Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Oncogene ; 35(50): 6429-6438, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27270425

ABSTRACT

The retinoblastoma tumor suppressor protein (RB) plays a critical role in cell proliferation and differentiation and its inactivation is a frequent underlying factor in tumorigenesis. While the regulation of RB function by phosphorylation is well studied, proteasome-mediated RB protein degradation is emerging as an important regulatory mechanism. Although our understanding of RB turnover is currently limited, there is evidence that the nuclear lamina filament protein Lamin A/C protects RB from proteasomal degradation. Here we show that SUMO1 conjugation of RB and Lamin A/C is modulated by the SUMO protease SENP1 and that sumoylation of both proteins is required for their interaction. Importantly, this SUMO1-dependent complex protects both RB and Lamin A/C from proteasomal turnover.


Subject(s)
Endopeptidases/physiology , Lamin Type A/physiology , Retinoblastoma Protein/physiology , Sumoylation , Animals , Cells, Cultured , Cysteine Endopeptidases , E2F Transcription Factors/physiology , Mice , Proteasome Endopeptidase Complex/physiology , Protein Stability
2.
Genesis ; 31(2): 72-7, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11668681

ABSTRACT

The Sil gene encodes a cytosolic protein required for mouse embryonic midline and left/right axial development. Based on the phenotype of Sil mutant embryos, we hypothesized that Sil may be required for the activity of Sonic Hedgehog (Shh), a secreted signaling molecule also critically important for the development of the embryonic axes and found mutated in multiple types of cancer. Here we tested the genetic interaction between Sil and the Shh pathway by generating and analyzing embryos carrying mutations in both Sil and Patched (Ptch), a Shh receptor that normally inhibits the signaling pathway in the absence of ligand and when mutated leads to constitutive activation of the pathway. We find that Sil(-/-) Ptch(-/-) embryos do not activate the Shh pathway and instead have a phenotype indistinguishable from Sil(-/-) embryos, in which there is a loss of activity of Shh. These results provide genetic evidence that Sil is an essential component of the Shh response, acting downstream to Ptch.


Subject(s)
Embryo, Mammalian/metabolism , Membrane Proteins/genetics , Oncogene Proteins, Fusion , Proteins/genetics , Proteins/metabolism , Signal Transduction , Trans-Activators/metabolism , Animals , Cell Death/genetics , Crosses, Genetic , Embryo, Mammalian/embryology , Epistasis, Genetic , Female , Gene Deletion , Gene Expression Regulation, Developmental , Genotype , Head/embryology , Hedgehog Proteins , In Situ Hybridization , In Situ Nick-End Labeling , Intracellular Signaling Peptides and Proteins , Male , Membrane Proteins/deficiency , Mice , Mice, Knockout , Patched Receptors , Patched-1 Receptor , RNA, Messenger/analysis , RNA, Messenger/genetics , Receptors, Cell Surface
3.
Dev Biol ; 236(1): 124-35, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11456449

ABSTRACT

Nodal has been thought to be an embryo-specific factor that regulates development, but nodal is also expressed in the mouse placenta beginning at midgestation, specifically in the spongiotrophoblasts. In an insertional null nodal mutant, not only is embryonic development disrupted, but mouse placental development is also grossly altered with the loss of the diploid spongiotrophoblasts and labyrinth and an expansion of the polyploid giant cell layer. A hypomorphic mutation in nodal results in an expansion of the giant cell and spongiotrophoblast layers, and a decrease in labyrinthine development. Expression of nodal in trophoblast cell cultures is sufficient to inhibit trophoblast giant cell differentiation, demonstrating that nodal can act directly on trophoblasts. The mechanism of nodal action includes the inhibition of junB gene transcription. These results suggest that nodal may be involved in redirecting trophoblast fate towards the midgestational expansion of the labyrinth region while maintaining the thin layer of trophoblast giant cells and the underlying layer of spongiotrophoblasts that form the boundary between the maternal and extraembryonic compartments.


Subject(s)
Gene Expression Regulation, Developmental , Placenta/physiology , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/physiology , Trophoblasts/physiology , Animals , Cell Differentiation , Cells, Cultured , DNA/metabolism , Giant Cells/metabolism , Heterozygote , Mice , Microscopy, Fluorescence , Nodal Protein , Phenotype , Placenta/metabolism , Proto-Oncogene Proteins c-jun/biosynthesis , RNA/metabolism , Rats , Signal Transduction , Time Factors , Transcription, Genetic , Transfection
4.
Development ; 128(10): 1831-43, 2001 May.
Article in English | MEDLINE | ID: mdl-11311163

ABSTRACT

Loss-of-function analysis has shown that the transforming growth factor-like signaling molecule nodal is essential for mouse mesoderm development. However, definitive proof of nodal function in other developmental processes in the mouse embryo has been lacking because the null mutation blocks gastrulation. We describe the generation and analysis of a hypomorphic nodal allele. Mouse embryos heterozygous for the hypomorphic allele and a null allele undergo gastrulation but then display abnormalities that fall into three distinct mutant phenotypic classes, which may result from expression levels falling below critical thresholds in one or more domains of nodal expression. Our analysis of each of these classes provides conclusive evidence for nodal-mediated regulation of several developmental processes in the mouse embryo, beyond its role in mesoderm formation. We find that nodal signaling is required for correct positioning of the anteroposterior axis, normal anterior and midline patterning, and the left-right asymmetric development of the heart, vasculature, lungs and stomach.


Subject(s)
Body Patterning/genetics , Body Patterning/physiology , Mutation , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/physiology , Alleles , Animals , Female , Gastrula/physiology , Gene Expression Regulation, Developmental , Heterozygote , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nodal Protein , Phenotype , Signal Transduction
5.
J Biol Chem ; 276(1): 656-61, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11024047

ABSTRACT

Nodal, a member of the transforming growth factor beta (TGF-beta) superfamily, is implicated in many events critical to the early vertebrate embryo, including mesoderm formation, anterior patterning, and left-right axis specification. Here we define the intracellular signaling pathway induced by recombinant nodal protein treatment of P19 embryonal carcinoma cells. Nodal signaling activates pAR3-Lux, a luciferase reporter previously shown to respond specifically to activin and TGF-beta. However, nodal is unable to induce pTlx2-Lux, a reporter specifically responsive to bone morphogenetic proteins. We also demonstrate that nodal induces p(CAGA)(12), a reporter previously shown to be specifically activated by Smad3. Expression of a dominant negative Smad2 significantly reduces the level of luciferase reporter activity induced by nodal treatment. Finally, we show that nodal signaling rapidly leads to the phosphorylation of Smad2. These results provide the first direct biochemical evidence that nodal signaling is mediated by both activin-TGF-beta pathway Smads, Smad2 and Smad3. We also show here that the extracellular cripto protein is required for nodal signaling, making it distinct from activin or TGF-beta signaling.


Subject(s)
DNA-Binding Proteins/metabolism , Homeodomain Proteins , Inhibins/metabolism , Intercellular Signaling Peptides and Proteins , Membrane Glycoproteins , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Trans-Activators/metabolism , Transcription Factors , Transforming Growth Factor beta/pharmacology , Xenopus Proteins , Activins , Animals , Bone Morphogenetic Proteins/physiology , DNA-Binding Proteins/genetics , Embryonal Carcinoma Stem Cells , Epidermal Growth Factor/physiology , GPI-Linked Proteins , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , Growth Substances/metabolism , Humans , Membrane Proteins/metabolism , Mice , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Nodal Protein , Phosphorylation/drug effects , Recombinant Proteins/pharmacology , Smad2 Protein , Smad3 Protein , Trans-Activators/genetics , Transfection , Tumor Cells, Cultured
6.
Genesis ; 28(1): 1-14, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11020711

ABSTRACT

Mice mutant for the TGF-beta family member, nodal, lack mesoderm and die between E8.5 and E9.5. The short ear-lethal (se(l) ) mutation, a deletion that eliminates Bmp-5, causes a strikingly similar gastrulation defect. Here we analyze se(l);nodal compound mutants and find a dosage effect. Embryos homozygous for one mutation show distinct gastrulation stage defects that depend on whether they are heterozygous or homozygous for the other mutation. Embryos mutant for nodal or se(l);nodal compound mutants fail to execute an antigenic shift indicative of mesoderm differentiation and ectoderm cells are shunted into an apoptotic pathway. Furthermore, we find a novel phenotype in se(l);nodal double mutant litters, in which two to four genetically different embryos are contained within the same deciduum. Both the gastrulation and implantation phenotypes can also arise in short ear-viable (se(v) ) and se(v); nodal mutant mice. These data indicate that loss of Bmp-5 may underlie the se(l) gastrulation phenotype and suggest that nodal and Bmp-5 interact during murine mesoderm formation. Our data also reveal an unsuspected role for Bmp-5 in implantation and the decidual response in the mouse.


Subject(s)
Bone Morphogenetic Proteins/physiology , Embryo Implantation , Mesoderm/physiology , Transforming Growth Factor beta/physiology , Animals , Apoptosis/genetics , Bone Morphogenetic Protein 5 , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Embryo Implantation/genetics , Embryonic and Fetal Development/genetics , Female , Gastrula/metabolism , Gastrula/physiology , Genotype , Male , Mice , Mice, Inbred C3H , Mice, Mutant Strains , Mutation/genetics , Nodal Protein , Phenotype , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
7.
Teratology ; 62(5): 317-24, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11029149

ABSTRACT

BACKGROUND: Normal development of the left/right (L/R) body axis leads to the characteristic sidedness of asymmetric body structures, e.g., the left-sided heart. Several genes are now known to be expressed with L/R asymmetry during embryogenesis, including nodal, a member of the transforming growth factor-beta (TGF-beta) family. Mutations or experimental treatments that affect L/R development, such as those that cause situs inversus (reversal of the sidedness of asymmetric body structures), have been shown to alter or abolish nodal's asymmetric expression. METHODS: In the present study, we examined the effects on nodal expression of alpha(1)-adrenergic stimulation, known to cause a 50% incidence of situs inversus in rat embryos grown in culture, using reverse transcription-polymerase chain reaction assay and whole-mount in situ hybridization assay. RESULTS: In embryos cultured with phenylephrine, an alpha(1)-adrenergic agonist, nodal's normal asymmetric expression only in the left lateral plate mesoderm was altered. In some treated embryos, nodal expression was detected in either the left or right lateral plate mesoderm. However, most treated embryos lacked lateral plate mesoderm expression. In addition, the embryos that did show expression were at a later stage than when nodal expression is normally found. CONCLUSIONS: Our results demonstrate that alpha(1)-adrenergic stimulation delays the onset and perturbs the normal asymmetric pattern of nodal expression. Either of these effects might contribute to situs inversus.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Embryo, Mammalian/drug effects , Phenylephrine/pharmacology , Receptors, Adrenergic, alpha-1/metabolism , Situs Inversus/genetics , Transforming Growth Factor beta/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA Primers/chemistry , Embryo, Mammalian/metabolism , Female , In Situ Hybridization , Molecular Sequence Data , Nodal Protein , Organ Culture Techniques , Polymerase Chain Reaction , Pregnancy , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Situs Inversus/etiology , Situs Inversus/metabolism , Situs Inversus/pathology , Transforming Growth Factor beta/biosynthesis
10.
Development ; 126(23): 5495-504, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10556073

ABSTRACT

Vertebrates develop distinct asymmetries along the left-right axis, which are consistently aligned with the anteroposterior and dorsoventral axes. The mechanisms that direct this handed development of left-right asymmetries have been elusive, but recent studies of mutations that affect left-right development have shed light on the molecules involved. One molecule implicated in left-right specification is left-right dynein (LRD), a microtubule-based motor protein. In the LRD protein of the inversus viscerum (iv) mouse, there is a single amino acid difference at a conserved position, and the lrd gene is one of many genes deleted in the legless (lgl) mutation. Both iv and lgl mice display randomized left-right development. Here we extend the analysis of the lrd gene at the levels of sequence, expression and function. The complete coding sequence of the lrd gene confirms its classification as an axonemal, or ciliary, dynein. Expression of lrd in the node at embryonic day 7.5 is shown to be symmetric. At embryonic day 8.0, however, a striking asymmetric expression pattern is observed in all three germ layers of the developing headfold, suggesting roles in both the establishment and maintenance of left-right asymmetries. At later times, expression of lrd is also observed in the developing floorplate, gut and limbs. These results suggest function for LRD protein in both ciliated and non-ciliated cells, despite its sequence classification as axonemal. In addition, a targeted mutation of lrd was generated that deletes the part of the protein required for ATP binding, and hence motor function. The resulting left-right phenotype, randomization of laterality, is identical to that of iv and lgl mutants. Gross defects in ciliary structure were not observed in lrd/lrd mutants. Strikingly, however, the monocilia on mutant embryonic node cells were immotile. These results prove the identity of the iv and lrd genes. Further, they argue that LRD motor function, and resulting nodal monocilia movement, are required for normal left-right development.


Subject(s)
Adenosine Triphosphate/metabolism , Body Patterning/genetics , Dyneins/genetics , Dyneins/metabolism , Amino Acid Sequence , Animals , Axonemal Dyneins , Binding Sites , Catalytic Domain , Cilia/physiology , Cloning, Molecular , Functional Laterality , Gene Expression Regulation, Developmental , Head/embryology , Male , Mice , Mice, Inbred Strains , Molecular Sequence Data , Mutation , Nervous System/embryology , Sequence Analysis , Sequence Deletion
11.
Nature ; 399(6737): 691-4, 1999 Jun 17.
Article in English | MEDLINE | ID: mdl-10385121

ABSTRACT

The establishment of the main body axis and the determination of left-right asymmetry are fundamental aspects of vertebrate embryonic development. A link between these processes has been revealed by the frequent finding of midline defects in humans with left-right anomalies. This association is also seen in a number of mutations in mouse and zebrafish, and in experimentally manipulated Xenopus embryos. However, the severity of laterality defects accompanying abnormal midline development varies, and the molecular basis for this variation is unknown. Here we show that mouse embryos lacking the early-response gene SIL have axial midline defects, a block in midline Sonic hedgehog (Shh) signalling and randomized cardiac looping. Comparison with Shh mutant embryos, which have axial defects but normal cardiac looping, indicates that the consequences of abnormal midline development for left-right patterning depend on the time of onset, duration and severity of disruption of the normal asymmetric patterns of expression of nodal, lefty-2 and Pitx2.


Subject(s)
Body Patterning/genetics , Embryonic and Fetal Development/genetics , Nuclear Proteins , Oncogene Proteins, Fusion , Proteins/genetics , Trans-Activators , Animals , Body Patterning/physiology , Embryo, Mammalian/abnormalities , Embryonic and Fetal Development/physiology , Gene Targeting , Heart/embryology , Hedgehog Proteins , Homeodomain Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins , Left-Right Determination Factors , Mice , Mice, Nude , Mutagenesis , Neural Tube Defects/genetics , Nodal Protein , Paired Box Transcription Factors , Proteins/metabolism , Proteins/physiology , Signal Transduction , Stem Cells , Transcription Factors/biosynthesis , Transforming Growth Factor beta/biosynthesis , Homeobox Protein PITX2
12.
Development ; 126(6): 1225-34, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10021341

ABSTRACT

Left-right asymmetry in vertebrates is controlled by activities emanating from the left lateral plate. How these signals get transmitted to the forming organs is not known. A candidate mediator in mouse, frog and zebrafish embryos is the homeobox gene Pitx2. It is asymmetrically expressed in the left lateral plate mesoderm, tubular heart and early gut tube. Localized Pitx2 expression continues when these organs undergo asymmetric looping morphogenesis. Ectopic expression of Xnr1 in the right lateral plate induces Pitx2 transcription in Xenopus. Misexpression of Pitx2 affects situs and morphology of organs. These experiments suggest a role for Pitx2 in promoting looping of the linear heart and gut.


Subject(s)
Body Patterning , Digestive System/embryology , Genes, Homeobox , Heart/embryology , Homeodomain Proteins/metabolism , Nuclear Proteins , Transcription Factors/metabolism , Vertebrates/embryology , Abnormalities, Multiple/embryology , Activins , Amino Acid Sequence , Animals , Cell Communication , Gastrula , Gene Expression Regulation, Developmental , Inhibins/metabolism , Intracellular Signaling Peptides and Proteins , Mice , Mice, Mutant Strains , Molecular Sequence Data , Morphogenesis , Nodal Protein , Paired Box Transcription Factors , Sequence Homology, Amino Acid , Species Specificity , Tissue Distribution , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/metabolism , Xenopus , Xenopus Proteins , Zebrafish , Zebrafish Proteins , Homeobox Protein PITX2
13.
Dev Biol ; 193(1): 77-89, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-9466889

ABSTRACT

Patterning along the left/right axes helps establish the orientation of visceral organ asymmetries, a process which is of fundamental importance to the viability of an organism. A linkage between left/right and axial patterning is indicated by the finding that a number of genes involved in left/right patterning also play a role in anteroposterior and dorsoventral patterning. We have recovered a spontaneous mouse mutation causing left/right patterning defects together with defects in anteroposterior and dorsoventral patterning. This mutation is recessive lethal and was named no turning (nt) because the mutant embryos fail to undergo embryonic turning. nt embryos exhibit cranial neural tube closure defects and malformed somites and are caudally truncated. Development of the heart arrests at the looped heart tube stage, with cardiovascular defects indicated by ballooning of the pericardial sac and the pooling of blood in various regions of the embryo. Interestingly, in nt embryos, the direction of heart looping was randomized. Nodal and lefty, two genes that are normally expressed only in the left lateral plate mesoderm, show expression in the right and left lateral plate mesoderm. Lefty, which is normally also expressed in the floorplate, is not found in the prospective floor plate of nt embryos. This suggests the possibility of notochordal defects. This was confirmed by histological analysis and the examination of sonic hedgehog, Brachyury, and HNF-3 beta gene expression. These studies showed that the notochord is present in the early nt embryo, but degenerates as development progresses. Overall, these findings support the hypothesis that the notochord plays an active role in left/right patterning. Our results suggest that nt may participate in this process by modulating the notochordal expression of HNF-3 beta.


Subject(s)
Body Patterning/physiology , Fetal Proteins , Heart Defects, Congenital/embryology , Mutation/physiology , Neural Tube Defects/embryology , Notochord/embryology , T-Box Domain Proteins , Trans-Activators , Animals , DNA-Binding Proteins/genetics , Embryonic and Fetal Development , Gene Expression Regulation, Developmental , Genes, Lethal , Heart Defects, Congenital/genetics , Hedgehog Proteins , Hepatocyte Nuclear Factor 3-beta , Left-Right Determination Factors , Mice , Neural Tube Defects/genetics , Nodal Protein , Nuclear Proteins/genetics , Proteins/genetics , Somites , Transcription Factors/genetics , Transforming Growth Factor beta/genetics
14.
Dev Biol ; 189(1): 57-67, 1997 Sep 01.
Article in English | MEDLINE | ID: mdl-9281337

ABSTRACT

Recently, a pathway of genes which are part of a cascade regulating the side on which the heart forms during chick development was characterized (M. Levin et al., 1995, Cell 82, 1-20). Here we extend these previous studies, showing that manipulation of at least one member of the cascade, Sonic hedgehog (Shh), can affect the situs of embryonic rotation and of the gut, in addition to the heart. Bilateral expression of Shh, which is normally found exclusively on the left, does not result in left isomerism (a bilaterally symmetrical embryo having two left sides) nor in a complete situs inversus phenotype. Instead, misexpression of Shh on the right side of the node, which in turn leads to bilateral nodal expression, produces a heterotaxia-like condition, where different aspects of laterality are determined independently. Heart situs has previously been shown to be altered by ectopic Shh and activin. However, the most downstream gene identified in the LR pathway, nodal, had not been functionally linked to heart laterality. We show that ectopic (right-sided) nodal expression is able to affect heart situs, suggesting that the randomization of heart laterality observed in Shh and activin misexpression experiments is a result of changes in nodal expression and that nodal is likely to regulate heart situs endogenously. The first defined asymmetric signal in the left-right patterning pathway is Shh, which is initially expressed throughout Hensen's node but becomes restricted to the left side at stage 4(+). It has been hypothesized that the restriction of Shh expression may be due to repression by an upstream activin-like factor. The involvement of such an activin-like factor on the right side of Hensen's node was suggested because ectopic activin protein is able to repress Shh on the left side of the node, as well as to induce ectopic expression of a normally right-sided marker, the activin receptor cAct-RIIa. Here we provide further evidence in favor of this model. We find that a member of this family, Activin betaB, is indeed expressed asymmetrically, only on the right side of Hensen's node, at the correct time for it to be the endogenous asymmetric activin signal. Furthermore, we show that application of follistatin-loaded beads eliminates the asymmetry in Shh expression, consistent with an inhibition of an endogenous member of the activin-BMP superfamily. This combined with the previous data on exogenous activin supports the model that Activin betaB functions in the chick embryo to initiate Shh asymmetry. While these data extend our understanding of the early signals which establish left-right asymmetry, they leave unanswered the interesting question of how the bilateral symmetry of the embryo is initially broken to define a consistent left-right axis. Analysis of spontaneous chick twins suggests that, whatever the molecular mechanism, left-right patterning is unlikely to be due to a blastodermal prepattern but rather is initiated in a streak-autonomous manner.


Subject(s)
Embryonic Induction , Heart/embryology , Proteins/physiology , Trans-Activators , Animals , Chick Embryo , Hedgehog Proteins , Proteins/genetics
15.
Nature ; 381(6578): 158-61, 1996 May 09.
Article in English | MEDLINE | ID: mdl-8610013

ABSTRACT

Vertebrates have characteristic and conserved left-right (L-R) visceral asymmetries, for example the left-sided heart. In humans, alterations of L-R development can have serious clinical implications, including cardiac defects. Although little is known about how the embryonic L-R axis is established, a recent study in the chick embryo revealed L-R asymmetric expression of several previously cloned genes, including Cnr-1 (for chicken nodal-related-1), and indicated how this L-R molecular asymmetry might be important for subsequent visceral morphogenesis. Here we show that nodal is asymmetrically expressed in mice at similar stages, as is Xnr-1 (for Xenopus nodal related-1) in frogs. We also examine nodal expression in two mouse mutations that perturb L-R development, namely situs inversus viscerum (iv), in which assignment of L-R asymmetry is apparently random and individuals develop either normally or are mirror-image-reversed (situs inversus), and inversion of embryonic turning (inv), in which all individuals develop with situs inversus. In both, nodal expression is strikingly affected, being reversed or converted to symmetry. These results further support a key role for nodal and nodal-related genes in interpreting and relaying L-R patterning information in vertebrates. To our knowledge, our results provide the first direct evidence that iv and inv normally function well before the appearance of morphological L-R asymmetry.


Subject(s)
Morphogenesis/genetics , Mutation , Situs Inversus/genetics , Transforming Growth Factor beta/genetics , Animals , Chick Embryo , Embryo, Mammalian/abnormalities , Embryo, Mammalian/metabolism , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/metabolism , Female , Gene Expression Regulation, Developmental , Heterozygote , Homozygote , Male , Mice , Mice, Inbred BALB C , Nodal Protein , RNA, Messenger/metabolism , Transforming Growth Factor beta/biosynthesis , Xenopus , Xenopus Proteins
16.
Development ; 121(11): 3651-62, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8582278

ABSTRACT

Mouse embryos homozygous for a null mutation in nodal arrest development at early gastrulation and contain little or no embryonic mesoderm. Here, two Xenopus nodal-related genes (Xnr-1 and Xnr-2) are identified and shown to be expressed transiently during embryogenesis, first within the vegetal region of late blastulae and later in the marginal zone during gastrulation, with enrichment in the dorsal lip. Xnrs and mouse nodal function as dose-dependent dorsoanterior and ventral mesoderm inducers in whole embryos and explanted animal caps. Using a plasmid vector to produce Xnr proteins during gastrulation, we show that, in contrast to activin and other TGF beta-like molecules, Xnr-1 and Xnr-2 can dorsalize ventral marginal zone explants and induce muscle differentiation. Xnr signalling also rescues a complete embryonic axis in UV-ventralized embryos. The patterns of Xnr expression, the activities of the proteins and the phenotype of mouse nodal mutants, all argue strongly that a signaling pathway involving nodal, or nodal-related peptides, is an essential conserved element in mesoderm differentiation associated with vertebrate gastrulation and axial patterning.


Subject(s)
Embryonic Induction/physiology , Gastrula/physiology , Mesoderm/physiology , Proteins , Transforming Growth Factor beta/genetics , Xenopus Proteins , Xenopus/embryology , Amino Acid Sequence , Animals , Chick Embryo , In Situ Hybridization , Mice , Molecular Sequence Data , Morphogenesis/drug effects , Morphogenesis/genetics , Nodal Protein , Nodal Signaling Ligands , Sequence Alignment , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta/physiology
17.
Development ; 121(2): 383-91, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7768180

ABSTRACT

One of the first intercellular signalling events in the vertebrate embryo leads to mesoderm formation and axis determination. In the mouse, a gene encoding a new member of the TGF-beta superfamily, nodal, is disrupted in a mutant deficient in mesoderm formation (Zhou et al., 1993, Nature 361, 543). nodal mRNA is found in prestreak mouse embryos, consistent with a role in the development of the dorsal axis. To examine the biological activities of nodal, we have studied the action of this factor in eliciting axis determination in the zebrafish, Danio rerio. Injection of nodal mRNA into zebrafish embryos caused the formation of ectopic axes that included notochord and somites. Axis duplication was preceded by the generation of an apparent ectopic shield (organizer equivalent) in nodal-injected embryos, as indicated by the appearance of a region over-expressing gsc and lim1; isolation and expression in the shield of the lim1 gene is reported here. These results suggest a role for a nodal-like factor in pattern formation in zebrafish.


Subject(s)
DNA-Binding Proteins/genetics , Genes, Homeobox , Homeodomain Proteins/genetics , Mesoderm/physiology , Repressor Proteins , Transcription Factors , Zebrafish/embryology , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Embryonic Induction/physiology , Gene Expression , Goosecoid Protein , In Situ Hybridization , Microinjections , Molecular Sequence Data , Morphogenesis/genetics , RNA, Messenger/administration & dosage , Zebrafish/genetics , Zebrafish Proteins
18.
Lab Anim Sci ; 43(1): 91-3, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8459684

ABSTRACT

We examined whether superovulation can be used to improve the efficiency of donor embryo collection in embryonic stem cell injection chimera experiments. Superovulation of prepuberal C57BL/6 mice was compared with spontaneous ovulation of mature mice with respect to production and survival of blastocysts. Our results indicate that, compared with spontaneous ovulation, superovulation results in an increased number of blastocysts per female with no difference in viability when transferred to outbred Swiss Webster foster mothers. The advantage of using superovulated females, in terms of maintaining smaller numbers of both females and stud males, is discussed.


Subject(s)
Chimera , Embryo Transfer/veterinary , Superovulation , Tissue Donors , Animals , Blastocyst , Cell Survival , Female , Male , Mice , Mice, Inbred C57BL , Ovulation , Stem Cells
19.
Nature ; 361(6412): 543-7, 1993 Feb 11.
Article in English | MEDLINE | ID: mdl-8429908

ABSTRACT

During gastrulation, the three germ layers of the embryo are formed and organized along the anterior-posterior body axis. In the mouse, gastrulation involves the delamination of ectodermal cells through the primitive streak and their differentiation into mesoderm. These processes do not occur in embryos homozygous for a retrovirally induced recessive prenatal lethal mutation, the strain 413-d insertional mutation. Instead of giving rise to mesoderm, embryonic ectoderm in 413-d mutants overproliferates and then rapidly degenerates, although extraembryonic lineages remain viable. Here we isolate a candidate for the mutated gene which encodes a new member of the transforming growth factor-beta (TGF-beta) superfamily. Expression is first detected in primitive streak-stage embryos at about the time of mesoderm formation. It then becomes highly localized in the node at the anterior of the primitive streak. This region is analogous to chick Hensen's node and Xenopus dorsal lip (Spemann's organizer), which can induce secondary body axes when grafted into host embryos (reviewed in refs 5 and 6). Our findings suggest that this gene, named nodal, encodes a signalling molecule essential for mesoderm formation and subsequent organization of axial structures in early mouse development.


Subject(s)
Gastrula , Genes , Transforming Growth Factor beta/genetics , Amino Acid Sequence , Animals , Base Sequence , In Situ Hybridization , Mice , Molecular Sequence Data , Mutation , Polymerase Chain Reaction , Restriction Mapping
20.
Dev Dyn ; 194(3): 198-208, 1992 Jul.
Article in English | MEDLINE | ID: mdl-1467556

ABSTRACT

A transgenic mouse strain derived from embryonic stem (ES) cells infected with multiple copies of a retroviral vector carries a recessive insertional mutation resulting in prenatal lethality. A detailed histological analysis of developing embryos has shown that the mutation results in hyperplasia of both embryonic and extraembryonic ectoderm and failure of mesoderm formation in the egg cylinder stage embryo. The number of cells in each lineage of normal and mutant embryos was estimated using stereological analysis of serial sections taken from implantation sites. We observed a 2-fold increase in the number of embryonic ectoderm cells in mutant embryos at 7.5 days postcoitum (dpc). In addition, we found that mutant embryonic ectoderm cells are only 0.6 times as large as normal cells. The number of extraembryonic ectoderm cells in mutant embryos at 7.5 dpc is also increased, by almost 4-fold. Mutant extraembryonic ectoderm cells are also smaller than normal, being only two-thirds the size of wild-type cells. The mutant phenotype suggests that the gene identified by this insertional mutation plays an important role in the growth control of early embryonic lineages.


Subject(s)
Embryonic and Fetal Development/physiology , Genes , Mutagenesis, Insertional , Animals , Cell Count , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/pathology , Genotype , Hyperplasia , Image Processing, Computer-Assisted , Mice , Mice, Transgenic , Mutation , Phenotype , Reference Values
SELECTION OF CITATIONS
SEARCH DETAIL
...