Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 125
Filter
1.
Vet Pathol ; 49(1): 64-70, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21233329

ABSTRACT

Embryonic lethality is a common phenotype that occurs in mice that are homozygous for genetically engineered mutations. These phenotypes highlight the time and place that a gene is first required during embryogenesis. Early embryonic lethality (ie, before and up to mid-gestation) can be straightforward to analyze because the stage at which death occurs suggests why an embryo has failed. Here we summarize general strategies for analyzing early embryonic lethal phenotypes in genetically engineered mouse mutants.


Subject(s)
Embryonic Development/genetics , Fetal Death/diagnosis , Phenotype , Animals , Female , Genes, Lethal/genetics , Genetic Engineering , Mice , Mice, Transgenic , Models, Animal , Mutation , Pregnancy
2.
Osteoarthritis Cartilage ; 18(1): 88-96, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19932218

ABSTRACT

OBJECTIVE: To characterize the in vivo role epiphycan (Epn) has in cartilage development and/or maintenance. METHODS: Epn-deficient mice were generated by disrupting the Epn gene in mouse embryonic stem cells. Epn/biglycan (Bgn) double-deficient mice were produced by crossing Epn-deficient mice with Bgn-deficient mice. Whole knee joint histological sections were stained using van Gieson or Fast green/Safranin-O to analyze collagen or proteoglycan content, respectively. Microarray analysis was performed to detect gene expression changes within knee joints. RESULTS: Epn-deficient and Epn/Bgn double-deficient mice appeared normal at birth. No significant difference in body weight or femur length was detected in any animal at 1 month of age. However, 9-month Epn/Bgn double-deficient mice were significantly lighter and had shorter femurs than wild type mice, regardless of gender. Male Epn-deficient mice also had significantly shorter femurs than wild type mice at 9 months. Most of the deficient animals developed osteoarthritis (OA) with age; the onset of OA was observed earliest in Epn/Bgn double-deficient mice. Message RNA isolated from Epn/Bgn double-deficient knee joints displayed increased matrix protein expression compared with wild type mice, including other small leucine-rich proteoglycan (SLRP) members such as asporin, fibromodulin and lumican. CONCLUSION: Similar to other previously studied SLRPs, EPN plays an important role in maintaining joint integrity. However, the severity of the OA phenotype in the Epn/Bgn double-deficient mouse suggests a synergy between these two proteins. These data are the first to show a genetic interaction involving class I and class III SLRPs in vivo.


Subject(s)
Knee Joint/chemistry , Osteoarthritis, Knee/physiopathology , Proteoglycans/analysis , Proteoglycans/deficiency , Animals , Blotting, Southern , Body Weight , Collagen/analysis , Femur/anatomy & histology , Immunohistochemistry , Knee Joint/pathology , Mice , Mice, Knockout , Microarray Analysis , Osteoarthritis, Knee/genetics , Phenotype , Polymerase Chain Reaction , Proteoglycans/genetics , RNA, Messenger/analysis
3.
Article in English | MEDLINE | ID: mdl-20028846

ABSTRACT

Mammals have evolved a variety of morphological adaptations that have allowed them to compete in their natural environments. The developmental genetic basis of this morphological diversity remains largely unknown. Bats are mammals that have the unique ability of powered flight. We have examined the molecular embryology of bats and investigated the developmental genetic basis for their highly derived limbs used for flight. Initially, we developed an embryo staging system for a model chiropteran, Carollia perspicillata, the short-tailed fruit bat that has subsequently been used for staging other bat species. Expression studies focusing on genes that regulate limb development indicate that there are similarities and differences between bats and mice. To determine the consequences of these expression differences, we have conducted an enhancer switch assay by gene targeting in mouse embryonic stem cells to create mice whose genes are regulated by bat sequences. Our studies indicate that cis-regulatory elements contribute to the morphological differences that have evolved among mammalian species.


Subject(s)
Biological Evolution , Mammals/growth & development , Mammals/genetics , Animals , Chiroptera/genetics , Chiroptera/growth & development , Chiroptera/physiology , Enhancer Elements, Genetic , Extremities/growth & development , Flight, Animal/physiology , Gene Expression Regulation, Developmental , Gene Targeting , Genes, Homeobox , Mammals/physiology , Mice , Models, Genetic , Mutation , Phenotype
4.
Sex Dev ; 3(1): 1-15, 2009.
Article in English | MEDLINE | ID: mdl-19339813

ABSTRACT

Male sexual differentiation is a complex process requiring the hormone-producing function of somatic cells in the gonad, including Sertoli cells and fetal Leydig cells (FLCs). FLCs are essential for virilization of the male embryo, but despite their crucial function, relatively little is known about their origins or development. Adult Leydig cells (ALCs), which arise at puberty, have been studied extensively and much of what has been learned about this cell population has been extrapolated to FLCs. This approach is problematic in that prevailing dogma in the field asserts that these 2 populations are distinct in origin. As such, it is imprudent to assume that FLCs arise and develop in a similar manner to ALCs. This review provides a critical assessment of studies performed on FLC populations, rather than those extrapolated from ALC studies to assemble a model for FLC origins and development. Furthermore, we underscore the need for conclusive identification of the source population of fetal Leydig cells.


Subject(s)
Cell Differentiation , Leydig Cells/cytology , Leydig Cells/physiology , Sex Differentiation , Testis/growth & development , Animals , Embryo, Mammalian , Fetus , Humans , Male , Models, Biological , Testis/cytology
5.
Biol Reprod ; 78(6): 994-1001, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18322278

ABSTRACT

Amniotes, regardless of genetic sex, develop two sets of genital ducts: the Wolffian and Müllerian ducts. For normal sexual development to occur, one duct must differentiate into its corresponding organs, and the other must regress. In mammals, the Wolffian duct differentiates into the male reproductive tract, mainly the vasa deferentia, epididymides, and seminal vesicles, whereas the Müllerian duct develops into the four components of the female reproductive tract, the oviducts, uterus, cervix, and upper third of the vagina. In males, the fetal Leydig cells produce testosterone, which stimulates the differentiation of the Wolffian duct, whereas the Sertoli cells of the fetal testes express anti-Müllerian hormone, which activates the regression of the Müllerian duct. Anti-Müllerian hormone is a member of the transforming growth factor-beta (TGF-beta) family of secreted signaling molecules and has been shown to signal through the BMP pathway. It binds to its type II receptor, anti-Müllerian hormone receptor 2 (AMHR2), in the Müllerian duct mesenchyme and through an unknown mechanism(s); the mesenchyme induces the regression of the Müllerian duct mesoepithelium. Using tissue-specific gene inactivation with an Amhr2-Cre allele, we have determined that two TGF-beta type I receptors (Acvr1 and Bmpr1a) and all three BMP receptor-Smads (Smad1, Smad5, and Smad8) function redundantly in transducing the anti-Müllerian hormone signal required for Müllerian duct regression. Loss of these genes in the Müllerian duct mesenchyme results in male infertility due to retention of Müllerian duct derivatives in an otherwise virilized male.


Subject(s)
Activin Receptors, Type I/metabolism , Bone Morphogenetic Protein Receptors, Type I/metabolism , Mullerian Ducts/embryology , Mullerian Ducts/metabolism , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism , Activin Receptors, Type I/deficiency , Activin Receptors, Type I/genetics , Animals , Anti-Mullerian Hormone/pharmacology , Bone Morphogenetic Protein Receptors, Type I/deficiency , Bone Morphogenetic Protein Receptors, Type I/genetics , Female , Infertility, Male/embryology , Infertility, Male/genetics , Infertility, Male/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Models, Biological , Mullerian Ducts/drug effects , Pregnancy , Signal Transduction , Smad1 Protein/genetics , Smad5 Protein/genetics , Smad8 Protein/genetics
6.
Cytogenet Genome Res ; 101(3-4): 229-36, 2003.
Article in English | MEDLINE | ID: mdl-14684988

ABSTRACT

Doublesex and mab3 related transcript (DMRT1) was identified as a candidate gene for human 9p24.3 associated sex reversal. DMRT1 orthologues have highly conserved roles in sexual differentiation from flies and worms to humans. A DMRT1 orthologue was isolated from a marsupial, the tammar wallaby Macropus eugenii. The wallaby gene is highly conserved with other vertebrate DMRT1 genes, especially within the P/S and DM domains. It is expressed in the differentiating testis from the late fetus, during pouch life and in the adult. As in eutherian mammals, DMRT1 protein was localized in the germ cells and the Sertoli cells of the testis, but in addition it was detected in the Leydig cells, peri-tubular myoid cells and within the acrosome of the sperm heads. DMRT1 protein was also detected in the fetal and adult ovary pre-granulosa, granulosa and germ cells. Similarly, we also detected DMRT1 in the granulosa cells of all developing follicles in the adult mouse ovary. This is the first report of DMRT1 expression in the adult mammalian ovary, and suggests a wider role for this gene in mammals, in both the testis and ovarian function.


Subject(s)
Mammals/metabolism , Ovary/metabolism , Testis/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , DNA, Complementary/isolation & purification , Female , Immunohistochemistry , Male , Marsupialia , Mice , Molecular Sequence Data , Ovary/anatomy & histology , Ovary/chemistry , Phylogeny , Sequence Alignment , Testis/anatomy & histology , Testis/chemistry , Transcription Factors/analysis , Transcription Factors/genetics
7.
BJU Int ; 89(1): 113-8, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11849175

ABSTRACT

OBJECTIVE: To determine, in mice with disrupted Müllerian inhibiting substance (MIS) receptor genes, whether MIS affects gubernacular development; MIS causes Müllerian duct regression and is proposed to be involved in the first stage of testicular descent, because gubernacular development is abnormal in humans with persistent Müllerian duct syndrome. MATERIALS AND METHODS: Ten wild-type, 11 heterozygotic and 12 homozygotic mice for MIS receptor mutations were killed at 17.5 or 18.5 days after conception or at birth, to provide serial sagittal sections of the pelvis. The amount of cremaster muscle, mitotic bodies in the gubernacular bulb, and gubernacular size were quantified by computer analysis (four mice/group). RESULTS: Müllerian ducts were present in the homozygous mutants, partially present in the heterozygotes and absent in the wild-type controls. All mice had descended testes. The cremaster muscle was significantly less developed in homozygous mutants than in wild-type controls (P < 0.001) and heterozygotes (P < 0.01) at birth. The mitotic index between the gubernacula of all groups was indistinguishable. There was no statistical difference in gubernacular area amongst the groups. Poor cremaster muscle development in homozygous mutants gave the muscle a loose mesenchymal appearance. CONCLUSIONS: Although there was an observable effect on cremaster muscle development in these mutant mice, gubernacular development and testicular descent were otherwise normal, and thus there must be other reasons for the observed differences in humans with persistent Müllerian duct syndrome.


Subject(s)
Glycoproteins , Growth Inhibitors/deficiency , Mullerian Ducts/growth & development , Receptors, Peptide/genetics , Testicular Hormones/deficiency , Abdominal Muscles/growth & development , Animals , Anti-Mullerian Hormone , Cryptorchidism/genetics , Growth Inhibitors/genetics , Heterozygote , Homozygote , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , Mullerian Ducts/abnormalities , Receptors, Transforming Growth Factor beta , Testicular Hormones/genetics
8.
Mol Biotechnol ; 19(3): 297-304, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11721625

ABSTRACT

Gene targeting in mouse embryonic stem (ES) cells is a fundamental methodology for generating mice with precise genetic modifications. Although there are many complex gene targeting strategies for creating a variety of diverse mutations in mice, most investigators initially choose to generate a null allele. Here we provide a guide for the novice to generate a null allele for a protein coding gene using a fundamental gene targeting strategy. Ultimately, a well considered gene targeting strategy saves significant amounts of time, money, and research animal lives. The straightforward strategy presented here bypasses many of the pitfalls associated with gene knockouts generated by novices. This guide also serves as a foundation for subsequently designing more complex gene targeting strategies.


Subject(s)
Gene Targeting , Animals , Cloning, Molecular , Genetic Vectors , Mice
9.
Dev Cell ; 1(2): 277-90, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11702786

ABSTRACT

L-Sox5 and Sox6 are highly identical Sry-related transcription factors coexpressed in cartilage. Whereas Sox5 and Sox6 single null mice are born with mild skeletal abnormalities, Sox5; Sox6 double null fetuses die with a severe, generalized chondrodysplasia. In these double mutants, chondroblasts poorly differentiate. They express the genes for all essential cartilage extracellular matrix components at low or undetectable levels and initiate proliferation after a long delay. All cartilages are thus extracellular matrix deficient and remain rudimentary. While chondroblasts in the center of cartilages ultimately activate prehypertrophic chondrocyte markers, epiphyseal chondroblasts ectopically activate hypertrophic chondrocyte markers. Thick intramembranous bone collars develop, but the formation of cartilage growth plates and endochondral bones is disrupted. L-Sox5 and Sox6 are thus redundant, potent enhancers of chondroblast functions, thereby essential for endochondral skeleton formation.


Subject(s)
Cartilage/embryology , Cartilage/physiology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , High Mobility Group Proteins/metabolism , High Mobility Group Proteins/physiology , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Transcription Factors , Animals , Bone Development , Bone and Bones/abnormalities , Cell Differentiation , Chondrocytes/cytology , Chondrocytes/metabolism , Exostoses, Multiple Hereditary/genetics , In Situ Hybridization , Mice , Microscopy, Fluorescence , Models, Biological , Models, Genetic , Mutation , Phenotype , SOXD Transcription Factors
10.
Development ; 128(22): 4449-61, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11714671

ABSTRACT

We demonstrate that signaling via the bone morphogenetic protein receptor IA (BMPR-IA) is required to establish two of the three cardinal axes of the limb: the proximal-distal axis and the dorsal-ventral axis. We generated a conditional knockout of the gene encoding BMPR-IA (Bmpr) that disrupted BMP signaling in the limb ectoderm. In the most severely affected embryos, this conditional mutation resulted in gross malformations of the limbs with complete agenesis of the hindlimbs. The proximal-distal axis is specified by the apical ectodermal ridge (AER), which forms from limb ectoderm at the distal tip of the embryonic limb bud. Analyses of the expression of molecular markers, such as Fgf8, demonstrate that formation of the AER was disrupted in the Bmpr mutants. Along the dorsal/ventral axis, loss of engrailed 1 (En1) expression in the non-ridge ectoderm of the mutants resulted in a dorsal transformation of the ventral limb structures. The expression pattern of Bmp4 and Bmp7 suggest that these growth factors play an instructive role in specifying dorsoventral pattern in the limb. This study demonstrates that BMPR-IA signaling plays a crucial role in AER formation and in the establishment of the dorsal/ventral patterning during limb development.


Subject(s)
Body Patterning , Extremities/embryology , Protein Serine-Threonine Kinases/metabolism , Receptors, Growth Factor/metabolism , Transforming Growth Factor beta , Animals , Antigens, Differentiation , Bone Morphogenetic Protein 4 , Bone Morphogenetic Protein 7 , Bone Morphogenetic Protein Receptors, Type I , Bone Morphogenetic Proteins/isolation & purification , Ectoderm , Epithelium/embryology , Fibroblast Growth Factor 8 , Fibroblast Growth Factors/isolation & purification , Hindlimb/abnormalities , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Integrases , Limb Deformities, Congenital , Mesoderm , Mice , Mice, Knockout , Models, Biological , Organizers, Embryonic , Protein Serine-Threonine Kinases/genetics , Receptors, Growth Factor/genetics , Signal Transduction , Viral Proteins
12.
Osteoarthritis Cartilage ; 9 Suppl A: S69-75, 2001.
Article in English | MEDLINE | ID: mdl-11680692

ABSTRACT

OBJECTIVE: This work was carried out to identify transcription factors controlling the differentiation of mesenchymal cells into chondrocytes. DESIGN: We delineated a cartilage-specific enhancer in the collagen type 2 gene (Col2a1) and identified transcription factors responsible for the activity of this enhancer in chondrocytes. We then analyzed the ability of these transcription factors to activate specific genes of the chondrocyte differentiation program and control cartilage formation in vivo. RESULTS: A 48-bp sequence in the first intron of Col2a1 drove gene expression specifically in cartilage in transgenic mouse embryos. The transcription factors L-Sox5, Sox6, and Sox9 bound and cooperatively activated this enhancer in vitro. They belong to the Sry-related family of HMG box DNA-binding proteins, which includes many members implicated in cell fate determination in various lineages. L-Sox5, Sox6, and Sox9 were coexpressed in all precartilaginous condensations in mouse embryos and continued to be expressed in chondrocytes until the cells underwent final hypertrophy. Whereas L-Sox5 and Sox6 are highly homologous proteins, they are totally different from Sox9 outside the HMG box domain. The three proteins cooperatively activated the Col2a1- and aggrecan genes in cultured cells. Heterozygous mutations in SOX9 in humans lead to campomelic dysplasia, a severe and generalized skeletal malformation syndrome. Embryonic cells with a homozygous Sox9 mutation were unable to form cartilage in vivo and activate essential chondrocyte marker genes. Preliminary data indicated that the mutation of Sox5 and Sox6 in the mouse led to severe skeletal malformations. CONCLUSIONS: L-Sox5, Sox6, and Sox9 play essential roles in chondrocyte differentiation and, thereby, in cartilage formation. Their discovery will help to understand further the molecular mechanisms controlling chondrogenesis in vivo, uncover genetic mechanisms underlying cartilage diseases, and develop novel strategies for cartilage repair.


Subject(s)
Cell Differentiation/physiology , Chondrocytes/cytology , DNA-Binding Proteins/physiology , High Mobility Group Proteins/physiology , Nuclear Proteins/physiology , Transcription Factors/physiology , Animals , Collagen Type II/genetics , Gene Expression , Mesoderm/cytology , Mice , Mice, Transgenic , SOX9 Transcription Factor , SOXD Transcription Factors
13.
Mol Cell Biol ; 21(21): 7243-55, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11585907

ABSTRACT

During mammalian spermiogenesis, major restructuring of chromatin takes place. In the mouse, the histones are replaced by the transition proteins, TP1 and TP2, which are in turn replaced by the protamines, P1 and P2. To investigate the role of TP2, we generated mice with a targeted deletion of its gene, Tnp2. Spermatogenesis in Tnp2 null mice was almost normal, with testis weights and epididymal sperm counts being unaffected. The only abnormality in testicular histology was a slight increase of sperm retention in stage IX to XI tubules. Epididymal sperm from Tnp2-null mice showed an increase in abnormal tail, but not head, morphology. The mice were fertile but produced small litters. In step 12 to 16 spermatid nuclei from Tnp2-null mice, there was normal displacement of histones, a compensatory translationally regulated increase in TP1 levels, and elevated levels of precursor and partially processed forms of P2. Electron microscopy revealed abnormal focal condensations of chromatin in step 11 to 13 spermatids and progressive chromatin condensation in later spermatids, but condensation was still incomplete in epididymal sperm. Compared to that of the wild type, the sperm chromatin of these mutants was more accessible to intercalating dyes and more susceptible to acid denaturation, which is believed to indicate DNA strand breaks. We conclude that TP2 is not a critical factor for shaping of the sperm nucleus, histone displacement, initiation of chromatin condensation, binding of protamines to DNA, or fertility but that it is necessary for maintaining the normal processing of P2 and, consequently, the completion of chromatin condensation.


Subject(s)
Chromatin/ultrastructure , Fertility/genetics , Nuclear Proteins/genetics , Spermatozoa/physiology , Spermatozoa/ultrastructure , Animals , Blotting, Northern , Cell Nucleus/metabolism , Chromatin/metabolism , DNA-Binding Proteins , Flow Cytometry , Gene Deletion , Genotype , Immunoblotting , Male , Mice , Mice, Transgenic , Microscopy, Electron , Models, Genetic , Mutagenesis, Site-Directed , Mutation , Spermatogenesis/physiology , Spermatozoa/metabolism , Testis/ultrastructure
14.
Gene ; 277(1-2): 209-19, 2001 Oct 17.
Article in English | MEDLINE | ID: mdl-11602358

ABSTRACT

In eutherian mammals, such as mice and humans, steroidogenic factor 1 (SF1) plays important roles in the development of the gonad and in its steroidogenic activity. Marsupial and eutherian mammals have been evolving independently for at least 100 million years and so we were interested in comparing SF1 of a marsupial with that of eutherians. To this end, we have cloned SF1 from an Australian marsupial, the tammar wallaby. Although the amino acid sequence of SF1 is highly conserved among vertebrate species, tammar SF1 appears to have diverged less from the ancestral SF1 than have eutherian SF1 proteins. Tammar SF1 is expressed by both ovaries and testes on the day of birth, just prior to the onset of testicular differentiation, until at least 8 days after birth by which time the ovary also has begun to sexually differentiate. SF1 transcripts are localized predominantly to the pre-granulosa and Sertoli cells of the ovary and testis, respectively. In the testis SF1 transcripts are also present in the interstitial cells, although at a lower level than that which is observed in the Sertoli cells. SF1 is also transcribed in adult testis and ovary. In the adult ovary SF1 is expressed in the interstitial gland, and in the granulosa cells and theca interna of small to medium-sized antral follicles, but is not expressed in large antral follicles. Thus, although the structure of tammar SF1 is divergent from that of eutherians, its expression profile is similar, supporting a conserved role in gonadal development and steroidogenesis.


Subject(s)
DNA-Binding Proteins/genetics , Macropodidae/genetics , Sex Differentiation/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Animals, Newborn , Blotting, Northern , Chromosome Mapping , Cloning, Molecular , DNA, Complementary/chemistry , DNA, Complementary/genetics , Female , Fushi Tarazu Transcription Factors , Gene Expression , Gene Expression Regulation, Developmental , Homeodomain Proteins , In Situ Hybridization , In Situ Hybridization, Fluorescence , Macropodidae/growth & development , Male , Molecular Sequence Data , Ovary/growth & development , Ovary/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Steroidogenic Factor 1 , Testis/growth & development , Testis/metabolism , Tissue Distribution
15.
Development ; 128(18): 3623-34, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11566865

ABSTRACT

An organizer population has been identified in the anterior end of the primitive streak of the mid-streak stage embryo, by the expression of Hnf3beta, Gsc(lacZ) and Chrd, and the ability of these cells to induce a second neural axis in the host embryo. This cell population can therefore be regarded as the mid-gastrula organizer and, together with the early-gastrula organizer and the node, constitute the organizer of the mouse embryo at successive stages of development. The profile of genetic activity and the tissue contribution by cells in the organizer change during gastrulation, suggesting that the organizer may be populated by a succession of cell populations with different fates. Fine mapping of the epiblast in the posterior region of the early-streak stage embryo reveals that although the early-gastrula organizer contains cells that give rise to the axial mesoderm, the bulk of the progenitors of the head process and the notochord are localized outside the early gastrula organizer. In the mid-gastrula organizer, early gastrula organizer derived cells that are fated for the prechordal mesoderm are joined by the progenitors of the head process that are recruited from the epiblast previously anterior to the early gastrula organizer. Cells that are fated for the head process move anteriorly from the mid-gastrula organizer in a tight column along the midline of the embryo. Other mid-gastrula organizer cells join the expanding mesodermal layer and colonize the cranial and heart mesoderm. Progenitors of the trunk notochord that are localized in the anterior primitive streak of the mid-streak stage embryo are later incorporated into the node. The gastrula organizer is therefore composed of a constantly changing population of cells that are allocated to different parts of the axial mesoderm.


Subject(s)
Gastrula/cytology , Mesoderm/cytology , Mice/embryology , Organizers, Embryonic , Animals , Body Patterning , Cell Differentiation , Cell Lineage , Cell Movement , Embryonic Induction , Endoderm/cytology , Mice, Transgenic , Morphogenesis , Somites/cytology , Stem Cells , Tissue Transplantation
16.
Biochem Biophys Res Commun ; 286(5): 1019-26, 2001 Sep 07.
Article in English | MEDLINE | ID: mdl-11527402

ABSTRACT

We report the isolation of the mouse Ovca1 gene, the orthologue of human OVCA1/DPH2L1, a putative tumor suppressor associated with ovarian cancer. Mouse Ovca1 contains at least 13 exons and spans approximately 17 kb. Northern analysis showed that Ovca1 is expressed in most adult mouse tissues. The most predominant Ovca1 transcript is 2.1 kb. RT-PCR analysis demonstrated Ovca1 expression in embryos from 8.5 days postcoitum (d.p.c.) to 10.5 d.p.c., and various organs of 14.5 d.p.c. embryos. Mouse Ovca1 encodes a protein of 438 amino acids and has high identity with human OVCA1. Western blot and immunohistochemistry revealed that mouse OVCA1 is a 50-kDa protein that is predominately localized in a punctate pattern in the nucleus. Based on gene homology, structure, and expression patterns, these findings indicate that mouse Ovca1 is the orthologue of human OVCA1/DPH2L1. This study will facilitate experiments to elucidate the in vivo role of Ovca1 in cancer.


Subject(s)
Genes, Tumor Suppressor , Protein Biosynthesis , Proteins/genetics , Tumor Suppressor Proteins , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Blotting, Western , Cell Nucleus/metabolism , Cloning, Molecular , Embryo, Mammalian/metabolism , Exons , Humans , Immunohistochemistry , Mice , Microscopy, Fluorescence , Minor Histocompatibility Antigens , Models, Genetic , Molecular Sequence Data , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
17.
Biochem Biophys Res Commun ; 286(1): 133-40, 2001 Aug 10.
Article in English | MEDLINE | ID: mdl-11485319

ABSTRACT

Gastrula stage mouse embryo RNA was screened by degenerate RT-PCR to yield a novel paired-like homeobox gene. The open reading frame encoded by the cDNA was most similar to human VSX1. Mouse Vsx1 encodes a protein of 363 amino acid residues that contains a CVC domain that was originally identified as a conserved motif among mouse CHX10, goldfish VSX-1 and C. elegans CEH-10. Linkage analysis showed that mouse Vsx1 mapped to the distal region of chromosome 2. RT-PCR analysis detected mouse Vsx1 transcripts from gastrulation and post-gastrulation stage mouse embryos, suggesting a role for Vsx1 during mouse embryogenesis. Analysis of the eyes of mouse chimeras generated with embryonic stem cells in which a lacZ reporter was targeted to the Vsx1 locus suggested that Vsx1 is expressed in the inner nuclear layer of the retina.


Subject(s)
Embryonic and Fetal Development/genetics , Eye Proteins/genetics , Gene Expression Regulation, Developmental , Genes, Homeobox , Homeodomain Proteins/genetics , Retina/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary , Eye Proteins/chemistry , Genetic Linkage , Homeodomain Proteins/chemistry , Mice , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
18.
Bioessays ; 23(6): 508-17, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11385630

ABSTRACT

In many animal species, the early development of the embryo follows a stereotypic pattern of cell cleavage, lineage allocation and generation of tissue asymmetry leading to delineation of the body plan with three primary embryonic axes. The mammalian embryo has been regarded as an exception and primary body axes of the mouse embryo were thought to develop after implantation. However, recent findings have challenged this view. Asymmetry in the fertilised oocyte, as defined by the position of the second polar body and the sperm entry point, can be correlated with the orientation of the animal-vegetal and the embryonic-abembryonic axes in the preimplantation blastocyst. Studies of the pattern of morphogenetic movement of cells and genetic activity in the peri-implantation embryo suggest that the animal-vegetal axis of the blastocyst might presage the orientation of the anterior-posterior axis of the gastrula. This suggests that the asymmetry of the zygote that is established at fertilisation and early cleavage has a lasting impact on the delineation of body axes during embryogenesis.


Subject(s)
Blastocyst/physiology , Body Patterning , Gastrula/physiology , Mice/embryology , Morphogenesis , Animals , Blastocyst/cytology , Endoderm/cytology , Endoderm/physiology , Gastrula/cytology
19.
Proc Natl Acad Sci U S A ; 98(12): 6698-703, 2001 Jun 05.
Article in English | MEDLINE | ID: mdl-11371614

ABSTRACT

In humans, SOX9 heterozygous mutations cause the severe skeletal dysmorphology syndrome campomelic dysplasia. Except for clinical descriptions, little is known about the pathogenesis of this disease. We have generated heterozygous Sox9 mutant mice that phenocopy most of the skeletal abnormalities of this syndrome. The Sox9(+/-) mice died perinatally with cleft palate, as well as hypoplasia and bending of many skeletal structures derived from cartilage precursors. In embryonic day (E)14.5 heterozygous embryos, bending of radius, ulna, and tibia cartilages was already prominent. In E12.5 heterozygotes, all skeletal elements visualized by using Alcian blue were smaller. In addition, the overall levels of Col2a1 RNA at E10.5 and E12.5 were lower than in wild-type embryos. We propose that the skeletal abnormalities observed at later embryonic stages were caused by delayed or defective precartilaginous condensations. Furthermore, in E18.5 embryos and in newborn heterozygotes, premature mineralization occurred in many bones, including vertebrae and some craniofacial bones. Because Sox9 is not expressed in the mineralized portion of the growth plate, this premature mineralization is very likely the consequence of allele insufficiency existing in cells of the growth plate that express Sox9. Because the hypertrophic zone of the heterozygous Sox9 mutants was larger than that of wild-type mice, we propose that Sox9 also has a role in regulating the transition to hypertrophic chondrocytes in the growth plate. Despite the severe hypoplasia of cartilages, the overall organization and cellular composition of the growth plate were otherwise normal. Our results suggest the hypothesis that two critical steps of the chondrocyte differentiation pathway are sensitive to Sox9 dosage. First, an early step presumably at the stage of mesenchymal condensation of cartilage primordia, and second, a later step preceding the transition of chondrocytes into hypertrophic chondrocytes.


Subject(s)
Bone and Bones/abnormalities , Calcification, Physiologic , Cartilage/abnormalities , High Mobility Group Proteins/genetics , Mutation , Transcription Factors/genetics , Animals , Bone Diseases, Developmental/genetics , Cell Differentiation , Chondrocytes/physiology , High Mobility Group Proteins/physiology , Humans , Mesoderm/physiology , Mice , Mice, Inbred C57BL , SOX9 Transcription Factor , Transcription Factors/physiology
20.
Genomics ; 74(1): 105-8, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11374906

ABSTRACT

A novel human homeobox gene related to the mouse Esx1 homeobox gene, which we have designated ESXR1, has been identified. ESXR1 and Esx1 share 65% identity within their homeodomains and have glutamic acid-rich and proline-rich N- and C-terminal regions, respectively. Unlike Esx1, ESXR1 contains 12 repeats of a unique nine amino acid motif, PPMAP(V/L)PPG, located C-terminal to the homeodomain. The general exon-intron structures of ESXR1 and Esx1 appear to be conserved. ESXR1 has been localized to human Xq22.1-q22.3, the same region of synteny shared by the map position of Esx1. ESXR1 expression appears to be restricted to the placenta and testis, the tissues in which Esx1 is also expressed. These data suggest that ESXR1 may be the orthologue of Esx1. The findings that there are similarities between ESXR1 and Esx1, yet differences between their encoded products, are consistent with the idea that placental genes evolve rapidly between mammalian species.


Subject(s)
Genes, Homeobox/genetics , Homeodomain Proteins/genetics , Placenta/metabolism , Testis/metabolism , X Chromosome/genetics , Amino Acid Sequence , Base Sequence , Blotting, Northern , DNA, Complementary/chemistry , DNA, Complementary/genetics , Female , Gene Expression , Genetic Linkage , Humans , Male , Molecular Sequence Data , RNA/genetics , RNA/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...