Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
2.
Gene Expr Patterns ; 4(5): 569-72, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15261835

ABSTRACT

Wnt signaling has been shown to be important for axis formation in vertebrates. However, no Wnt ligand or receptor has been shown to be specifically expressed in all the organizer tissues in the mouse embryo. Here we report that the mouse frizzled 8 (mfz8) gene, a Wnt receptor, is expressed in the anterior visceral endoderm (AVE) and the anterior primitive streak, which have been shown to possess organizer activity. mFz8 is also expressed in the descendents of the anterior streak that comprise the anterior mesendoderm (AME) at midgastrulation, with subsequent expression in the anterior neurectoderm, which is specified and patterned by the AVE and AME. Thus, mfz8 is specifically expressed in the organizer tissues that establish the anterior-posterior axis in the mouse embryo.


Subject(s)
Body Patterning , Endoderm/metabolism , Gastrula/metabolism , Gene Expression Regulation, Developmental , Mice/embryology , Receptors, Cell Surface/metabolism , Animals , DNA, Complementary/genetics , Expressed Sequence Tags , In Situ Hybridization , Mice/genetics , Mice/metabolism , Receptors, Cell Surface/genetics , Xenopus Proteins/genetics
3.
J Environ Manage ; 67(3): 239-54, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12667474

ABSTRACT

Patterns of vegetation across Great Britain (GB) between 1990 and 1998 were quantified based on an analysis of plant species data from a total of 9596 fixed plots. Plots were established on a stratified random basis within 501 1 km sample squares located as part of the Countryside Survey of GB. Results are primarily conveyed in terms of a classification of national land-cover into 22 mutually exclusive Broad Habitat types. Each of the fixed vegetation plots could be assigned to the Broad Habitat in which they were located in either year. Two types of analysis are reported, both based on changes in plant species composition within monitoring plots. The first examined turnover and net change between Broad Habitat types. The second quantified more subtle changes that had occurred within each Broad Habitat using a series of condition measures that summarized multivariate plant species data as a single scalar value for each plot at each time. There are major difficulties in using uncontrolled, large-scale surveillance data to unravel causal linkages and no attempt was made to quantitatively partition variation among competing causes. However, it was clear that results were broadly consistent with environmental drivers known to have operated prior to and during the survey interval. Large-scale vegetation changes could be summarized in terms of shifts along gradients of substrate fertility and disturbance. Changes implied increased nutrient availability across upland and lowland ecosystems while, in lowland landscapes, linear features and small biotope fragments saw a marked shift to species compositions associated with greater shade and less disturbance.


Subject(s)
Conservation of Natural Resources , Environmental Monitoring , Plants , Data Collection , Ecosystem , Light , Nitrogen , Phosphorus , Population Dynamics , United Kingdom
4.
Development ; 128(18): 3609-21, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11566864

ABSTRACT

The Smad proteins are important intracellular mediators of the transforming growth factor beta (TGFbeta) family of secreted growth factors. Smad1 is an effector of signals provided by the bone morphogenetic protein (BMP) sub-group of TGFbeta molecules. To understand the role of Smad1 in mouse development, we have generated a Smad1 loss-of-function allele using homologous recombination in ES cells. Smad1-/- embryos die by 10.5 dpc because they fail to connect to the placenta. Mutant embryos are first recognizable by 7.0 dpc, owing to a characteristic localized outpocketing of the visceral endoderm at the posterior embryonic/extra-embryonic junction, accompanied by a dramatic twisting of the epiblast and nascent mesoderm. Chimera analysis reveals that these two defects are attributable to a requirement for Smad1 in the extra-embryonic tissues. By 7.5 dpc, Smad1-deficient embryos show a marked impairment in allantois formation. By contrast, the chorion overproliferates, is erratically folded within the extra-embryonic space and is impeded in proximal migration. BMP signals are known to be essential for the specification and proliferation of primordial germ cells. We find a drastic reduction of primordial germ cells in Smad1-deficient embryos, suggesting an essential role for Smad1-dependent signals in primordial germ cell specification. Surprisingly, despite the key involvement of BMP signaling in tissues of the embryo proper, Smad1-deficient embryos develop remarkably normally. An examination of the expression domains of Smad1, Smad5 and Smad8 in early mouse embryos show that, while Smad1 is uniquely expressed in the visceral endoderm at 6.5 dpc, in other tissues Smad1 is co-expressed with Smad5 and/or Smad8. Collectively, these data have uncovered a unique function for Smad1 signaling in coordinating the growth of extra-embryonic structures necessary to support development within the uterine environment.


Subject(s)
Bone Morphogenetic Proteins/metabolism , DNA-Binding Proteins/genetics , Extraembryonic Membranes/cytology , Genes, Lethal , Germ Cells/cytology , Trans-Activators/genetics , Animals , Cell Differentiation , Cell Lineage , Crosses, Genetic , DNA-Binding Proteins/isolation & purification , Ectoderm/pathology , Embryonic and Fetal Development , Gastrula/pathology , Genotype , Heterozygote , Mesoderm/pathology , Mice , Mice, Mutant Strains , Phenotype , Phosphoproteins/isolation & purification , Smad Proteins , Smad1 Protein , Smad5 Protein , Smad8 Protein , Stem Cells , Tissue Distribution , Trans-Activators/isolation & purification
5.
Dev Biol ; 235(2): 449-66, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11437450

ABSTRACT

The mature heart valves and septa are derived from the cardiac cushions which initially form as local outgrowths of mesenchymal cells within the outflow tract and atrioventricular regions. Endocardial cells respond to signals from the overlying myocardium and undergo an epithelial-to-mesenchymal transformation to invade the intervening extracellular matrix. The molecules that can induce and maintain these cell populations are not known, but many candidates, including several TGFbetas and BMPs, have been proposed based on their expression patterns and activities in other systems. In the present study, we describe the expression of Bmp6 and Bmp7 in overlapping and adjacent sites, including the cardiac cushions during mouse embryonic development. Previous analyses demonstrate that neither of these BMPs is required during cardiogenesis, but analysis of Bmp6;Bmp7 double mutants uncovers a marked delay in the formation of the outflow tract endocardial cushions. A proportion of Bmp6;Bmp7 mutants also display defects in valve morphogenesis and chamber septation, and the embryos die between 10.5 and 15.5 dpc due to cardiac insufficiency. These data provide the first genetic evidence that BMPs are involved in the formation of the cardiac cushions.


Subject(s)
Bone Morphogenetic Proteins/physiology , Heart/embryology , Myocardium/metabolism , Animals , Bone Morphogenetic Protein 6 , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/genetics , Bromodeoxyuridine/metabolism , Cell Division , Genotype , In Situ Hybridization , In Situ Nick-End Labeling , Mice , Mutation , Neurons/metabolism , Protein Binding , RNA, Messenger/metabolism , Time Factors , Transforming Growth Factor beta/metabolism , beta-Galactosidase/metabolism
6.
Curr Opin Genet Dev ; 11(4): 384-92, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11448624

ABSTRACT

Although much remains unknown about how the embryonic axis is laid down in the mouse, it is now clear that reciprocal interactions between the extraembryonic and embryonic lineages establish and reinforce patterning of the embryo. At early post-implantation stages, the extraembryonic ectoderm appears to impart proximal-posterior identity to the adjacent proximal epiblast, whereas the distal visceral endoderm signals to the underlying epiblast to restrict posterior identity as it moves anteriorward. At gastrulation, the visceral endoderm is necessary for specifying anterior primitive streak derivatives, which, in turn, pattern the anterior epiblast. Polarity of these extraembryonic tissues can be traced back to the blastocyst stage, where asymmetry has been linked to the point of sperm entry at fertilization.


Subject(s)
Axis, Cervical Vertebra/embryology , Body Patterning/physiology , Animals , Blastocyst/cytology , Cell Differentiation , Cell Movement , Ectoderm/physiology , Embryonic Development/physiology , Embryonic and Fetal Development , Female , Fertilization/physiology , Gastrula/physiology , Male , Mesoderm/physiology , Mice , Pregnancy , Spermatozoa/physiology , Zygote/physiology
7.
Nature ; 411(6840): 965-9, 2001 Jun 21.
Article in English | MEDLINE | ID: mdl-11418863

ABSTRACT

Shortly after implantation the mouse embryo comprises three tissue layers. The founder tissue of the embryo proper, the epiblast, forms a radially symmetric cup of epithelial cells that grows in close apposition to the extra-embryonic ectoderm and the visceral endoderm. This simple cylindrical structure exhibits a distinct molecular pattern along its proximal-distal axis. The anterior-posterior axis of the embryo is positioned later by coordinated cell movements that rotate the pre-existing proximal-distal axis. The transforming growth factor-beta family member Nodal is known to be required for formation of the anterior-posterior axis. Here we show that signals from the epiblast are responsible for the initiation of proximal-distal polarity. Nodal acts to promote posterior cell fates in the epiblast and to maintain molecular pattern in the adjacent extra-embryonic ectoderm. Both of these functions are independent of Smad2. Moreover, Nodal signals from the epiblast also pattern the visceral endoderm by activating the Smad2-dependent pathway required for specification of anterior identity in overlying epiblast cells. Our experiments show that proximal-distal and subsequent anterior-posterior polarity of the pregastrulation embryo result from reciprocal cell-cell interactions between the epiblast and the two extra-embryonic tissues.


Subject(s)
Body Patterning , Embryo, Mammalian/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Cell Polarity , DNA-Binding Proteins/metabolism , Ectoderm/metabolism , Embryo, Mammalian/cytology , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Mice , Nodal Protein , Smad2 Protein , Trans-Activators/metabolism , Transforming Growth Factor beta/genetics
8.
J Immunol ; 166(8): 5087-98, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11290790

ABSTRACT

Current ideas about DM actions have been strongly influenced by studies of mutant strains expressing the H-2(b) haplotype. To evaluate DM contributions to class II activities in BALB/c mice, we generated a novel mutation at the DMa locus via embryonic stem cell technology. Unlike long-lived A(b)/class II-associated invariant chain-derived peptide (CLIP) complexes, mature A(d) and E(d) molecules are loosely occupied by class II-associated invariant chain-derived peptide and are SDS unstable. BALB/c DM mutants weakly express BP107 conformational epitopes and toxic shock syndrome toxin-1 superantigen-binding capabilities, consistent with partial occupancy by wild-type ligands. Near normal numbers of mature CD4(+) T cells fail to undergo superantigen-mediated negative selection, as judged by TCR Vbeta usage. Ag presentation assays reveal consistent differences for A(d)- and E(d)-restricted T cells. Indeed, the mutation leads to decreased peptide capture by A(d) molecules, and in striking contrast causes enhanced peptide loading by E(d) molecules. Thus, DM requirements differ for class II structural variants coexpressed under physiological conditions in the intact animal.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Mice, Inbred BALB C/immunology , Peptides/immunology , Peptides/metabolism , Alleles , Animals , Antigen Presentation/genetics , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Clone Cells , Crosses, Genetic , Dimerization , Female , Gene Targeting , Haplotypes , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/genetics , Lymphocyte Activation/genetics , Male , Mice , Mice, Inbred BALB C/genetics , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Peptides/genetics , Protein Conformation , Sequence Deletion , Sodium Dodecyl Sulfate
9.
Planta ; 211(1): 150-7, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10923716

ABSTRACT

The peroxisome targeting signal (PTS) required for import of the rat acyl-CoA oxidase (AOX; EC 1.3.3.6) and the Candida tropicalis multifunctional protein (MFP) in plant peroxisomes was assessed in transgenic Arabidopsis thaliana (L.) Heynh. The native rat AOX accumulated in peroxisomes in A. thaliana cotyledons and targeting was dependent on the presence of the C-terminal tripeptide S-K-L. In contrast, the native C. tropicalis MFP, containing the consensus PTS sequence A-K-I was not targeted to plant peroxisomes. Modification of the carboxy terminus to the S-K-L tripeptide also failed to deliver the MFP to peroxisomes while addition of the last 34 amino acids of the Brassica napus isocitrate lyase, containing the terminal tripeptide S-R-M, enabled import of the fusion protein into peroxisomes. These results underline the influence of the amino acids adjacent to the terminal tripeptide of the C. tropicalis MFP on peroxisomal targeting, even in the context of a protein having a consensus PTS sequence S-K-L.


Subject(s)
Arabidopsis Proteins , Candida/genetics , Peroxisomes/metabolism , Acyl-CoA Oxidase , Animals , Arabidopsis/metabolism , Candida/metabolism , Fungal Proteins/metabolism , Fungal Proteins/physiology , Oxidoreductases/metabolism , Peroxisome-Targeting Signal 1 Receptor , Peroxisomes/genetics , Peroxisomes/physiology , Plants, Genetically Modified/metabolism , Rats , Receptors, Cytoplasmic and Nuclear/physiology
10.
Development ; 127(14): 3079-90, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10862745

ABSTRACT

TGFbeta growth factors specify cell fate and establish the body plan during early vertebrate development. Diverse cellular responses are elicited via interactions with specific cell surface receptor kinases that in turn activate Smad effector proteins. Smad2-dependent signals arising in the extraembryonic tissues of early mouse embryos serve to restrict the site of primitive streak formation and establish anteroposterior identity in the epiblast. Here we have generated chimeric embryos using lacZ-marked Smad2-deficient ES cells. Smad2 mutant cells extensively colonize ectodermal and mesodermal populations without disturbing normal development, but are not recruited into the definitive endoderm lineage during gastrulation. These experiments provide the first evidence that TGFbeta signaling pathways are required for specification of the definitive endoderm lineage in mammals and identify Smad2 as a key mediator that directs epiblast derivatives towards an endodermal as opposed to a mesodermal fate. In largely Smad2-deficient chimeras, asymmetric nodal gene expression is maintained and expression of pitx2, a nodal target, is also unaffected. These results strongly suggest that other Smad(s) act downstream of Nodal signals in mesodermal populations. We found Smad2 and Smad3 transcripts both broadly expressed in derivatives of the epiblast. However, Smad2 and not Smad3 mRNA is expressed in the visceral endoderm, potentially explaining why the primary defect in Smad2 mutant embryos originates in this cell population.


Subject(s)
DNA-Binding Proteins/metabolism , Endoderm/physiology , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation/physiology , Cell Lineage , DNA-Binding Proteins/genetics , Embryo, Mammalian/physiology , Female , Gene Expression Regulation, Developmental , Homozygote , Male , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Mutant Strains , Nodal Protein , Signal Transduction , Smad2 Protein , Smad3 Protein , Trans-Activators/genetics , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
11.
Genes Dev ; 14(9): 1146-55, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10809672

ABSTRACT

In vertebrates, specification of anteroposterior (A/P) and left-right (L/R) axes depends on TGFbeta-related signals, including Nodal, Lefty, and BMPs. Endoproteolytic maturation of these proteins is probably mediated by the proprotein convertase SPC1/Furin. In addition, precursor processing may be regulated by related activities such as SPC4 (also known as PACE4). Here, we show that a proportion of embryos lacking SPC4 develop situs ambiguus combined with left pulmonary isomerism or complex craniofacial malformations including cyclopia, or both. Gene expression analysis during early somite stages indicates that spc4 is genetically upstream of nodal, pitx2, lefty1, and lefty2 and perhaps maintains the balance between Nodal and BMP signaling in the lateral plate that is critical for L/R axis formation. Furthermore, genetic interactions between nodal and spc4, together with our analysis of chimeric embryos, strongly suggest that during A/P axis formation, SPC4 acts primarily in the foregut. These findings establish an important role for SPC4 in patterning the early mouse embryo.


Subject(s)
Body Patterning/genetics , Embryonic and Fetal Development/genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Signal Transduction/physiology , Abnormalities, Multiple/embryology , Abnormalities, Multiple/genetics , Animals , Bone Morphogenetic Proteins/physiology , Chromosome Mapping , Gene Expression Regulation, Developmental , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Mice , Mice, Knockout , Nodal Protein , Proprotein Convertases , Serine Endopeptidases/deficiency , Transforming Growth Factor beta/physiology
12.
Development ; 127(2): 245-54, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10603343

ABSTRACT

Furin, the mammalian prototype of a family of serine proteases, is required for ventral closure and axial rotation, and formation of the yolk sac vasculature. Here we show additionally that left-sided expression of pitx2 and lefty-2 are also perturbed in Furin-deficient embryos. These tissue abnormalities are preceded by a marked delay in the expansion of the definitive endoderm during gastrulation. Using a chimera approach, we show that Furin activity is required in epiblast derivatives, including the primitive heart, gut and extraembryonic mesoderm, whereas it is nonessential in the visceral endoderm. Thus, chimeric embryos, derived by injecting wild-type embryonic stem (ES) cells into fur(-/-) blastocysts, develop normally until at least 9.5 d.p.c. In contrast, Furin-deficient chimeras developing in the context of wild-type visceral endoderm fail to undergo ventral closure, axial rotation and yolk sac vascularization. Fur(-/-) cells are recruited into all tissues examined, including the yolk sac vasculature and the midgut, even though these structures fail to form in fur mutants. The presence of wild-type cells in the gut strikingly correlates with the ability of chimeric embryos to undergo turning. Overall, we conclude that Furin activity is essential in both extraembryonic and precardiac mesoderm, and in definitive endoderm derivatives.


Subject(s)
Chimera/genetics , Heart/embryology , Nuclear Proteins , Subtilisins/genetics , Animals , Endoderm/metabolism , Furin , Gastrula/metabolism , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/genetics , In Situ Hybridization , Left-Right Determination Factors , Mice , Mice, Knockout , Morphogenesis , Mutation , Paired Box Transcription Factors , Phenotype , RNA, Messenger/metabolism , Stem Cells/metabolism , Transcription Factors/genetics , Transforming Growth Factor beta/genetics , Viscera/embryology , Homeobox Protein PITX2
13.
Mol Cell ; 4(3): 287-98, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10518210

ABSTRACT

Mammalian lefty and zebrafish antivin form a subgroup of the TGF beta superfamily. We report that mouse mutants for lefty2 have an expanded primitive streak and form excess mesoderm, a phenotype opposite to that of mutants for the TGF beta gene nodal. Analogously, overexpression of Antivin or Lefty2 in zebrafish embryos blocks head and trunk mesoderm formation, a phenotype identical to that of mutants caused by loss of Nodal signaling. The lefty2 mutant phenotype is partially suppressed by heterozygosity for nodal. Similarly, the effects of Antivin and Lefty2 can be suppressed by overexpression of the nodal-related genes cyclops and squint or the extracellular domain of ActRIIB. Expression of antivin is dependent on Nodal signaling, revealing a feedback loop wherein Nodal signals induce their antagonists Lefty2 and Antivin to restrict Nodal signaling during gastrulation.


Subject(s)
Body Patterning , Gastrula/physiology , Mice/embryology , Transforming Growth Factor beta/metabolism , Zebrafish Proteins , Zebrafish/embryology , Activin Receptors, Type II , Animals , Feedback , Gene Expression Regulation, Developmental , Genes, Lethal , Heterozygote , Histocytochemistry , In Situ Hybridization , Left-Right Determination Factors , Mesoderm , Mice, Mutant Strains , Mutagenesis , Nodal Protein , Phenotype , RNA, Messenger , Receptors, Growth Factor/metabolism , Signal Transduction , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/genetics
14.
Int J Dev Biol ; 43(5): 405-11, 1999.
Article in English | MEDLINE | ID: mdl-10535316

ABSTRACT

Members of the Bone morphogenetic protein (BMP) family have been shown to be important signaling molecules throughout mouse development. Accordingly, many BMPs are also expressed during organogenesis of the metanephric kidney. However, only BMP7 has been shown to be absolutely required for proper formation of the kidney, thus the majority of information known involves this family member. BMP7 is expressed in both the ureteric epithelium and the mesenchyme throughout embryonic development and has been shown to function as a survival factor for the nephrogenic mesenchyme. However, there has been some controversy over the role of BMP7 as an inducing molecule for the metanephric mesenchyme. Recent studies have shown that BMP7 functions as an anti-differentiation factor for this mesenchyme cell population. The function of BMPs in the ureter and in the more differentiated epithelial structures of the nephron is less well understood.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Kidney/embryology , Transforming Growth Factor beta , Animals , Bone Morphogenetic Protein 7 , Cell Differentiation , Embryonic and Fetal Development , Gene Expression Regulation, Developmental , Humans , Kidney/metabolism , Mesoderm/metabolism , Mesoderm/physiology , Mice , Morphogenesis , Nephrons/embryology , Nephrons/metabolism , Wolffian Ducts/embryology , Wolffian Ducts/metabolism
15.
Mech Dev ; 85(1-2): 35-47, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10415345

ABSTRACT

Site specific recombinases have provided the experimental strategy necessary to modulate the expression of gene products in the mouse embryo. In this study we have exploited Cre recombinase to develop a widely applicable cell marking system which functions efficiently even at early post-implantation embryonic stages. Importantly, the techniques and reagents derived in this study are generally applicable to any recombinase driven approach, including strategies to temporally and spatially modulate endogenous or ectopic gene expression in the embryo. The cell marking scheme has two essential components which were derived as separate mouse lines. The first line carries a universal conditional lacZ reporter (UCR) locus which was prepared by using gene targeting in a novel approach to modify a ubiquitously expressed retroviral lacZ promoter trap insertion. The UCR locus is silent until it undergoes a Cre mediated DNA rearrangement to restore lacZ expression. To generate the Cre expressing allele, we outline a flexible strategy which requires the introduction of a novel IRES-Cre cassette into exon sequence of an endogenous locus by gene targeting. We successfully demonstrate this approach by generating a Cre expressing allele of the EphA2 gene, an Eph receptor protein tyrosine kinase expressed early in development. Analysis of double heterozygote embryos clearly demonstrates that Cre recombinase is expressed in vivo from the EphA2 IRES-Cre allele, and that the conditional reporter locus is efficiently restored in EphA2-expressing cells as early as 7.5 dpc. This cell marking experiment establishes the feasibility of expressing Cre recombinase from a single copy allele in the embryo and demonstrates the utility of the conditional reporter mouse which can be used in the analysis of any Cre expressing allele.


Subject(s)
Embryonic and Fetal Development/genetics , Gene Expression Regulation, Developmental , Integrases/genetics , Mutagenesis, Insertional , Viral Proteins , Animals , Base Sequence , Gene Targeting , Mice , Molecular Sequence Data , Mutagenesis, Insertional/methods
16.
Plant J ; 18(6): 651-62, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10417716

ABSTRACT

ARC (accumulation and replication of chloroplasts) genes control different aspects of the chloroplast division process in higher plants. In order to establish the hierarchy of the ARC genes in the chloroplast division process and to provide evidence for their specific roles, double mutants were constructed between arc11, arc6, arc5, arc3 and arc1 in all combinations and phenotypically analysed. arc11 is a new nuclear recessive mutant with 29 chloroplasts compared with 120 in wild type. All the phenotypes of the double mutants are unambiguous. ARC1 down-regulates proplastid division but is on a separate pathway from ARC3, ARC5, ARC6 and ARC11. ARC6 initiates both proplastid and chloroplast division. ARC3 controls the rate of chloroplast expansion and ARC11 the central positioning of the final division plane in chloroplast division. ARC5 facilitates separation of the two daughter chloroplasts. ARC5 maps to chromosome 3 and ARC11 and ARC6 map approximately 60 cM apart on chromosome 5.


Subject(s)
Arabidopsis/genetics , Chloroplasts , Genes, Plant , Arabidopsis/cytology , Chromosome Mapping , Genes, Recessive , Mutation
17.
Genes Dev ; 13(12): 1575-88, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10385626

ABSTRACT

The TGFbeta-related molecule Nodal is required for establishment of the anterior-posterior (A-P) and left-right (L-R) body axes of the vertebrate embryo. In mouse, several discrete sites of nodal activity closely correlate with its highly dynamic expression domains. nodal function in the posterior epiblast promotes primitive streak formation, whereas transient nodal expression in the extraembryonic visceral endoderm is essential for patterning the rostral central nervous system. Asymmetric nodal expression in the developing node and at later stages in left lateral plate mesoderm has been implicated as a key regulator of L-R axis determination. We have analyzed the cis-regulatory elements controlling nodal expression domains during early development. We show that the regulatory sequences conferring node-specific expression are contained in an upstream region of the locus, whereas early expression in the endoderm and epiblast and asymmetric expression at later stages on the left side of the body axis are controlled by a 600-bp intronic enhancer. Targeted deletion of a 100-bp subregion of this intronic enhancer eliminates nodal expression in the early epiblast and visceral endoderm and disrupts asymmetric expression in the node and lateral plate mesoderm. Thus, developmentally regulated nodal expression at distinct tissue sites during A-P and L-R axis formation is potentially controlled by common transcriptional activators.


Subject(s)
Gene Expression Regulation, Developmental , Regulatory Sequences, Nucleic Acid , Transforming Growth Factor beta/genetics , Animals , Base Sequence , Body Patterning , DNA, Complementary , Enhancer Elements, Genetic , Genes, Overlapping , Introns , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Molecular Sequence Data , Nodal Protein , Promoter Regions, Genetic , Somites
18.
Genes Dev ; 13(12): 1601-13, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10385628

ABSTRACT

Nephrogenesis in the mouse kidney begins at embryonic day 11 and ends approximately 10 days postpartum. During this period, new nephrons are continually being generated from a stem-cell population-the nephrogenic mesenchyme-in response to signals emanating from the tips of the branching ureter. Relatively little is known about the mechanism by which the nephrogenic mesenchyme cell population is maintained at the tips of the ureter in the presence of signals promoting tubulogenesis. Previous studies have shown that a loss of Bmp7 function leads to kidney defects that are a likely result of progressive loss of nephrogenic mesenchyme by apoptosis. The studies presented here demonstrate that BMP7 signaling can prevent apoptosis in explants of metanephric mesenchyme. The surviving mesenchyme cell population, however, is not competent to respond to signals promoting tubulogenesis. In conjunction with FGF2, BMP7 promotes growth and maintains competence of the mesenchyme in vitro. In addition, FGF2 and BMP7 signaling, both independently and in combination, inhibit tubulogenesis. Interestingly, FGF2 and BMP7 also promote expansion of the stromal progenitor cell population in whole kidney culture. Because BMP7 is not produced by stromal progenitor cells, these data suggest a novel interaction between the nephrogenic mesenchyme and stromal progenitor cell populations. A model for the regulation of nephrogenesis by FGF and BMP signaling is presented.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Fibroblast Growth Factor 2/metabolism , Kidney/embryology , Signal Transduction , Transforming Growth Factor beta , Animals , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/pharmacology , Fibroblast Growth Factor 2/genetics , Gene Expression , Kidney Tubules/embryology , Mesoderm , Mice , Receptors, Fibroblast Growth Factor/genetics
19.
Dev Biol ; 207(1): 176-88, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10049573

ABSTRACT

Targeted inactivation of the Bmp7 gene in mouse leads to eye defects with late onset and variable penetrance (A. T. Dudley et al., 1995, Genes Dev. 9, 2795-2807; G. Luo et al., 1995, Genes Dev. 9, 2808-2820). Here we report that the expressivity of the Bmp7 mutant phenotype markedly increases in a C3H/He genetic background and that the phenotype implicates Bmp7 in the early stages of lens development. Immunolocalization experiments show that BMP7 protein is present in the head ectoderm at the time of lens placode induction. Using an in vitro culture system, we demonstrate that addition of BMP7 antagonists during the period of lens placode induction inhibits lens formation, indicating a role for BMP7 in lens placode development. Next, to integrate Bmp7 into a developmental pathway controlling formation of the lens placode, we examined the expression of several early lens placode-specific markers in Bmp7 mutant embryos. In these embryos, Pax6 head ectoderm expression is lost just prior to the time when the lens placode should appear, while in Pax6-deficient (Sey/Sey) embryos, Bmp7 expression is maintained. These results could suggest a simple linear pathway in placode induction in which Bmp7 functions upstream of Pax6 and regulates lens placode induction. At odds with this interpretation, however, is the finding that expression of secreted Frizzled Related Protein-2 (sFRP-2), a component of the Wnt signaling pathway which is expressed in prospective lens placode, is absent in Sey/Sey embryos but initially present in Bmp7 mutants. This suggests a different model in which Bmp7 function is required to maintain Pax6 expression after induction, during a preplacodal stage of lens development. We conclude that Bmp7 is a critical component of the genetic mechanism(s) controlling lens placode formation.


Subject(s)
Bone Morphogenetic Proteins/genetics , Ectoderm/metabolism , Eye/growth & development , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins , Lens, Crystalline/growth & development , Membrane Proteins , Proteins , Transforming Growth Factor beta , Zebrafish Proteins , Animals , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Embryonic and Fetal Development , Eye/embryology , Eye Proteins/genetics , Gene Targeting , Immunochemistry , In Situ Hybridization , Lens, Crystalline/embryology , Mice , Mice, Knockout , Mutation/genetics , Organ Culture Techniques , PAX6 Transcription Factor , Paired Box Transcription Factors , Phenotype , Proto-Oncogene Proteins/genetics , Repressor Proteins , Signal Transduction/genetics , Wnt Proteins
20.
Development ; 126(8): 1753-68, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10079236

ABSTRACT

Members of the BMP family of signaling molecules display a high conservation of structure and function, and multiple BMPs are often coexpressed in a variety of tissues during development. Moreover, distinct BMP ligands are capable of activating common pathways. Here we describe the coexpression of two members of the 60A subfamily of BMPs, Bmp5 and Bmp7, at a number of different sites in the embryo from gastrulation onwards. Previous studies demonstrate that loss of either Bmp5 or Bmp7 has negligible effects on development, suggesting these molecules functionally compensate for each other at early stages of embryonic development. Here we show this is indeed the case. Thus we find that Bmp5;Bmp7 double mutants die at 10.5 dpc and display striking defects primarily affecting the tissues where these factors are coexpressed. The present analysis also uncovers novel roles for BMP signaling during the development of the allantois, heart, branchial arches, somites and forebrain. Bmp5 and Bmp7 do not appear to be involved in establishing pattern in these tissues, but are instead necessary for the proliferation and maintenance of specific cell populations. These findings are discussed with respect to potential mechanisms underlying cooperative signaling by multiple members of the TGF-beta superfamily.


Subject(s)
Bone Morphogenetic Proteins/physiology , Transforming Growth Factor beta , Allantoin , Animals , Apoptosis , Bone Morphogenetic Protein 5 , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/genetics , Chorion , Embryonic and Fetal Development , Epithelium/embryology , Gastrula , Gene Expression Regulation, Developmental , Heart/embryology , Humans , Mice , Morphogenesis , Mutagenesis , Neural Crest , RNA, Messenger , Somites
SELECTION OF CITATIONS
SEARCH DETAIL
...