Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Placenta ; 28(5-6): 412-20, 2007.
Article in English | MEDLINE | ID: mdl-16822546

ABSTRACT

Voltage-dependent Ca(2+) channels (VDCC) exist in most excitable cells and their properly regulated activity is essential for critical biological processes as many of these are sensitive to cellular Ca(2+) ion concentration. The ancillary cytoplasmic Ca(2+) channel beta subunits (CACNB) modulate Ca(2+) channel function and are required to enhance the number of functional channels in the plasma membrane. There are four genes encoding CACNB subunits and the gene encoding CACNB3 is over expressed in hyperplastic placentas of mouse interspecies hybrids. To determine the role of CACNB3 in the mouse placenta, we performed an expression and function analysis. Our results show that Cacnb3 exhibits specific spatial and temporal expression in the mouse placenta. Deletion of Cacnb3 does not produce a strong placental phenotype, which may be due to expression of other CACNB subunit encoding genes; however, sporadic occurrence of a labyrinthine architecture phenotype, characterized by reduced density of fetal blood vessels and decrease in pericyte number, could be observed. Down-regulation of Cacnb3 expression did not rescue placental hyperplasia in a model of interspecies hybrid placentas, which indicates that up-regulation in the hyperplastic placentas is a downstream event.


Subject(s)
Calcium Channels/genetics , Gene Expression Regulation , Placenta/physiology , Animals , Calcium Channels/deficiency , Calcium Channels/physiology , Cell Membrane/physiology , DNA/genetics , DNA/isolation & purification , DNA Primers , Female , Gene Deletion , Humans , Mice , Placenta/pathology , Pregnancy , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
2.
Placenta ; 27(9-10): 1014-21, 2006.
Article in English | MEDLINE | ID: mdl-16338003

ABSTRACT

The extracellular matrix protein fibulin-1 (FBLN1) is an important component of blood vessel walls, as shown by the lethality of mice with homozygous targeted deletion of the Fbln1 gene. Here, we show that a murine placental overgrowth phenotype is associated with elevated Fbln1 transcript levels, suggesting that the gene and its product have a functional role in placentation. Fbln1 exhibits a specific expression pattern in the mouse placenta. Transcripts could not be detected prior to day 12. In subsequent stages, Fbln1 was expressed strongly in the spongiotrophoblast. Other sites of expression were endothelia of large fetal blood vessels, a tissue type reported to not express this gene. In addition, a subset of giant cells expressed the gene. This giant cell specific expression was strongly increased in hyperplastic placentas. Analysis of the placentation in fibulin null mice did not show any abnormality. Attempts to rescue the placental phenotypes of a congenic model of interspecies hybrid placental dysplasia (IHPD) by normalizing expression of Fbln1 proved that Fbln1 alone is not the key cause of phenotypes in these models of placental hyperplasia.


Subject(s)
Calcium-Binding Proteins/physiology , Placenta/pathology , Placentation/physiology , Animals , Calcium-Binding Proteins/metabolism , Female , Gene Expression , Hyperplasia/metabolism , Mice , Mice, Inbred BALB C , Mutation , Placenta/metabolism , Pregnancy
4.
J Evol Biol ; 17(2): 453-60, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15009278

ABSTRACT

Interspecific hybridization in the rodent genera Peromyscus and Mus results in abnormal placentation. In the Peromyscus interspecies hybrids, abnormal allelic interaction between an X-linked locus and the imprinted paternally expressed Peg3 locus was shown to cause the placental defects. In addition, loss-of-imprinting (LOI) of Peg3 was positively correlated with increased placental size. As in extreme cases this placental dysplasia constitutes a post-zygotic barrier against interspecies hybridization, this finding was the first direct proof that imprinted genes may be important in speciation and thus in evolution. In the Mus interspecies hybrids, a strong role of an X-linked locus in placental dysplasia has also been detected. However, here we show by backcross and allele specific expression analyses that neither LOI of Peg3 nor abnormal interactions between Peg3 and an X-linked locus are involved in generating placental dysplasia in Mus hybrids, although the placental phenotypes observed in the two genera seem to be identical. In contrast to this, another dysgenesis effect common to Peromyscus and Mus hybrids, altered foetal growth, is caused at least in part by the same X-chromosomal regions in both genera. These findings first underline the strong involvement of the X-chromosome in the genetics of speciation. Secondly, they indicate that disruption of epigenetic states, such as LOI, at specific loci may be involved in hybrid dysgenesis effects in one group, but not in another. Thus, we conclude that even in closely related groups divergent molecular mechanisms may be involved in the production of phenotypically similar post-zygotic barriers against hybridization.


Subject(s)
Hybridization, Genetic , Muridae/physiology , Peromyscus/physiology , Placenta/abnormalities , Reproduction/physiology , X Chromosome/genetics , Alleles , Animals , Chromosome Mapping , DNA Primers , Epigenesis, Genetic/genetics , Genomic Imprinting , Histological Techniques , Lod Score , Muridae/genetics , Peromyscus/genetics , Polymorphism, Single-Stranded Conformational , Protein Kinases/genetics , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity , Transcription Factors/genetics
5.
Hum Mol Genet ; 10(26): 2983-7, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11751680

ABSTRACT

In the normal diploid mouse embryo, active demethylation of the paternal genome but not of the maternal genome occurs within only a few hours and in a highly coordinated fashion as the zygote proceeds through the first G1 phase. This zygotic demethylation may be necessary to reprogram the sperm genome for somatic development. Immunofluorescence staining with an antibody against 5-methylcytosine shows that the cellular machinery of the fertilized egg cannot demethylate the second maternal genome in parthenogenetic, gynogenetic and triploid digynic embryos or remethylate the additional (already demethylated) paternal genome in androgenetic and triploid diandric embryos. This suggests that differential zygotic demethylation results from differences in the remodeling of paternal and maternal chromatin structures after fertilization, i.e. sperm nuclear decondensation and protamine-histone exchange. A proportion of embryos derived from normal matings display abnormal methylation patterns some of which are indistinguishable from those in androgenetic or gynogenetic embryos. We conclude that methylation reprogramming defects in mammalian zygotes contribute to the high incidence of early pregnancy failure.


Subject(s)
DNA Methylation , Embryo, Mammalian/physiology , Animals , Antibodies, Antinuclear , Antibodies, Monoclonal , Embryonic and Fetal Development , Female , Fluorescent Antibody Technique , Male , Mice , Parthenogenesis
6.
Proc Natl Acad Sci U S A ; 98(23): 13126-31, 2001 Nov 06.
Article in English | MEDLINE | ID: mdl-11698681

ABSTRACT

The placenta is a highly specialized organ essential for embryonic growth and development. Here, we have applied cDNA subtraction between extraembryonic tissues of early- (day 7.5 of gestation) and late-stage embryos (day 17.5) to generate stage-specific cDNA pools that were used for screening of high-density mouse UniGene cDNA arrays containing 25,000 clones. A total of 638 clones were identified, 488 with the e7.5-specific probe and 150 with the e17.5-specific probe. Importantly, 363/638 (56.9%) of the hybridizing clones were not known to be expressed during placental development before. Differential regulation was confirmed by Northern blot and in situ hybridization for a total of 44/44 of positive clones. Thus, this combination of cDNA subtraction and array hybridization was highly successful for identification of genes expressed and regulated during placental development. These included growth factors and receptors, components of the transcriptional and translational machinery, cell cycle regulators, molecular chaperones, and cytoskeletal elements. The extensive in situ hybridization analysis revealed extraembryonic structures with a high density of differentially expressed genes, most strikingly the ectoplacental cone and the spongiotrophoblast. This large-scale identification of genes regulated during placentogenesis is extremely useful to further elucidate the molecular basis of extraembryonic development.


Subject(s)
DNA, Complementary/genetics , Oligonucleotide Array Sequence Analysis , Placentation , Transcription, Genetic , Animals , Cloning, Molecular , Gene Expression Regulation , In Situ Hybridization , Mice , Mice, Inbred Strains , Placenta/metabolism
7.
Trends Genet ; 17(12): 697-701, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11718922

ABSTRACT

The incidence of mental disability is 30% higher in males than in females. We have examined entries in the OMIM database that are associated with mental disability and for several other common defects. Our findings indicate that compared with the autosomes, the X chromosome contains a significantly higher number of genes that, when mutated, cause mental impairment. We propose that these genes are involved in the development of cognitive abilities and thus exert a large X-chromosome effect on general intelligence in humans. We discuss these conclusions with regard to the conservation of the vertebrate X-chromosomal linkage group and to human evolution.


Subject(s)
Cognition , Evolution, Molecular , Genetic Linkage , Intellectual Disability/genetics , Intelligence/genetics , X Chromosome , Animals , Brain/metabolism , Cognition Disorders/genetics , Conserved Sequence , Female , Fertility , Gene Frequency , Genes , Haplotypes , Humans , Male , Models, Genetic , Mutation , Selection, Genetic , Testis/physiology
8.
Genetics ; 157(1): 341-8, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11139514

ABSTRACT

It has been shown previously that abnormal placental growth, i.e., hyper- and hypoplasia, occurs in crosses and backcrosses between different mouse (Mus) species. A locus that contributes to this abnormal development has been mapped to the X chromosome. Unexpectedly, an influence of fetal sex on placental development has been observed, in that placentas attached to male fetuses tended to exhibit a more pronounced phenotype than placentas attached to females. Here, we have analyzed this sex dependence in more detail. Our results show that differences between male and female placental weights are characteristic of interspecific matings and are not observed in intraspecific Mus musculus matings. The effect is retained in congenic lines that contain differing lengths of M. spretus-derived X chromosome. Expression of the X-linked gene Pgk1 from the maternal allele only and lack of overall activity of two paternally inherited X-linked transgenes indicate that reactivation or lack of inactivation of the paternal X chromosome in trophoblasts of interspecific hybrids is not a frequent occurrence. Thus, the difference between male and female placentas seems not to be caused by faulty preferential X-inactivation. Therefore, these data suggest that the sex difference of placental weights in interspecific hybrids is caused by interactions with the Y chromosome.


Subject(s)
Dosage Compensation, Genetic , Placenta/abnormalities , Y Chromosome/genetics , Animals , Crosses, Genetic , Female , Hybridization, Genetic , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Muridae , Phenotype , Placentation , Pregnancy , Species Specificity
10.
Dev Biol ; 222(1): 158-69, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10885754

ABSTRACT

Trophoblast invasion is a critical process in development of most mammals that shares similarities with the invasive behavior of tumor cells. In the present investigation, a cDNA subtraction library was constructed between invasive trophoblast at day 8 of murine development and mature noninvasive placenta at day 18 of gestation. One of the differentially expressed clones, Epcs26, was mapped to the X chromosome and revealed no homology to any known gene. It was predominantly expressed in parietal endoderm, undifferentiated cells of the ectoplacental cone, and a few trophoblast giant cells. Another gene, designated Epcs50, was mapped to chromosome 19. It exhibited homologies to the mouse Mps1 gene and, like Mps1, may have a distant relationship to the lytic protein perforin. High expression was detected in parietal endoderm cells and in a subset of secondary trophoblast giant cells. Two sequences, Epcs24 and Epcs68, exhibited an extensive open reading frame that shared the common features of the cysteine proteinase cathepsin L. Expression was confined to an undefined subpopulation of trophoblast giant cells. Both genes were mapped to chromosome 13 in close proximity to cathepsins L and J. The known functions of MPS1 and cathepsin L proteins indicate that the related proteins EPCS50, EPCS24, and EPCS68 participate in conferring invasive properties to the mouse trophoblast.


Subject(s)
Cell Movement/genetics , Endopeptidases , Gene Expression , Proteins/genetics , Trophoblasts/cytology , Amino Acid Sequence , Animals , Base Sequence , Cathepsin L , Cathepsins/chemistry , Cloning, Molecular , Cysteine Endopeptidases , DNA Primers , DNA, Complementary , Enzyme Precursors/chemistry , Female , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Open Reading Frames , Proteins/chemistry , Sequence Homology, Amino Acid , Trophoblasts/metabolism
11.
Curr Biol ; 10(8): 475-8, 2000 Apr 20.
Article in English | MEDLINE | ID: mdl-10801417

ABSTRACT

DNA methylation is essential for the control of a number of biological mechanisms in mammals [1]. Mammalian development is accompanied by two major waves of genome-wide demethylation and remethylation: one during germ-cell development and the other after fertilisation [2] [3] [4] [5] [6] [7]. Most previous studies have suggested that the genome-wide demethylation observed after fertilisation occurs passively, that is, by the lack of maintenance methylation following DNA replication and cell division [6] [7], although one other study has reported that replication-independent demethylation may also occur during early embryogenesis [8]. Here, we report that genes that are highly methylated in sperm are rapidly demethylated in the zygote only hours after fertilisation, before the first round of DNA replication commences. By contrast, the oocyte-derived maternal alleles are unaffected by this reprogramming. They either remain methylated after fertilisation or become further methylated de novo. These results provide the first direct evidence for active demethylation of single-copy genes in the mammalian zygote and, moreover, reveal a striking asymmetry in epigenetic methylation reprogramming. Whereas paternally (sperm)-derived sequences are exposed to putative active demethylases in the oocyte cytoplasm, maternally (oocyte)-derived sequences are protected from this reaction. These results, whose generality is supported by findings of Mayer et al. [9], have important implications for the establishment of biparental genetic totipotency after fertilisation, the establishment and maintenance of genomic imprinting, and the reprogramming of somatic cells during cloning.


Subject(s)
DNA Methylation , Spermatozoa/chemistry , Zygote/metabolism , Actins/genetics , Animals , CpG Islands/genetics , DNA/genetics , DNA/metabolism , Female , Insulin-Like Growth Factor II/genetics , Male , Mice , Oocytes/chemistry , Transgenes , Zygote/chemistry
12.
Mamm Genome ; 11(4): 288-91, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10754104

ABSTRACT

We have studied the process of tspy gene silencing in murine evolution. We have isolated functional tspy sequences from Apodemus agrarius, A. sylvaticus, A. flavicollis, and Mus platythrix (subgenus Pyromys) and nonfunctional tspy sequences from species of the subgenus Mus. We present two alternative models as to how tspy may have lost its function in the murine lineage.


Subject(s)
Biological Evolution , DNA-Binding Proteins/genetics , Gene Silencing , Nuclear Proteins , Transcription Factors , Y Chromosome , Animals , Base Sequence , DNA Primers , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Sex-Determining Region Y Protein
13.
Dev Dyn ; 217(1): 1-10, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10679925

ABSTRACT

In the mouse fetus, Mest is widely expressed in mesoderm derived tissues. In separate studies in mice and in humans, it has been shown to be maternally imprinted, that is, only the paternally inherited allele is active. Here, we show that starting with implantation, Mest is also expressed in maternal decidua of the mouse and in placenta of both humans and mice. Expression in murine decidua was restricted to endothelial cells. After Day 7, expression in the decidua gradually decreased. Mest-specific RT-PCR and restriction fragment length variant (RFLV) analysis of decidualized endometrium isolated from (M. musculus x M. spretus)F1 females showed that only the paternally derived Mest allele was activated in the decidual endothelium. In the mouse extraembryonic tissues, Mest transcripts were detected in derivatives of extraembryonic mesoderm only. Here, hemangioblast precursor cells and endothelial cells were positive. At all developmental stages of the mouse, trophoblast-derived cells were clearly devoid of Mest transcripts. In the human placenta MEST transcripts were also detected in hemangioblast precursor cells, however, MEST was also expressed in villous and invasive cytotrophoblast. In a human choriocarcinoma/trophoblastic tumour grown in a nude mouse, human MEST was expressed in the tumour cells, whereas murine Mest was expressed in endothelia of the murine capillaries. The expression pattern exhibited by both Mest and MEST in extraembryonic tissues during development and during formation of choriocarcinoma/trophoblast tumour suggests a functional role of the MEST proteins related to oncofetal angiogenesis. Dev Dyn 2000;217:1-10.


Subject(s)
Gene Expression Regulation, Developmental , Neovascularization, Physiologic/genetics , Placenta/physiology , Proteins/genetics , Animals , Female , Genomic Imprinting , Humans , Mice , Pregnancy , Protein Biosynthesis
14.
J Cell Biol ; 148(4): 629-34, 2000 Feb 21.
Article in English | MEDLINE | ID: mdl-10684246

ABSTRACT

We have used two different experimental approaches to demonstrate topological separation of parental genomes in preimplantation mouse embryos: mouse eggs fertilized with 5-bromodeoxyuridine (BrdU)-labeled sperm followed by detection of BrdU in early diploid embryos, and differential heterochromatin staining in mouse interspecific hybrid embryos. Separation of chromatin according to parental origin was preserved up to the four-cell embryo stage and then gradually disappeared. In F1 hybrid animals, genome separation was also observed in a proportion of somatic cells. Separate nuclear compartments during preimplantation development, when extreme chromatin remodelling occurs, and possibly in some differentiated cell types, may be associated with epigenetic reprogramming.


Subject(s)
Blastocyst/cytology , Blastocyst/metabolism , Cell Nucleus/metabolism , Fathers , Genome , Mothers , Animals , Bromodeoxyuridine/metabolism , Cell Differentiation , Cell Nucleus/genetics , Cells, Cultured , Centromere/genetics , Centromere/metabolism , DNA/genetics , DNA/metabolism , Diploidy , Female , Fibroblasts , Heterochromatin/genetics , Heterochromatin/metabolism , Hybridization, Genetic , In Situ Hybridization, Fluorescence , Interphase , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Spermatozoa/cytology , Spermatozoa/metabolism , Zygote/cytology , Zygote/metabolism
17.
Genetics ; 153(1): 383-90, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10471720

ABSTRACT

Interspecific hybridization in the genus Mus results in male sterility and X-linked placental dysplasia. We have generated several congenic laboratory mouse lines (Mus musculus) in which different parts of the maternal X chromosome were derived from M. spretus. A strict positive correlation between placental weight and length of the M. spretus-derived part of the X chromosome was shown. Detailed analysis was carried out with one congenic strain that retained a M. spretus interval between 12.0 and 30.74 cM. This strain consistently produced hyperplastic placentas that exhibited an average weight increase of 180% over the weight of control placentas. In derived subcongenic strains, however, increased placental weight could no longer be observed. Morphometric analysis of these placentas revealed persistence of abnormal morphology. Fully developed placental hyperplasia could be reconstituted by recombination of proximal and central M. spretus intervals with an intervening M. musculus region. These results may suggest that placental dysplasia of interspecific mouse hybrids is caused by multiple loci clustered on the X chromosome that act synergistically. Alternatively, it is possible that changes in chromatin structure in interspecific hybrids that influence gene expression are dependent on the length of the alien chromosome.


Subject(s)
Genetic Linkage/genetics , Placenta/abnormalities , X Chromosome/genetics , Animals , Crosses, Genetic , Female , Fetus/abnormalities , Gene Expression , Genetic Markers/genetics , Genotype , Haplotypes/genetics , Male , Mice , Mice, Congenic , Mice, Inbred C3H , Mice, Inbred C57BL , Organ Size , Phenotype , Placenta/embryology , Placenta/pathology , Pregnancy
18.
Anat Embryol (Berl) ; 200(3): 335-43, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10463348

ABSTRACT

The placenta plays a pivotal role in fetal growth control and is considered a major site of genetic conflict between maternal and paternal genomes within the conceptus and, in addition, the genome of the mother. Accordingly, placental development is a strictly controlled process, and both placental and fetal weights do not vary much in intraspecific crosses of laboratory mice (Mus musculus). In mouse interspecific crosses and backcrosses [(M. musculus x M. spretus) x M. musculus], tremendous variation of placental, but not of fetal weight was observed. We have studied trophoblast cell type distribution and differentiation, and their effect on the associated placentas and fetuses in such backcrosses. Differentiation of spongious trophoblast, but not size of materno-fetal interface, correlated with fetal weight. Giant fetuses were observed only if less than one third of the spongiotrophoblast was formed by glycogen cells. Thus, placental efficiency was inversely related to the amount of glycogen cells. This influence of a trophoblast-derived cell type on fetal growth was not anticipated. We conclude that: (1) glycogen cells are able to negatively modulate fetal growth by an as yet unidentified mechanism; (2) correlation between fetal and placental weights is weak or absent in interspecific hybrids; (3) impaired control over placental and embryonic development in hybrids may contribute to post-mating isolation of species.


Subject(s)
Mice/genetics , Placenta/embryology , Trophoblasts/physiology , Animals , Body Constitution , Embryonic and Fetal Development , Female , Glycogen/metabolism , Hybridization, Genetic , In Situ Hybridization , Male , Maternal-Fetal Exchange , Mice/embryology , Placenta/anatomy & histology , Pregnancy , Species Specificity
19.
Gene ; 232(1): 35-42, 1999 May 17.
Article in English | MEDLINE | ID: mdl-10333519

ABSTRACT

We describe the cloning and characterization of the murine G90 gene, identified by subtractive hybridization based on the differential presence of its transcript in large and small intestine. The full-length cDNA and genomic sequences were cloned and found to produce a 1.5kb transcript that is polyadenylated but has no open reading frame larger than 249bp. The G90 gene was mapped to the proximal region of mouse chromosome 6. Expression analysis by Northern blotting showed that G90 is transcribed at very high levels in the small intestine and at lower levels in large intestine, testis and kidney of the mouse. In situ hybridization analysis on sections of small and large intestine and testis showed that G90 transcripts are present only in post-mitotic cells.


Subject(s)
Gene Expression , Intestine, Large/metabolism , Intestine, Small/metabolism , Open Reading Frames/genetics , RNA/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Chromosome Mapping , DNA, Complementary/genetics , In Situ Hybridization, Fluorescence , Mice , Molecular Sequence Data , Poly A/metabolism , RNA/metabolism , Sequence Analysis, DNA
20.
Hum Mol Genet ; 8(2): 229-36, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9931330

ABSTRACT

The von Hippel-Lindau (VHL) tumour suppressorgene product is believed to be involved in the down-regulation of transcriptional elongation by preventing the association of elongin B and C with the catalytic subunit elongin A. Alterations in the human VHL gene lead to VHL disease which is associated with various rare neoplasias, including haemangioblastoma of the central nervous system, retinal angioma, clear cell renal carcinoma and pheochromocytoma. Recently, a protein (VBP1) was isolated that was found to bind to the VHL protein in vivo. We have used the murine Vbp1 homologous cDNA to investigate the expression of the Vbp1 mRNA in the mouse by in situ hybridization and northern blot analysis. In fetal stages between days 9 and 18 of gestation, Vbp1 was expressed mainly in the central nervous system, retina and liver. In addition, at day 12, high expression was observed in the labyrinthine region of the placenta. In later stage placentas, Vbp1 expression was, however, considerably reduced. Northern blot analysis of adult mouse tissues showed that Vbp1 was ubiquitously expressed. In situ analysis on several adult tissues showed that in most tissues, transcripts were evenly distributed. In brain, eye, kidney and intestine, however, Vbp1 was expressed in specific cell types. Moreover, expression of the human VBP1 gene was investigated in cerebellum and in various tumours of VHL patients encompassinghaemangioblastomas, renal cell carcinomas and pheochromocytomas. In all of these tissues, VBP1 was ubiquitously expressed at low levels. However, no consistent differences in VBP1 expression levels could be detected between tumours and normal tissue. Mapping of the murine Vbp1 gene revealed conserved chromosomal localization between mouse and human in a region homologous to human Xq28.


Subject(s)
Carrier Proteins/genetics , Fetus/metabolism , Animals , Cerebellum/metabolism , Chromosome Mapping , Cytoskeletal Proteins , Embryo, Mammalian/metabolism , Embryonic and Fetal Development , Female , Gene Expression Regulation, Developmental , Humans , In Situ Hybridization , Male , Mice , Molecular Chaperones , Muridae , RNA/genetics , RNA/metabolism , Tissue Distribution , von Hippel-Lindau Disease/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...