promote deadenylation prior to MBT, indicating that turnover of the maternal mRNAs is limited by the amount of accumulated miR-427. Injected transcripts comprised solely of the cyclin mRNA 3' untranslated regions or bearing a 5' ApppG cap undergo deadenylation, showing that translation of the targeted RNA is not required. miR-427 is not unique in promoting deadenylation, as an unrelated miRNA, let-7, can substitute for miR-427 if the reporter RNA contains an appropriate let-7 target site. We propose that miR-427, like the orthologous miR-430 of zebrafish, functions to down-regulate expression of maternal mRNAs early in development.


Subject(s)
Adenine/metabolism , MicroRNAs/metabolism , RNA, Messenger/metabolism , Xenopus laevis/metabolism , 3' Untranslated Regions , Animals , Base Sequence , Cyclin B2/genetics , Cyclin B2/metabolism , Gene Expression Regulation, Developmental , MicroRNAs/genetics , Molecular Sequence Data , Protein Biosynthesis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Xenopus laevis/genetics
14.
Proc Natl Acad Sci U S A ; 105(40): 15317-22, 2008 Oct 07.
Article in English | MEDLINE | ID: mdl-18824697

ABSTRACT

We identified a functional domain (XlePABP2-TRP) of Xenopus laevis embryonic type II poly(A)-binding protein (XlePABP2). The NMR structure of XlePABP2-TRP revealed that the protein is a homodimer formed by the antiparallel association of beta-strands from the single RNA recognition motif (RRM) domain of each subunit. In each subunit of the homodimer, the canonical RNA recognition site is occluded by a polyproline motif. Upon poly(A) binding, XlePABP2-TRP undergoes a dimer-monomer transition that removes the polyproline motif from the RNA recognition site and allows it to be replaced by the adenosine nucleotides of poly(A). Our results provide high-resolution structural information concerning type II PABPs and an example of a single RRM domain protein that transitions from a homodimer to a monomer upon RNA binding. These findings advance our understanding of RRM domain regulation, poly(A) recognition, and are relevant to understanding how type II PABPs function in mRNA processing and human disease.


Subject(s)
Poly(A)-Binding Protein II/chemistry , Poly(A)-Binding Protein II/metabolism , RNA/metabolism , Xenopus Proteins/chemistry , Xenopus Proteins/metabolism , Animals , Binding Sites , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Poly A/metabolism , Protein Conformation , Protein Structure, Tertiary , RNA/chemistry , Structure-Activity Relationship , Xenopus/metabolism
15.
Nat Commun ; 12(1): 5482, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34531379

ABSTRACT

Rotating cilia at the vertebrate left-right organizer (LRO) generate an asymmetric leftward flow, which is sensed by cells at the left LRO margin. Ciliary activity of the calcium channel Pkd2 is crucial for flow sensing. How this flow signal is further processed and relayed to the laterality-determining Nodal cascade in the left lateral plate mesoderm (LPM) is largely unknown. We previously showed that flow down-regulates mRNA expression of the Nodal inhibitor Dand5 in left sensory cells. De-repression of the co-expressed Nodal, complexed with the TGFß growth factor Gdf3, drives LPM Nodal cascade induction. Here, we show that post-transcriptional repression of dand5 is a central process in symmetry breaking of Xenopus, zebrafish and mouse. The RNA binding protein Bicc1 was identified as a post-transcriptional regulator of dand5 and gdf3 via their 3'-UTRs. Two distinct Bicc1 functions on dand5 mRNA were observed at pre- and post-flow stages, affecting mRNA stability or flow induced translational inhibition, respectively. To repress dand5, Bicc1 co-operates with Dicer1, placing both proteins in the process of flow sensing. Intriguingly, Bicc1 mediated translational repression of a dand5 3'-UTR mRNA reporter was responsive to pkd2, suggesting that a flow induced Pkd2 signal triggers Bicc1 mediated dand5 inhibition during symmetry breakage.


Subject(s)
Body Patterning/genetics , Gene Expression Regulation, Developmental , Intercellular Signaling Peptides and Proteins/genetics , RNA-Binding Proteins/genetics , Ribonuclease III/genetics , Xenopus laevis/genetics , Zebrafish/genetics , 3' Untranslated Regions/genetics , Animals , Embryonic Development/genetics , Mice , RNA Stability/genetics , Xenopus laevis/embryology , Zebrafish/embryology
16.
BMC Dev Biol ; 9: 7, 2009 Jan 28.
Article in English | MEDLINE | ID: mdl-19175933

ABSTRACT

BACKGROUND: Vertebrate development relies on the regulated translation of stored maternal mRNAs, but how these regulatory mechanisms may have evolved to control translational efficiency of individual mRNAs is poorly understood. We compared the translational regulation and polyadenylation of the cyclin B1 mRNA during zebrafish and Xenopus oocyte maturation. Polyadenylation and translational activation of cyclin B1 mRNA is well characterized during Xenopus oocyte maturation. Specifically, Xenopus cyclin B1 mRNA is polyadenylated and translationally activated during oocyte maturation by proteins that recognize the conserved AAUAAA hexanucleotide and U-rich Cytoplasmic Polyadenylation Elements (CPEs) within cyclin B1 mRNA's 3'UnTranslated Region (3'UTR). RESULTS: The zebrafish cyclin B1 mRNA was polyadenylated during zebrafish oocyte maturation. Furthermore, the zebrafish cyclin B1 mRNA's 3'UTR was sufficient to stimulate translation of a reporter mRNA during zebrafish oocyte maturation. This stimulation required both AAUAAA and U-rich CPE-like sequences. However, in contrast to AAUAAA, the positions and sequences of the functionally defined CPEs were poorly conserved between Xenopus and zebrafish cyclin B1 mRNA 3'UTRs. To determine whether these differences were relevant to translation efficiency, we analyzed the translational activity of reporter mRNAs containing either the zebrafish or Xenopus cyclin B1 mRNA 3'UTRs during both zebrafish and Xenopus oocyte maturation. The zebrafish cyclin B1 3'UTR was quantitatively less effective at stimulating polyadenylation and translation compared to the Xenopus cyclin B1 3'UTR during both zebrafish and Xenopus oocyte maturation. CONCLUSION: Although the factors that regulate translation of maternal mRNAs are highly conserved, the target sequences and overall sequence architecture within the 3'UTR of the cyclin B1 mRNA have diverged to affect translational efficiency, perhaps to optimize levels of cyclin B1 protein required by these different species during their earliest embryonic cell divisions.


Subject(s)
Cyclin B/genetics , Gene Expression Regulation, Developmental , Oocytes/metabolism , RNA, Messenger/genetics , 3' Untranslated Regions/genetics , Animals , Cyclin B1 , Female , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Polyadenylation/genetics , Polyadenylation/physiology , Protein Biosynthesis/genetics , Protein Biosynthesis/physiology , Xenopus , Zebrafish
17.
Methods Mol Biol ; 1920: 33-39, 2019.
Article in English | MEDLINE | ID: mdl-30737684

ABSTRACT

The earliest steps of animal development depend upon posttranscriptional events that drive the embryonic cell cycle and guide cell fate decisions. The analysis of post-transcriptional regulatory events has relied upon the use of chimeric reporter mRNAs that encode firefly luciferase fused to potential regulatory sequences. A new and more sensitive luciferase developed recently called NanoLuc has the potential to improve reporter studies and provide new insights into the regulation of embryonic processes. Here I describe how to create and analyze reporter mRNAs encoding NanoLuc luciferase using extracts from microinjected Xenopus embryos.


Subject(s)
Gene Expression , Genes, Reporter , Luciferases/genetics , Luminescent Measurements , Microinjections , RNA, Messenger/genetics , Xenopus laevis/genetics , Animals , Embryo, Nonmammalian , In Vitro Techniques , Luminescent Measurements/methods , Plasmids/genetics , RNA, Messenger/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription, Genetic , Xenopus laevis/metabolism
18.
Cell Signal ; 44: 1-9, 2018 04.
Article in English | MEDLINE | ID: mdl-29284139

ABSTRACT

Sonic Hedgehog (Shh) signaling plays key regulatory roles in embryonic development and postnatal homeostasis and repair. Modulation of the Shh pathway is known to cause malformations and malignancies associated with dysregulated tissue growth. However, our understanding of the molecular mechanisms by which Shh regulates cellular proliferation is incomplete. Here, using mouse embryonic fibroblasts, we demonstrate that the Forkhead box gene Foxd1 is transcriptionally regulated by canonical Shh signaling and required for downstream proliferative activity. We show that Foxd1 deletion abrogates the proliferative response to SHH ligand while FOXD1 overexpression alone is sufficient to induce cellular proliferation. The proliferative response to both SHH ligand and FOXD1 overexpression was blocked by pharmacologic inhibition of cyclin-dependent kinase signaling. Time-course experiments revealed that Shh pathway activation of Foxd1 is followed by downregulation of Cdkn1c, which encodes a cyclin-dependent kinase inhibitor. Consistent with a direct transcriptional regulation mechanism, we found that FOXD1 reduces reporter activity of a Fox enhancer sequence in the second intron of Cdkn1c. Supporting the applicability of these findings to specific biological contexts, we show that Shh regulation of Foxd1 and Cdkn1c is recapitulated in cranial neural crest cells and provide evidence that this mechanism is operational during upper lip morphogenesis. These results reveal a novel Shh-Foxd1-Cdkn1c regulatory circuit that drives the mitogenic action of Shh signaling and may have broad implications in development and disease.


Subject(s)
Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Forkhead Transcription Factors/metabolism , Hedgehog Proteins/metabolism , Neural Crest/growth & development , Animals , Cell Proliferation , Cells, Cultured , Down-Regulation , Gene Expression Regulation , Mice , Primary Cell Culture , Signal Transduction
19.
Cell Rep ; 24(9): 2248-2260, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30157421

ABSTRACT

Length-dependent axonopathy of the corticospinal tract causes lower limb spasticity and is characteristic of several neurological disorders, including hereditary spastic paraplegia (HSP) and amyotrophic lateral sclerosis. Mutations in Trk-fused gene (TFG) have been implicated in both diseases, but the pathomechanisms by which these alterations cause neuropathy remain unclear. Here, we biochemically and genetically define the impact of a mutation within the TFG coiled-coil domain, which underlies early-onset forms of HSP. We find that the TFG (p.R106C) mutation alters compaction of TFG ring complexes, which play a critical role in the export of cargoes from the endoplasmic reticulum (ER). Using CRISPR-mediated genome editing, we engineered human stem cells that express the mutant form of TFG at endogenous levels and identified specific defects in secretion from the ER and axon fasciculation following neuronal differentiation. Together, our data highlight a key role for TFG-mediated protein transport in the pathogenesis of HSP.


Subject(s)
Axon Fasciculation/genetics , Proteins/genetics , Proteins/metabolism , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism , Axons/metabolism , Axons/pathology , Base Sequence , Humans , Mutation , Neurons/metabolism , Neurons/pathology , Protein Transport , Spastic Paraplegia, Hereditary/pathology
20.
J Vis Exp ; (125)2017 07 28.
Article in English | MEDLINE | ID: mdl-28784977

ABSTRACT

Native polyacrylamide gel electrophoresis is a fundamental tool of molecular biology that has been used extensively for the biochemical analysis of RNA-protein interactions. These interactions have been traditionally analyzed with polyacrylamide gels generated between two glass plates and samples electrophoresed vertically. However, polyacrylamide gels cast in trays and electrophoresed horizontally offers several advantages. For example, horizontal gels used to analyze complexes between fluorescent RNA substrates and specific proteins can be imaged multiple times as electrophoresis progresses. This provides the unique opportunity to monitor RNA-protein complexes at several points during the experiment. In addition, horizontal gel electrophoresis makes it possible to analyze many samples in parallel. This can greatly facilitate time course experiments as well as analyzing multiple reactions simultaneously to compare different components and conditions. Here we provide a detailed protocol for generating and using horizontal native gel electrophoresis for analyzing RNA-Protein interactions.


Subject(s)
Electrophoresis, Polyacrylamide Gel , Proteins/metabolism , RNA/metabolism , Animals , Fluorescent Dyes/chemistry , Neoplasm Proteins/genetics , Protein Binding , Proteins/chemistry , RNA/chemistry , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Video Recording , Xenopus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL