Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
J Clin Transl Sci ; 2(2): 63-65, 2018 Apr.
Article in English | MEDLINE | ID: mdl-30364657

ABSTRACT

Several reports have shown that doctoral and postdoctoral trainees in biomedical research pursue diverse careers that advance science meaningful to society. Several groups have proposed a three-tier career taxonomy to showcase these outcomes. This three-tier taxonomy will be a valuable resource for institutions committed to greater transparency in reporting outcomes, to not only be transparent in reporting their own institutional data but also to lend greater power to a central repository.

2.
J Clin Invest ; 96(6): 2947-54, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8675666

ABSTRACT

Elevated levels of endogenous angiotensin can cause hypertensive nephrosclerosis as a result of the potent vasopressor action of the peptide. We have produced by gene targeting mice homozygous for a null mutation in the angiotensinogen gene (Atg-1-). Postnatally, Atg-1- animals show a modest delay in glomerular maturation. Although Atg-1- animals are hypotensive by 7 wk of age, they develop, by 3 wk of age, pronounced lesions in the renal cortex, similar to those of hypertensive nephrosclerosis. In addition, the papillae of homozygous mutant kidneys are reduced in size. These lesions are accompanied by local up-regulation of PDGF-B and TGF-beta1 mRNA in the cortex and down-regulation of PDGF-A mRNA in the papilla. The study demonstrates an important requirement for angiotensin in achieving and maintaining the normal morphology of the kidney. The mechanism through which angiotensin maintains the volume homeostasis in mammals includes promotion of the maturational growth of the papilla.


Subject(s)
Angiotensin II/metabolism , Angiotensinogen/deficiency , Angiotensinogen/genetics , Gene Expression , Growth Substances/biosynthesis , Kidney/growth & development , Aging , Angiotensinogen/blood , Animals , Blood Pressure , Body Weight , Homeostasis , Kidney/metabolism , Kidney/pathology , Kidney Cortex/growth & development , Kidney Cortex/pathology , Kidney Glomerulus/growth & development , Kidney Glomerulus/pathology , Kidney Medulla/growth & development , Kidney Medulla/pathology , Mice , Mice, Knockout , Mice, Mutant Strains , Organ Size , Platelet-Derived Growth Factor/biosynthesis , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Restriction Mapping , Transforming Growth Factor beta/biosynthesis
3.
Mech Dev ; 76(1-2): 185-90, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9767163

ABSTRACT

The embryonic neural crest is a unique group of cells that gives rise to the peripheral nervous system as well as many other cell types. Most of the work describing the behavior and specification of these cells has been done in the avian system, a system amenable to experimental manipulations such as tissue grafting, cell transplantation and lineage tracing. This work has been greatly facilitated by the use of molecular markers of neural crest cells such as HNK-1 and slug, markers that are not yet available for the study of mouse neural crest. Here we demonstrate that Hfh2 (HNF3 forkhead homologue 2), a member of the 'winged helix' or 'forkhead' transcription factor gene family, is expressed in premigratory and migrating neural crest cells in the early mouse embryo and in motorneuron progenitors in the developing spinal cord. Using linkage analysis we have localized the Hfh2 gene to chromosome 4 at 44.91 cM from the centromere.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins , Gene Expression Regulation, Developmental , Nerve Tissue Proteins , Neural Crest/embryology , Repressor Proteins/genetics , Spinal Cord/embryology , Animals , Binding Sites , Chromosome Mapping , DNA-Binding Proteins/metabolism , Early Growth Response Protein 2 , Forkhead Transcription Factors , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , In Situ Hybridization/methods , LIM-Homeodomain Proteins , Mice , Mice, Inbred Strains , Molecular Sequence Data , Motor Neurons/metabolism , Neural Crest/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Repressor Proteins/metabolism , Rhombencephalon/embryology , Rhombencephalon/metabolism , Spinal Cord/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
5.
J Thromb Haemost ; 6(2): 339-45, 2008 02.
Article in English | MEDLINE | ID: mdl-18036190

ABSTRACT

BACKGROUND: Activated factor X (FXa) is a vitamin K-dependent serine protease that plays a pivotal role in blood coagulation by converting prothrombin to thrombin. There are no reports of humans with complete deficiency of FX, and knockout of murine F10 is embryonic or perinatal lethal. OBJECTIVE: We sought to generate a viable mouse model of FX deficiency. METHODS: We used a socket-targeting construct to generate F10-knockout mice by eliminating F10 exon 8 (knockout allele termed F10(tm1Ccmt), abbreviated as '-'; wild-type '+'), and a plug-targeting construct to generate mice expressing a FX variant with normal antigen levels but low levels of FX activity [4-9% normal in humans carrying the defect, Pro343-->Ser, termed FX Friuli (mutant allele termed F10(tm2Ccmt), abbreviated as F)]. RESULTS: F10 knockout mice exhibited embryonic or perinatal lethality. In contrast, homozygous Friuli mice [F10 (F/F)] had FX activity levels of approximately 5.5% (sufficient to rescue both embryonic and perinatal lethality), but developed age-dependent iron deposition and cardiac fibrosis. Interestingly, F10 (-/F) mice with FX activity levels of 1-3% also showed complete rescue of lethality. Further study of this model provides evidence supporting a role of maternal FX transfer in the embryonic survival. CONCLUSIONS: We demonstrate that, while complete absence of FX is incompatible with murine survival, minimal FX activity as low as 1-3% is sufficient to rescue the lethal phenotype. This viable low-FX mouse model will facilitate the development of FX-directed therapies as well as investigation of the FX role in embryonic development.


Subject(s)
Factor X Deficiency/genetics , Factor X/genetics , Genomic Imprinting/genetics , Mice, Transgenic/genetics , Models, Animal , Amino Acid Substitution , Animals , Cardiomyopathies/etiology , Exons/genetics , Factor X Deficiency/complications , Female , Fetal Death/genetics , Fibrosis , Gene Targeting/methods , Genes, Lethal , Genetic Complementation Test , Genotype , Hemosiderosis/etiology , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic/blood , Myocardium/pathology
6.
Dev Biol ; 159(1): 232-44, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8103490

ABSTRACT

We wish to identify genes involved in mediating early lineage decisions in the mouse embryo. F9 teratocarcinoma cells treated with retinoic acid (RA) in suspension culture develop into embryoid bodies (EBs) with an outer layer of visceral endoderm. In order to identify genes that are involved in establishing this extraembryonic endoderm lineage we have employed a PCR-based approach using cDNAs from early EBs as templates. PCR reactions were performed with degenerate oligonucleotide primers coding for the highly conserved regions of the homeodomains of the Drosophila Antennapedia, bicoid, and zerknüllt proteins. Among the PCR products were representatives of previously identified mouse genes, including Hox-A5 (1.3), A1 (1.6), A9 (1.7), B8 (2.4), B2 (2.8), C8 (3.1), and D12 (4.7). Whole mount in situ hybridization analysis, performed to examine the temporal and spatial distribution of transcripts, suggests a possible role for the Hox-D12 gene during endoderm differentiation in F9 EBs. Whereas the expression patterns of several other homeobox genes are essentially uniform throughout the aggregates, Hox-D12 expression is restricted to the outer surface of early EBs at a time when lineage decisions may be occurring. In order to establish the relationship between the Hox-D12 expression pattern and the role of RA in inducing F9 EB differentiation, we examined PSA-1 EBs that differentiate in the absence of added RA. PSA-1 EBs show similar temporal and spatial localization of Hox-D12 when compared to F9 EBs. These data suggest that the pattern of Hox-D12 expression correlates with endoderm differentiation and not with RA treatment and point to a possible role for homeobox-containing genes during the early stages of mouse embryogenesis.


Subject(s)
Cell Differentiation/genetics , Endoderm/cytology , Genes, Homeobox , Amino Acid Sequence , Animals , Base Sequence , DNA , In Situ Hybridization , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Homology, Amino Acid , Teratoma , Tretinoin/pharmacology , Tumor Cells, Cultured
7.
Development ; 120(11): 3197-204, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7720562

ABSTRACT

Primordial germ cells of the mouse cultured on feeder layers with leukemia inhibitory factor, Steel factor and basic fibroblast growth factor give rise to cells that resemble undifferentiated blastocyst-derived embryonic stem cells. These primordial germ cell-derived embryonic germ cells can be induced to differentiate extensively in culture, form teratocarcinomas when injected into nude mice and contribute to chimeras when injected into host blastocysts. Here, we report the derivation of multiple embryonic germ cell lines from 8.5 days post coitum embryos of C57BL/6 inbred mice. Four independent embryonic germ cell lines with normal male karyotypes have formed chimeras when injected into BALB/c host blastocysts and two of these lines have transmitted coat color markers through the germline. We also show that pluripotent cell lines capable of forming teratocarcinomas and coat color chimeras can be established from primordial germ cells of 8.0 days p.c. embryos and 12.5 days p.c. genital ridges. We have examined the methylation status of the putative imprinting box of the insulin-like growth factor type 2 receptor gene (Igf2r) in these embryonic germ cell lines. No correlation was found between methylation pattern and germline competence. A significant difference was observed between embryonic stem cell and embryonic germ cell lines in their ability to maintain the methylation imprint of the Igf2r gene in culture. This may illustrate a fundamental difference between these two cell types.


Subject(s)
Genes , Genomic Imprinting , Germ Cells/physiology , Receptor, IGF Type 2/genetics , Stem Cells/physiology , Animals , Base Sequence , Blotting, Southern , Cell Differentiation/genetics , Cell Line , DNA Primers/genetics , Germ Cells/cytology , Methylation , Mice , Mice, Inbred Strains , Mice, Transgenic , Molecular Sequence Data , Polymerase Chain Reaction , Stem Cells/cytology
8.
Ciba Found Symp ; 182: 157-68; discussion 168-78, 1994.
Article in English | MEDLINE | ID: mdl-7835148

ABSTRACT

When primordial germ cells of the mouse are cultured on feeder layers with the addition of the polypeptide signalling molecules leukaemia inhibitory factor, Steel factor and basic fibroblast growth factor they give rise to cells that resemble undifferentiated blastocyst-derived embryonic stem cells. These primordial germ cell-derived embryonic germ cells (EG cells) can be induced to differentiate extensively in culture and also form teratocarcinomas when injected into nude mice. Additionally, they contribute to chimeras when injected into host blastocysts. We have derived multiple EG cell lines from 8.5 days post coitum (dpc) embryos of C57BL/6 inbred mice. Four independent EG cell lines with normal male karyotypes have formed chimeras (up to 70% coat colour chimerism) when injected into BALB/c host blastocysts. Chimeric mice from all four cell lines are fertile, but only those from one line have transmitted coat colour markers through the germline. Studies have also been carried out to determine whether gonadal primordial germ cells can give rise to pluripotent EG cells. Germ cells from gonads of 15.5 dpc C57BL/6 embryos and newborn mice failed to produce EG cell lines. EG cell lines capable of forming teratocarcinomas and coat colour chimeras have been established from primordial germ cells of 12.5 dpc genital ridges. We are currently testing the genomic imprinting status of the insulin-like growth factor type 2 receptor gene (Igf2r) in our different EG cell lines.


Subject(s)
Germ Cells/cytology , Mice/embryology , Animals , Blastocyst , Cell Differentiation , Cell Line , Cell Transplantation , Chimera , Female , Genomic Imprinting , Growth Substances/physiology , Hair Color/genetics , Male , Mice, Inbred Strains , Mice, Nude , Receptor, IGF Type 2/genetics , Stem Cells/cytology , Teratocarcinoma/etiology , Teratocarcinoma/pathology
9.
Development ; 128(21): 4127-38, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11684651

ABSTRACT

The neural crest is a migratory cell population that gives rise to multiple cell types in the vertebrate embryo. The intrinsic determinants that segregate neural crest cells from multipotential dorsal progenitors within the neural tube are poorly defined. In this study, we show that the winged helix transcription factor Foxd3 is expressed in both premigratory and migratory neural crest cells. Foxd3 is genetically downstream of Pax3 and is not expressed in regions of Pax3 mutant mice that lack neural crest, implying that Foxd3 may regulate aspects of the neural crest differentiation program. We show that misexpression of Foxd3 in the chick neural tube promotes a neural crest-like phenotype and suppresses interneuron differentiation. Cells that ectopically express Foxd3 upregulate HNK1 and Cad7, delaminate and emigrate from the neural tube at multiple dorsoventral levels. Foxd3 does not induce Slug and RhoB, nor is its ability to promote a neural crest-like phenotype enhanced by co-expression of Slug. Together these results suggest Foxd3 can function independently of Slug and RhoB to promote the development of neural crest cells from neural tube progenitors.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Neural Crest/cytology , Neurons/cytology , Repressor Proteins/genetics , Animals , Avian Proteins , Biomarkers , CD57 Antigens/genetics , Cadherins/genetics , Cell Differentiation/genetics , Cell Movement , Chick Embryo , DNA-Binding Proteins/metabolism , Embryonic Induction/genetics , Forkhead Transcription Factors , Helix-Turn-Helix Motifs , Mice , Mice, Mutant Strains , PAX3 Transcription Factor , Paired Box Transcription Factors , Repressor Proteins/metabolism , Snail Family Transcription Factors , Spinal Cord/embryology , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , rhoB GTP-Binding Protein/genetics
10.
Genes Dev ; 9(17): 2105-16, 1995 Sep 01.
Article in English | MEDLINE | ID: mdl-7657163

ABSTRACT

Bone morphogenetic protein-4 (BMP-4) is a member of the TGF-beta superfamily of polypeptide signaling molecules, closely related to BMP-2 and to Drosophila decapentaplegic (DPP). To elucidate the role of BMP-4 in mouse development the gene has been inactivated by homologous recombination in ES cells. Homozygous mutant Bmp-4tm1blh embryos die between 6.5 and 9.5 days p.c., with a variable phenotype. Most Bmp-4tm1blh embryos do not proceed beyond the egg cylinder stage, do not express the mesodermal marker T(Brachyury), and show little or no mesodermal differentiation. Some homozygous mutants develop to the head fold or beating heart/early somite stage or beyond. However, they are developmentally retarded and have truncated or disorganized posterior structures and a reduction in extraembryonic mesoderm, including blood islands. These results provide direct genetic evidence that BMP-4 is essential for several different processes in early mouse development, beginning with gastrulation and mesoderm formation. Moreover, in the presumed absence of zygotic ligand, it appears that homozygous mutants can be rescued partially by related proteins or by maternal BMP-4.


Subject(s)
Embryonic and Fetal Development , Gastrula/physiology , Mesoderm/physiology , Proteins/physiology , Animals , Base Sequence , Bone Morphogenetic Proteins , Cell Line , Chimera , Crosses, Genetic , Female , Gene Targeting , Heterozygote , Homozygote , Male , Mesoderm/cytology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Phenotype , Proteins/genetics , Stem Cells
11.
Genes Dev ; 10(13): 1657-69, 1996 Jul 01.
Article in English | MEDLINE | ID: mdl-8682296

ABSTRACT

Bone morphogenetic protein 8B (BMP8B) is a member of the TGFbeta superfamily of growth factors. In the mouse, Bmp8b is expressed in male germ cells of the testis and trophoblast cells of the placenta, suggesting that it has a role in spermatogenesis and reproduction. To investigate these possibilities, we have generated mice with a targeted mutation in Bmp8b. Here, we show that homozygous Bmp8b(tm1blh) mutant males exhibit variable degrees of germ-cell deficiency and infertility. Detailed analysis reveals two separable defects in the homozygous mutant testes. First, during early puberty (2 weeks old or younger) the germ cells of all homozygous mutants either fail to proliferate or show a marked reduction in proliferation and a delayed differentiation. Second, in adults, there is a significant increase in programmed cell death (apoptosis) of spermatocytes, leading to germ-cell depletion and sterility. Sertoli cells and Leydig cells appear relatively unaffected in mutants. This study therefore provides the first genetic evidence that a murine germ cell-produced factor, BMP8B, is required for the resumption of male germ-cell proliferation in early puberty, and for germ-cell survival and fertility in the adult.


Subject(s)
Growth Substances/physiology , Proteins/physiology , Spermatogenesis/genetics , Animals , Apoptosis , Bone Morphogenetic Proteins , Cell Differentiation , Cell Division , Female , Gene Expression Regulation, Developmental , Gene Targeting , Genes/physiology , Growth Substances/genetics , Homozygote , Infertility, Male/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Proteins/genetics , RNA, Messenger/analysis , Sertoli Cells , Sexual Maturation/genetics , Spermatozoa/cytology , Testis/cytology , Testis/growth & development , Testis/pathology
12.
Genomics ; 34(2): 241-5, 1996 Jun 01.
Article in English | MEDLINE | ID: mdl-8661058

ABSTRACT

Members of the winged helix family of transcription factors are required for the normal embryonic development of the mouse. Using the interspecific backcross panel from The Jackson Laboratory, we have determined the chromosomal locations of four genes that encode winged helix containing proteins. Mf1 was assigned to mouse Chromosome 8, Mf2 to Chromosome 4, Mf3 to Chromosome 9, and Mf4 to Chromosome 13. Since Mf3 is located in a region of Chromosome 9 containing many well-characterized mouse mutations such as short ear (se), ashen (ash), and dilute (d), we have analyzed deletion mutants to determine the location of Mf3 more precisely.


Subject(s)
Chromosome Mapping , Gene Expression Regulation, Developmental , Mice/genetics , Transcription Factors/genetics , Animals , Crosses, Genetic , DNA Probes , DNA, Complementary , Genetic Markers , Genomic Library , Mice, Mutant Strains , Multigene Family , Muridae , Restriction Mapping , Transcription Factors/biosynthesis
13.
Development ; 127(5): 1095-104, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10662648

ABSTRACT

BMP signaling is essential for a wide variety of developmental processes. To evaluate the role of Bmp2/4 in cranial neural crest (CNC) formation or differentiation after its migration into the branchial arches, we used Xnoggin to block their activities in specific areas of the CNC in transgenic mice. This resulted in depletion of CNC cells from the targeted areas. As a consequence, the branchial arches normally populated by the affected neural crest cells were hypomorphic and their skeletal and neural derivatives failed to develop. In further analyses, we have identified Bmp2 as the factor required for production of migratory cranial neural crest. Its spatial and temporal expression patterns mirror CNC emergence and Bmp2 mutant embryos lack both branchial arches and detectable migratory CNC cells. Our results provide functional evidence for an essential role of BMP signaling in CNC development.


Subject(s)
Bone Morphogenetic Proteins/physiology , Brain/embryology , Gene Expression Regulation, Developmental , Neural Crest/physiology , Osteogenesis , Transforming Growth Factor beta , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/genetics , Bone and Bones/embryology , Carrier Proteins , Embryonic and Fetal Development , Mice , Mice, Transgenic , Proteins/genetics , Proteins/physiology , Recombinant Proteins/metabolism , Signal Transduction
14.
Dev Biol ; 227(2): 495-509, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11071769

ABSTRACT

TGFbeta signals play important roles in establishing the body axes and germ layers in the vertebrate embryo. Vg1 is a TGFbeta-related gene that, due to its maternal expression and vegetal localization in Xenopus, has received close examination as a potential regulator of development in Xenopus, zebrafish, and chick. However, a mammalian Vg1 ortholog has not been identified. To isolate mammalian Vg1 we screened a mouse expression library with a Vg1-specific monoclonal antibody and identified a single cross-reactive clone encoding mouse Gdf1. Gdf1 is expressed uniformly throughout the embryonic region at 5.5-6.5 days postcoitum and later in the node, midbrain, spinal cord, paraxial mesoderm, lateral plate mesoderm, and limb bud. When expressed in Xenopus embryos, native GDF1 is not processed, similar to Vg1. In contrast, a chimeric protein containing the prodomain of Xenopus BMP2 fused to the GDF1 mature domain is efficiently processed and signals via Smad2 to induce mesendoderm and axial duplication. Finally, right-sided expression of chimeric GDF1, but not native GDF1, reverses laterality and results in right-sided Xnr1 expression and reversal of intestinal and heart looping. Therefore, GDF1 can regulate left-right patterning, consistent with the Gdf1 loss-of-function analysis in the mouse (C. T. Rankin, T. Bunton, A. M. Lawler, and S. J. Lee, 2000, Nature Genet. 24, 262-265) and a proposed role for Vg1 in Xenopus. Our results establish that Gdf1 is posttranslationally regulated, that mature GDF1 activates a Smad2-dependent signaling pathway, and that mature GDF1 is sufficient to reverse the left-right axis. Moreover, these findings demonstrate that GDF1 and Vg1 are equivalent in biochemical and functional assays, suggesting that Gdf1 provides a Vg1-like function in the mammalian embryo.


Subject(s)
Body Patterning/genetics , Bone Morphogenetic Proteins/genetics , Glycoproteins/genetics , Amino Acid Sequence , Animals , Antigens/genetics , Bone Morphogenetic Proteins/immunology , Bone Morphogenetic Proteins/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Glycoproteins/immunology , Glycoproteins/physiology , Growth Differentiation Factors , In Situ Hybridization , Mesoderm/cytology , Mesoderm/metabolism , Mice , Molecular Sequence Data , Protein Processing, Post-Translational , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Signal Transduction , Smad2 Protein , Trans-Activators/genetics , Trans-Activators/physiology , Transforming Growth Factor beta , Xenopus , Xenopus Proteins
15.
Genesis ; 29(2): 60-71, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11170346

ABSTRACT

The ability to lactate is a process restricted to mammals and is necessary for the survival of nonhuman mammals. Female mice carrying a null mutation in the winged helix transcription factor Foxb1 (previously Mf3/Fkh5/TWH) have lactation defects on inbred genetic backgrounds. To determine the cellular basis of the Foxb1 lactation defect we have inserted a tau-lacZ lineage marker into the locus to follow the fate of Foxb1 expressing cells. This approach has revealed that Foxb1 is expressed in epithelial cells of developing and adult mammary glands as well as previously described regions of the central nervous system. Mammary glands from C57BL/6 Foxb1-/- mice have incomplete lobuloalveolar development. In addition, the tau-lacZ lineage label was used to determine that the mammillothalamic tract was lost in all Foxb1-/- mice. Finally, morphological defects in the inferior colliculi of the midbrain in Foxb1-/- mice correlate with the inability to lactate, suggesting that the midbrain defect, but not the loss of the mammillothalamic tract, may be responsible for the lactation defect.


Subject(s)
Brain/growth & development , DNA-Binding Proteins/genetics , Gene Targeting/methods , Mammary Glands, Animal/growth & development , Mammillary Bodies/growth & development , Milk Ejection/physiology , Stem Cells/physiology , Transcription Factors/genetics , Animals , Brain/embryology , Crosses, Genetic , Female , Forkhead Transcription Factors , Heterozygote , Homozygote , Immunoenzyme Techniques , Lac Operon/physiology , Male , Mammary Glands, Animal/embryology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Phenotype , Reflex/genetics , tau Proteins/metabolism
16.
Proc Natl Acad Sci U S A ; 94(4): 1402-7, 1997 Feb 18.
Article in English | MEDLINE | ID: mdl-9037065

ABSTRACT

Matrix metalloproteinases (MMPs) classically have been implicated in basement membrane destruction associated with late-stage tumor cell invasion and metastasis. However, recent studies have demonstrated that one MMP family member, matrilysin, is expressed in a high percentage of early-stage human colorectal tumors. We analyzed matrilysin expression in benign intestinal tumors from mice heterozygous for the ApcMin allele (Min/+) and found that the mRNA was induced in the majority (88%) of these adenomas. Protein was detected in the tumor cells, where, surprisingly, it was predominantly immunolocalized to the lumenal surface of dysplastic glands rather than the basement membrane or extracellular matrix. To address the role of matrilysin in Min intestinal tumorigenesis, we generated Min/+ mice deficient in this MMP by gene targeting and homologous recombination. The absence of matrilysin resulted in a reduction in mean tumor multiplicity in Min/+ animals of approximately 60% and a significant decrease in the average tumor diameter. Based on these findings, we conclude that matrilysin is a suppressor of the Min phenotype, possibly by functioning in a capacity independent of matrix degradation. These results argue for the use of MMP inhibitors in the treatment and prevention of early-stage colon cancer.


Subject(s)
Adenoma/genetics , Adenomatous Polyposis Coli/genetics , Genes, APC , Genes, Tumor Suppressor , Metalloendopeptidases/deficiency , Animals , Clone Cells , Disease Models, Animal , Immunohistochemistry , In Situ Hybridization , Matrix Metalloproteinase 7 , Metalloendopeptidases/genetics , Metalloendopeptidases/isolation & purification , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutagenesis, Site-Directed , Phenotype , Stem Cells , Tissue Distribution
17.
Dev Biol ; 166(1): 170-9, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7958444

ABSTRACT

We previously reported the isolation of a cDNA clone for a homeobox-containing gene designated Pem, shown by Northern analysis of Day 7 through Day 16 mouse embryos to be expressed in extraembryonic tissues. In this study, Pem gene expression was further examined using in situ hybridization and immunocytochemistry to determine the spatial distribution of Pem transcripts and protein in peri-implantation embryos and in embryoid bodies (EBs). Low amounts of Pem mRNA were detected in undifferentiated EBs. When EBs were induced to differentiate, the outer cell layer of visceral or parietal endoderm expressed both Pem mRNA and protein. In developing embryos, no Pem protein was detectable in the uncompacted morula; 12% of the nuclei in compacted morulae were Pem positive, while 25% of the blastocyst trophectoderm and 15% of inner cell mass cells expressed Pem protein. Shortly after implantation, in 5.5 and 6.5 d.p.c. embryos, Pem expression was limited to extraembryonic tissues and was present in distal and proximal visceral endoderm, parietal endoderm, and ectoplacental cone. By 7.5-8.5 d.p.c. neither Pem RNA nor protein was found in the distal squamous visceral endoderm, which surrounds the embryonic region of the egg cylinder, nor in the parietal endoderm. Expression was retained in the proximal columnar epithelium of the visceral endoderm. Prominent Pem expression was observed in the chorion, in trophoblast-derived cells of the ectoplacental cone, and in secondary giant cells, localized in the nuclear compartment. Pem was localized to the X chromosome and found to be expressed in cell lineages where only the maternal X chromosome is active. The data indicate a possible role for Pem in regulating genes involved in the differentiation of extraembryonic tissues.


Subject(s)
DNA-Binding Proteins/biosynthesis , Embryo, Mammalian/physiology , Gene Expression , Genes, Homeobox , Homeodomain Proteins , Transcription Factors/biosynthesis , X Chromosome , Animals , Cell Differentiation/drug effects , Cricetinae , Cricetulus , DNA-Binding Proteins/genetics , Embryo, Mammalian/cytology , Embryonic and Fetal Development , Hybrid Cells , Immunohistochemistry , In Situ Hybridization , Mice , Oligonucleotide Probes , Oligonucleotides, Antisense , Recombination, Genetic , Teratoma , Transcription Factors/genetics , Transcription, Genetic , Tretinoin/pharmacology , Tumor Cells, Cultured
18.
Nature ; 377(6551): 748-50, 1995 Oct 26.
Article in English | MEDLINE | ID: mdl-7477267

ABSTRACT

There are two major angiotensin II receptor isoforms, AT1 and AT2. AT1 mediates the well-known pressor and mitogenic effects of angiotensin II, but the signalling mechanism and physiological role of AT2 has not been established. Its abundant expression in fetal tissues and certain brain nuclei suggest possible roles in growth, development and neuronal functions. Here we report the unexpected finding that the targeted disruption of the mouse AT2 gene resulted in a significant increase in blood pressure and increased sensitivity to the pressor action of angiotensin II. Thus AT2 mediates a depressor effect and antagonizes the AT1-mediated pressor action of angiotensin II. In addition, disruption of the AT2 gene attenuated exploratory behaviour and lowered body temperature. Our results show that angiotensin II activates AT1 and AT2, which have mutually counteracting haemodynamic effects, and that AT2 regulates central nervous system functions, including behaviour.


Subject(s)
Blood Pressure/physiology , Exploratory Behavior/physiology , Receptors, Angiotensin/physiology , Angiotensin II/physiology , Animals , Body Temperature , Cell Line , Crosses, Genetic , Female , Gene Targeting , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Receptors, Angiotensin/deficiency , Receptors, Angiotensin/genetics , Vasoconstrictor Agents
19.
Development ; 126(12): 2631-42, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10331975

ABSTRACT

Mammalian Tolloid-like 1 (mTLL-1) is an astacin-like metalloprotease, highly similar in domain structure to the morphogenetically important proteases bone morphogenetic protein-1 (BMP-1) and Drosophila Tolloid. To investigate possible roles for mTLL-1 in mammalian development, we have used gene targeting in ES cells to produce mice with a disrupted allele for the corresponding gene, Tll1. Homozygous mutants were embryonic lethal, with death at mid-gestation from cardiac failure and a unique constellation of developmental defects that were apparently confined solely to the heart. Constant features were incomplete formation of the muscular interventricular septum and an abnormal and novel positioning of the heart and aorta. Consistent with roles in cardiac development, Tll1 expression was specific to precardiac tissue and endocardium in 7.5 and 8.5 days p.c. embryos, respectively. Tll1 expression was also high in the developing interventricular septum, where expression of the BMP-1 gene, Bmp1, was not observed. Cardiac structures that were not affected in Tll1-/- embryos either showed no Tll1 expression (atrio-ventricular cushions) or showed overlapping expression of Tll1 and Bmp1 (aortico-pulmonary septum), suggesting that products of the Bmp1 gene may be capable of functionally substituting for mTLL-1 at sites in which they are co-expressed. Together, the various data show that mTLL-1 plays multiple roles in formation of the mammalian heart and is essential for formation of the interventricular septum.


Subject(s)
Bone Morphogenetic Proteins , Heart Defects, Congenital/genetics , Heart/embryology , Proteins/genetics , Animals , Bone Morphogenetic Protein 1 , Female , Fetal Death/genetics , Gene Expression Regulation, Developmental , Heart Septal Defects, Atrial/genetics , Heart Septal Defects, Ventricular/genetics , Heart Ventricles/abnormalities , Homozygote , Male , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Metalloproteases , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Myocardium/metabolism , Phenotype , Proteins/metabolism , Recombination, Genetic , Tolloid-Like Metalloproteinases
20.
Development ; 122(3): 983-95, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8631275

ABSTRACT

It has been proposed that the Xenopus homeobox gene, XlHbox8, is involved in endodermal differentiation during pancreatic and duodenal development (Wright, C.V.E., Schnegelsberg, P. and De Robertis, E.M. (1988). Development 105, 787-794). To test this hypothesis directly, gene targeting was used to make two different null mutations in the mouse XlHbox8 homolog, pdx-1. In the first, the second pdx-1 exon, including the homeobox, was replaced by a neomycin resistance cassette. In the second, a lacZ reporter was fused in-frame with the N terminus of PDX-1, replacing most of the homeodomain. Neonatal pdx-1 -/- mice are apancreatic, in confirmation of previous reports (Jonsson, J., Carlsson, L., Edlund, T. and Edlund, H. (1994). Nature 371, 606-609). However, the pancreatic buds do form in homozygous mutants, and the dorsal bud undergoes limited proliferation and outgrowth to form a small, irregularly branched, ductular tree. This outgrowth does not contain insulin or amylase-positive cells, but glucagon-expressing cells are found. The rostral duodenum shows a local absence of the normal columnar epithelial lining, villi, and Brunner's glands, which are replaced by a GLUT2-positive cuboidal epithelium resembling the bile duct lining. Just distal of the abnormal epithelium, the numbers of enteroendocrine cells in the villi are greatly reduced. The PDX-1/beta-galactosidase fusion allele is expressed in pancreatic and duodenal cells in the absence of functional PDX-1, with expression continuing into perinatal stages with similar boundaries and expression levels. These results offer additional insight into the role of pdx-1 in the determination and differentiation of the posterior foregut, particularly regarding the proliferation and differentiation of the pancreatic progenitors.


Subject(s)
Duodenum/embryology , Genes, Homeobox , Homeodomain Proteins , Pancreas/embryology , Trans-Activators/physiology , Animals , Cholecystokinin/metabolism , Endoderm , Gene Expression Regulation, Developmental , Glucose Transporter Type 2 , Heterozygote , Mice , Mice, Inbred C57BL , Mice, Knockout , Monosaccharide Transport Proteins/genetics , RNA, Messenger/genetics , Secretin/metabolism , Serotonin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL