Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nat Med ; 23(10): 1191-1202, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28892064

ABSTRACT

Embryonal tumors with multilayered rosettes (ETMRs) have recently been described as a new entity of rare pediatric brain tumors with a fatal outcome. We show here that ETMRs are characterized by a parallel activation of Shh and Wnt signaling. Co-activation of these pathways in mouse neural precursors is sufficient to induce ETMR-like tumors in vivo that resemble their human counterparts on the basis of histology and global gene-expression analyses, and that point to apical radial glia cells as the possible tumor cell of origin. Overexpression of LIN28A, which is a hallmark of human ETMRs, augments Sonic-hedgehog (Shh) and Wnt signaling in these precursor cells through the downregulation of let7-miRNA, and LIN28A/let7a interaction with the Shh pathway was detected at the level of Gli mRNA. Finally, human ETMR cells that were transplanted into immunocompromised host mice were responsive to the SHH inhibitor arsenic trioxide (ATO). Our work provides a novel mouse model in which to study this tumor type, demonstrates the driving role of Wnt and Shh activation in the growth of ETMRs and proposes downstream inhibition of Shh signaling as a therapeutic option for patients with ETMRs.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenicals/pharmacology , Brain Neoplasms/genetics , Hedgehog Proteins/genetics , Neoplasms, Germ Cell and Embryonal/genetics , Oxides/pharmacology , Wnt Signaling Pathway/genetics , Animals , Arsenic Trioxide , Blotting, Western , Brain Neoplasms/metabolism , Cell Line, Tumor , Disease Models, Animal , Down-Regulation , Gene Expression Profiling , Hedgehog Proteins/antagonists & inhibitors , Humans , Immunohistochemistry , Mice , Mice, Transgenic , MicroRNAs/genetics , Neoplasms, Germ Cell and Embryonal/metabolism , RNA-Binding Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Xenograft Model Antitumor Assays , Zinc Finger Protein GLI1/genetics
2.
Aging Cell ; 15(1): 111-7, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26507795

ABSTRACT

Impaired growth is often associated with an extension of lifespan. However, the negative correlation between somatic growth and life expectancy is only true within, but not between, species. This can be observed because smaller species have, as a rule, a shorter lifespan than larger species. In insects and worms, reduced reproductive development and increased fat storage are associated with prolonged lifespan. However, in mammals the relationship between the dynamics of reproductive development, fat metabolism, growth rate, and lifespan are less clear. To address this point, female transgenic mice that were overexpressing similar levels of either intact (D-mice) or mutant insulin-like growth factor-binding protein-2 (IGFBP-2) lacking the Arg-Gly-Asp (RGD) motif (E- mice) were investigated. Both lines of transgenic mice exhibited a similar degree of growth impairment (-9% and -10%) in comparison with wild-type controls (C-mice). While in D-mice, sexual maturation was found to be delayed and life expectancy was significantly increased in comparison with C-mice, these parameters were unaltered in E-mice in spite of their reduced growth rate. These observations indicate that the RGD-domain has a major influence on the pleiotropic effects of IGFBP-2 and suggest that somatic growth and time of sexual maturity or somatic growth and life expectancy are less closely related than thought previously.


Subject(s)
Body Weight/physiology , Insulin-Like Growth Factor Binding Protein 2/metabolism , Life Expectancy , Lipid Metabolism/physiology , Organ Size/physiology , Animals , Body Weight/genetics , Female , Insulin-Like Growth Factor Binding Protein 2/genetics , Lipid Metabolism/genetics , Mice, Inbred C57BL , Mice, Transgenic , Organ Size/genetics , Time Factors
4.
Cardiovasc Toxicol ; 16(1): 90-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25666561

ABSTRACT

The non-nucleoside reverse transcriptase inhibitor efavirenz is a widely prescribed antiretroviral drug used in combined antiretroviral therapy. Despite being an essential and life-saving medication, the required lifelong use of HIV drugs has been associated with a variety of adverse effects, including disturbances in lipid metabolism and increased cardiovascular risk. Efavirenz belongs to those HIV drugs for which cardiovascular and endothelial dysfunctions have been reported. It is here shown that elevated concentrations of efavirenz can inhibit endothelial meshwork formation on extracellular matrix gels by normal and immortalized human umbilical vein cells. This inhibition was associated with an increase in oxidative stress markers, endoplasmic reticulum (ER) stress markers, and autophagy. Induction of ER stress occurred at pharmacologically relevant concentrations of efavirenz and resulted in reduced proliferation and cell viability of endothelial cells, which worsened in the presence of elevated efavirenz concentrations. In combination with the HIV protease inhibitor nelfinavir, both oxidative stress and ER stress became elevated in endothelial cells. These data indicate that pharmacologically relevant concentrations of efavirenz can impair cell viability of endothelial cells and that these effects may be aggravated by either elevated concentrations of efavirenz or by a combined use of efavirenz with other oxidative stress-inducing medications.


Subject(s)
Anti-HIV Agents/toxicity , Autophagy/drug effects , Benzoxazines/toxicity , Endoplasmic Reticulum Stress/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Oxidative Stress/drug effects , Reverse Transcriptase Inhibitors/toxicity , Alkynes , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclopropanes , HIV Protease Inhibitors/toxicity , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Microtubules/drug effects , Microtubules/pathology , Nelfinavir/toxicity
5.
Proc Natl Acad Sci U S A ; 110(52): 21024-9, 2013 Dec 24.
Article in English | MEDLINE | ID: mdl-24297939

ABSTRACT

In recent years protein O-mannosylation has become a focus of attention as a pathomechanism underlying severe congenital muscular dystrophies associated with neuronal migration defects. A key feature of these disorders is the lack of O-mannosyl glycans on α-dystroglycan, resulting in abnormal basement membrane formation. Additional functions of O-mannosylation are still largely unknown. Here, we identify the essential cell-cell adhesion glycoprotein epithelial (E)-cadherin as an O-mannosylated protein and establish a functional link between O-mannosyl glycans and cadherin-mediated cell-cell adhesion. By genetically and pharmacologically blocking protein O-mannosyltransferases, we found that this posttranslational modification is essential for preimplantation development of the mouse embryo. O-mannosylation-deficient embryos failed to proceed from the morula to the blastocyst stage because of defects in the molecular architecture of cell-cell contact sites, including the adherens and tight junctions. Using mass spectrometry, we demonstrate that O-mannosyl glycans are present on E-cadherin, the major cell-adhesion molecule of blastomeres, and present evidence that this modification is generally conserved in cadherins. Further, the use of newly raised antibodies specific for an O-mannosyl-conjugated epitope revealed that these glycans are present on early mouse embryos. Finally, our cell-aggregation assays demonstrated that O-mannosyl glycans are crucial for cadherin-based cell adhesion. Our results redefine the significance of O-mannosylation in humans and other mammals, showing the immense impact of cadherins on normal as well as pathogenic cell behavior.


Subject(s)
Adherens Junctions/metabolism , Cadherins/metabolism , Cell Adhesion/physiology , Embryo, Mammalian/cytology , Embryonic Development/physiology , Mannose/metabolism , Animals , DNA Primers/genetics , Dogs , Embryo, Mammalian/physiology , Fluorescent Antibody Technique , Glycosylation , Madin Darby Canine Kidney Cells , Mass Spectrometry , Mice , Polysaccharides/metabolism
6.
Hum Mol Genet ; 21(3): 473-84, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22010047

ABSTRACT

Pur-alpha (Purα) plays an important role in a variety of cellular processes including transcriptional regulation, cell proliferation and oncogenic transformation. To better understand the role of Purα in the developing and mature brain, we generated Purα-deficient mice, which we were able to raise to the age of six months. Purα(-/-) mice were born with no obvious pathological condition. We obtained convincing evidence that lack of Purα prolongs the postnatal proliferation of neuronal precursor cells both in the hippocampus and in the cerebellum, however, without affecting the overall number of postmitotic neurons. Independent of these findings, we observed alterations in the expression and distribution of the dendritic protein MAP2, the translation of which has been proposed previously to be Purα-dependent. At the age of 2 weeks, Purα(-/-) mice generated a continuous tremor which persisted throughout lifetime. Finally, adult Purα(-/-) mice displayed a megalencephaly and histopathological findings including axonal swellings and hyperphosphorylation of neurofilaments. Our studies underline the importance of Purα in the proliferation of neuronal precursor cells during postnatal brain development and suggest a role for Purα in the regulation of the expression and cellular distribution of dendritic and axonal proteins. Since recent studies implicate a link between Purα and the fragile X tremor/ataxia syndrome, our Purα(-/-) mouse model will provide new opportunities for understanding the mechanisms of neurodegeneration.


Subject(s)
Brain/growth & development , Brain/pathology , DNA-Binding Proteins/physiology , Nerve Tissue Proteins/physiology , Animals , Axons/metabolism , Brain Chemistry , Cell Proliferation , Cerebellum/cytology , Cerebellum/growth & development , Cerebellum/pathology , Cerebrum/growth & development , Cerebrum/pathology , DNA-Binding Proteins/genetics , Hippocampus/cytology , Hippocampus/growth & development , Hypertrophy , Mice , Mice, Knockout , Microtubule-Associated Proteins/analysis , Nerve Tissue Proteins/genetics , Neurofilament Proteins/metabolism , Phosphorylation
7.
Horm Cancer ; 1(6): 320-30, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21761363

ABSTRACT

The insulin analog glargine has a higher binding affinity than regular insulin for the insulin-like growth factor 1 receptor in vitro, raising questions about increased mitogenicity in vivo. Observational studies in humans have recently reported a potential differential association between insulin glargine and malignancies, but available evidence remains inconclusive. We directly compared glargine vs. neutral protamine Hagedorn (NPH) insulin's effects on cell proliferation in colonic mucosa and on formation of aberrant crypt foci in diabetic mice, i.e., early stages of colorectal cancer development. Mice (BKS.Cg-+Lepr(db)/+Lepr(db)/OlaHsd) were treated with insulin glargine (G), NPH insulin (NPH), or saline (NaCl). We assessed epithelial proliferation after long-term insulin treatment (18 weeks) by 5-bromo-2'-deoxyuridine and Ki67 staining and analyzed the formation of aberrant crypt foci (ACF) in mice treated with insulin glargine or NPH insulin or 10 weeks after initiation with 1,2-dimethylhydrazine. Insulin glargine treatment did not result in significantly different epithelial colonic proliferation compared to NPH insulin (G, 137 ± 22; NPH, 136 ± 15; NaCl, 100 ± 20 (relative proliferation index)), but both insulin-treated groups of mice had a higher proliferation index compared to the NaCl control group (p<0.001). Similarly, we observed no difference in ACF formation between glargine- and NPH-insulin-treated mice (G, 132 ± 12; NPH, 138 ± 9; NaCl, 100 ± 7 (relative number of ACF)), but ACF formation was significantly higher in insulin-treated mice than in NaCl-treated control mice (p=0.001). Chronic insulin treatment results in higher colonic epithelial proliferation and ACF formation, but the use of insulin glargine vs. NPH insulin is not associated with increased risk.


Subject(s)
Colon/drug effects , Epithelial Cells/drug effects , Epithelial Cells/pathology , Hypoglycemic Agents/adverse effects , Insulin, Isophane/adverse effects , Insulin, Long-Acting/adverse effects , Animals , Cell Proliferation/drug effects , Colon/pathology , Diabetes Mellitus, Type 2/drug therapy , Female , Insulin Glargine , Mice
8.
Mol Cell Endocrinol ; 315(1-2): 219-24, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-19682540

ABSTRACT

The availability of regulatory sequences directing tissue-specific expression of transgenes in genetically modified mice and large animals is a prerequisite for the development of adequate models for human diseases. The rat insulin 2 gene (Ins2) promoter, widely used to achieve transgene expression in pancreatic beta-cells of mice, also directs expression to extrapancreatic tissues and performs poorly in isolated pancreatic islets of human, mouse, and pig. To evaluate whether the full 5' untranslated region (UTR) of the porcine insulin gene (INS) confers robust and specific expression in beta-cells we generated an expression cassette containing 1500bp of the porcine INS 5' UTR and the 3' UTR of the bovine growth hormone gene (GH). The cassette was designed to allow easy exchange of the sequences to be expressed and easy removal of the vector backbone from the expression cassette. To evaluate the properties of the cassette, we initially inserted a cDNA encoding human betacellulin, a growth factor known to affect structural and functional parameters of beta-cells. After confirming the functionality and specificity of the construct in vitro, transgenic mouse lines were generated by pronuclear DNA microinjection. Using RT-PCR, immunohistochemistry and immunofluorescence, we show that transgenic mice expressed human betacellulin exclusively in beta-cells. Confirming the proposed insulinotropic effect of betacellulin, transgenic mice showed improved glucose tolerance. We conclude that the newly designed expression cassette containing 1500bp of the porcine insulin promoter 5' UTR confers robust and specific transgene expression to beta-cells in vitro and in transgenic mice.


Subject(s)
Gene Expression Regulation , Insulin-Secreting Cells/physiology , Insulin/genetics , Promoter Regions, Genetic , Transgenes , Animals , Base Sequence , Betacellulin , Blood Glucose/metabolism , Cattle , Humans , Insulin-Secreting Cells/cytology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Transgenic , Molecular Sequence Data , Phenotype , Rats , Swine , Tissue Distribution , Untranslated Regions/genetics
9.
J Mol Biol ; 388(4): 721-9, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19324055

ABSTRACT

The 37-kDa/67-kDa laminin receptor (LRP/LR) was identified as a cell surface receptor for prion proteins. The laminin receptor mutant LRP102-295::FLAG interfered with PrP(Sc) propagation in murine neuronal cells presumably acting as a decoy in a transdominant negative fashion by trapping PrP molecules in the extracellular matrix. Here, we generated hemizygous transgenic mice expressing LRP102-295::FLAG in the brain. Scrapie-infected transgenic mice exhibit a significantly prolonged incubation time in comparison to scrapie-infected wild-type (FVB) mice. At the terminal stage, transgenic mice revealed significantly reduced proteinase-K-resistant PrP levels by 71% compared to wild-type mice. Our results recommend the laminin receptor decoy mutant as an alternative therapeutic tool for treatment of transmissible spongiform encephalopathies.


Subject(s)
Mice, Transgenic , Prions/metabolism , Receptors, Laminin , Scrapie/physiopathology , Animals , Brain/metabolism , Brain/pathology , Genotype , Humans , Kaplan-Meier Estimate , Mice , Prions/genetics , Receptors, Laminin/genetics , Receptors, Laminin/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Scrapie/pathology , Spleen/metabolism , Spleen/pathology
10.
Int J Cancer ; 124(9): 2220-5, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19142966

ABSTRACT

Colon cancer patients frequently show increased levels of serum insulin-like growth factor-binding protein-2 (IGFBP-2), however, the pathogenetic relevance of this phenomenon for colorectal cancer is unclear. Therefore, we have used IGFBP-2 transgenic animals which overexpress IGFBP-2 systemically and locally in the intestine to study its role in chemically induced colorectal carcinogenesis. Mice received intraperitoneal injections of 1,2-dimethylhydrazine (DMH) (40 mg/kg body weight) once a week for 6 weeks to selectively induce aberrant crypt foci (ACF) and tumors in the colon. While tumor incidence was comparable in transgenic and control mice, the volume of adenomas in IGFBP-2 transgenic mice was reduced more than 2-fold. Furthermore, serum IGFBP-2 levels negatively correlated with tumor volume in the IGFBP-2 transgenic group. Histological examination showed that IGFBP-2 transgenic mice developed significantly less dysplastic ACF with a high potential to progress to advanced stages. The reduced tumor volume in IGFBP-2 transgenic animals was due to significantly reduced proliferative capacity, evidenced by a lower proportion of cells positive for Ki67. Our results demonstrate for the first time in an experimental model that IGFBP-2 overabundance prior to the onset and during colorectal carcinogenesis reduces tumor growth by inhibition of cell proliferation.


Subject(s)
Adenoma/metabolism , Adenoma/prevention & control , Colonic Neoplasms/metabolism , Colonic Neoplasms/prevention & control , Insulin-Like Growth Factor Binding Protein 2/metabolism , 1,2-Dimethylhydrazine/toxicity , Adenoma/chemically induced , Animals , Body Weight , Carcinogens/toxicity , Cell Proliferation , Cells, Cultured , Colonic Neoplasms/chemically induced , Female , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
11.
Transgenic Res ; 17(4): 479-88, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18097769

ABSTRACT

Transgenic mice overexpressing growth hormone (GH) display a plethora of phenotypic alterations and provide unique models for studying and influencing consequences of chronic GH excess. Since the first report on GH transgenic mice was published in 1982, many different mouse models overexpressing GH from various species at different levels and with different tissue specificities were established, most of them on random-bred or hybrid genetic background. We have generated a new transgenic mouse model on FVB/N inbred background, expressing bovine (b) GH under the control of the chicken beta-actin promoter (cbetaa). cbetaa-bGH transgenic mice exhibit ubiquitous expression of bGH mRNA and protein and circulating bGH levels in the range of several microg/ml, resulting in markedly stimulated growth and the characteristic spectrum of pathological lesions which were described in previous GH overexpressing mouse models. Importantly, a consistent sequence of renal alterations is observed, mimicking progressive kidney disease in human patients. The novel, genetically standardized GH transgenic mouse model is ideal for holistic transcriptome and proteome studies aiming at the identification of the molecular mechanisms underlying GH-induced pathological alterations especially in the kidney. Moreover, genetically defined cbetaa-bGH mice facilitate random mutagenesis screens for modifier genes which influence the effects of chronic GH excess and associated pathological lesions.


Subject(s)
Growth Hormone/physiology , Kidney Diseases/etiology , Kidney Diseases/pathology , Animals , Blotting, Western , Body Weight , Cattle , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression , Male , Mice , Mice, Transgenic , Organ Size , Plasmids , Promoter Regions, Genetic
12.
Biochem J ; 409(3): 741-9, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17961124

ABSTRACT

SePP (selenoprotein P) is central for selenium transport and distribution. Targeted inactivation of the Sepp gene in mice leads to reduced selenium content in plasma, kidney, testis and brain. Accordingly, activities of selenoenzymes are reduced in Sepp(-/-) organs. Male Sepp(-/-) mice are infertile. Unlike selenium deficiency, Sepp deficiency leads to neurological impairment with ataxia and seizures. Hepatocyte-specific inactivation of selenoprotein biosynthesis reduces plasma and kidney selenium levels similarly to Sepp(-/-) mice, but does not result in neurological impairment, suggesting a physiological role of locally expressed SePP in the brain. In an attempt to define the role of liver-derived circulating SePP in contrast with locally expressed SePP, we generated Sepp(-/-) mice with transgenic expression of human SePP under control of a hepatocyte-specific transthyretin promoter. Secreted human SePP was immunologically detectable in serum from SEPP1-transgenic mice. Selenium content and selenoenzyme activities in serum, kidney, testis and brain of Sepp(-/-;SEPP1) (SEPP1-transgenic Sepp(-/-)) mice were increased compared with Sepp(-/-) controls. When a selenium-adequate diet (0.16-0.2 mg/kg of body weight) was fed to the mice, liver-specific expression of SEPP1 rescued the neurological defects of Sepp(-/-) mice and rendered Sepp(-/-) males fertile. When fed on a low-selenium diet (0.06 mg/kg of body weight), Sepp(-/-;SEPP1) mice survived 4 weeks longer than Sepp(-/-) mice, but ultimately developed the neurodegenerative phenotype. These results indicate that plasma SePP derived from hepatocytes is the main transport form of selenium supporting the kidney, testis and brain. Nevertheless, local Sepp expression is required to maintain selenium content in selenium-privileged tissues such as brain and testis during dietary selenium restriction.


Subject(s)
Gene Expression Regulation , Hepatocytes/metabolism , Infertility, Male/metabolism , Motor Activity , Selenium/metabolism , Selenoprotein P/deficiency , Selenoprotein P/metabolism , Animals , Biological Transport , Humans , Infertility, Male/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Organ Specificity , Phenotype , Selenoprotein P/genetics , Spermatozoa/metabolism
13.
Biochem Biophys Res Commun ; 361(3): 574-9, 2007 Sep 28.
Article in English | MEDLINE | ID: mdl-17669363

ABSTRACT

Apolipoprotein E (ApoE) plays an important role in the development of atherosclerosis. Previous studies provide evidence for an atheroprotective role of ApoE in mouse models on the ApoE deficient (ApoE-/-) background. However, it is not clear whether this is also true on the LDL-receptor deficient (LDLR-/-) background. Transgenic mice carrying hApoE coding sequences in a chicken lysozyme expression cassette were generated. Transgene expression was directed into macrophages, expressing low levels of hApoE. Expression of the hApoE transgene was not sufficient to correct hypercholesterolemia. However, lesion area at the brachiocephalic artery (BCA) was significantly reduced (-72%) in female hApoE transgenic mice on the LDLR-/- background. This was associated with increased cholesterol efflux in macrophages of transgenic animals on the ApoE-/- background. We conclude that over-expression of ApoE in macrophages might be useful as a therapeutic principle for the prevention of atherosclerosis.


Subject(s)
Apolipoproteins E/metabolism , Arteriosclerosis/metabolism , Macrophages/metabolism , Receptors, LDL/genetics , Animals , Apolipoproteins E/blood , Apolipoproteins E/genetics , Arteriosclerosis/etiology , Cells, Cultured , Cholesterol/blood , Cholesterol/metabolism , Female , Humans , Mice , Mice, Knockout , Mice, Transgenic , RNA, Messenger/metabolism , Receptors, LDL/metabolism
14.
Stem Cells Dev ; 14(4): 402-7, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16137229

ABSTRACT

Isolated and expanded scl (+) adult murine progenitors show a strong endothelial and hematopoietic differentiation potential and have been considered to be the adult equivalent of the hemangioblast. These unique cells may provide effective therapeutic approaches to tissue damage resulting from hypoxemia or chronic ischemia. Here, we study the fate of adult scl (+/+) during development and their ability to reverse genetic defects in scl expression. scl (+/+) adult stem cells (clone RM26) did not persist during embryonic development after injection into blastocysts of allogeneic wild-type mice on day E 3.5. However, GFP(+)-marked scl (+/+) cells were detected in all possible genotypes from allogeneic scl (+/+) intercrosses (scl (+/+), scl (+/-), scl (-/-) on day E 9.5 after the cloned cells were injected into scl-mutant blastocysts on day E 3.5. Nevertheless, there was no indication of phenotypic rescue of the mutant blastocysts despite the continued presence of scl (+/+) RM26 cells in the allogeneic embryonic environment. The results show that differentiated stem cells providing a defective gene may exert effects during development when there is a reparative demand, but they are not capable of reversing the effects of a mutant phenotype during embryonic development. These effects should be considered when evaluating the efficacy of stem cells for therapeutic reversal of inborn errors of development.


Subject(s)
Blastocyst/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Mutation , Neovascularization, Physiologic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Transcription Factors/genetics , Transcription Factors/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors , Blood Cells/cytology , Cell Differentiation , DNA/metabolism , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/metabolism , Hypoxia , Ischemia , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Phenotype , Stem Cells/cytology , T-Cell Acute Lymphocytic Leukemia Protein 1 , Time Factors , Transplantation, Homologous
15.
Transgenic Res ; 14(2): 145-58, 2005 Apr.
Article in English | MEDLINE | ID: mdl-16022386

ABSTRACT

The ability of a 470 bp sub-fragment of the murine whey acidic protein (WAP) promoter in the context of a retroviral expression plasmid to direct gene expression to mammary epithelial cells was analysed in a number of independent transgenic mouse lines. In contrast to previous findings with the genuine 2.5 kb promoter fragment, our studies revealed a highly mammary gland-specific expression detectable only in non-lactating animals. This suggested a mainly progesterone-regulated activity of the short fragment. Therefore, transgene expression was examined in the progesterone-determined estrous cycle and during pregnancy. In accordance with in vitro data from stably transfected cell lines, in both situations expression was upregulated at stages associated with high progesterone levels. Taken together these data provide deeper insight into WAP-promoter regulation and stress the usefulness of the shortened fragment for a lactation independent mammary-targeted expression.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma/genetics , Carcinoma/pathology , Gene Expression Regulation , Mammary Glands, Animal/physiology , Milk Proteins/genetics , Animals , Epithelial Cells/physiology , Estrus/physiology , Female , Genetic Vectors , Lactation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Progesterone/physiology , Promoter Regions, Genetic , Retroviridae , Tumor Cells, Cultured
16.
Proc Natl Acad Sci U S A ; 101(39): 14126-31, 2004 Sep 28.
Article in English | MEDLINE | ID: mdl-15383666

ABSTRACT

O-mannosylation is an important protein modification in eukaryotes that is initiated by an evolutionarily conserved family of protein O-mannosyltransferases. The first mammalian protein O-mannosyltransferase gene described was the human POMT1. Mutations in the hPOMT1 gene are responsible for Walker-Warburg syndrome (WWS), a severe recessive congenital muscular dystrophy associated with defects in neuronal migration that produce complex brain and eye abnormalities. During embryogenesis, the murine Pomt1 gene is prominently expressed in the neural tube, the developing eye, and the mesenchyme. These sites of expression correlate with those in which the main tissue alterations are observed in WWS patients. We have inactivated a Pomt1 allele by gene targeting in embryonic stem cells and produced chimeras transmitting the defect allele to offspring. Although heterozygous mice were viable and fertile, the total absence of Pomt1(-/-) pups in the progeny of heterozygous intercrosses indicated that this genotype is embryonic lethal. An analysis of the mutant phenotype revealed that homozygous Pomt1(-/-) mice suffer developmental arrest around embryonic day (E) 7.5 and die between E7.5 and E9.5. The Pomt1(-/-) embryos present defects in the formation of Reichert's membrane, the first basement membrane to form in the embryo. The failure of this membrane to form appears to be the result of abnormal glycosylation and maturation of dystroglycan that may impair recruitment of laminin, a structural component required for the formation of Reichert's membrane in rodents. The targeted disruption of mPomt1 represents an example of an engineered deletion of a known glycosyltransferase involved in O-mannosyl glycan synthesis.


Subject(s)
Abnormalities, Multiple/embryology , Abnormalities, Multiple/genetics , Fetal Death/genetics , Mannosyltransferases/genetics , Abnormalities, Multiple/enzymology , Animals , Base Sequence , Brain/abnormalities , Brain/embryology , Extracellular Matrix/physiology , Eye Abnormalities/genetics , Female , Fetal Death/embryology , Gene Expression/physiology , Gene Targeting , Glycosylation , Hematoxylin/metabolism , Humans , Immunohistochemistry , In Situ Hybridization , Laminin/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , Pregnancy , Recombination, Genetic , Syndrome
17.
Transgenic Res ; 13(1): 81-5, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15070079

ABSTRACT

The 37-kDa/67-kDa laminin receptor (LRP/LR) plays a major role in the propagation of PrPSc, the abnormal form of the prion protein. In order to ablate the expression of LRP/LR in mouse brain we generated transgenic mice ectopically expressing antisense LRP RNA in the brain under control of the neuron-specific enolase (NSE) promoter. Hemizygous transgenic mice TgN(NSEasLRP)2 showed a significant reduction of LRP/LR protein levels in hippocampal and cerebellar brain regions. These mice might act as powerful tools to investigate the role of the laminin receptor in scrapie pathogenesis.


Subject(s)
Brain/metabolism , Gene Expression , Mice/genetics , RNA, Antisense/metabolism , Receptors, Laminin/metabolism , Animals , Cell Line , Mice, Transgenic , Molecular Weight , Phosphopyruvate Hydratase/genetics , Promoter Regions, Genetic , Rats , Receptors, Laminin/genetics
18.
J Biol Chem ; 278(20): 18524-31, 2003 May 16.
Article in English | MEDLINE | ID: mdl-12637572

ABSTRACT

Prion diseases are neurodegenerative infectious disorders for which no prophylactic regimens are known. In order to induce antibodies/auto-antibodies directed against surface-located PrP(c), we used a covalently linked dimer of mouse prion protein expressed recombinantly in Escherichia coli. Employing dimeric PrP as an immunogen we were able to effectively overcome autotolerance against murine PrP in PrP wild-type mice without inducing obvious side effects. Treatment of prion-infected mouse cells with polyclonal anti-PrP antibodies generated in rabbit or auto-antibodies produced in mice significantly inhibited endogenous PrP(Sc) synthesis. We show that polyclonal antibodies are binding to surface-located PrP(c), thereby interfering with prion biogenesis. This effect is much more pronounced in the presence of full IgG molecules, which, unlike Fab fragments, seem to induce a significant cross-linking of surface PrP. In addition, we found immune responses against different epitopes when comparing antibodies induced in rabbits and PrP wild-type mice. Only in the auto-antibody situation in mice an immune reaction against a region of PrP is found that was reported to be involved in the PrP(Sc) conversion process. Our data point to the possibility of developing means for an active immunoprophylaxis against prion diseases.


Subject(s)
Prions/immunology , Animals , Cells, Cultured , Dimerization , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes , Escherichia coli/metabolism , Female , Immunoblotting , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Mice , Mice, Inbred C57BL , Precipitin Tests , Prions/chemistry , Prions/metabolism , Protein Binding , Rabbits , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
19.
FASEB J ; 17(2): 247-9, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12475886

ABSTRACT

Megalin is an endocytic receptor highly expressed in the proximal tubules of the kidney. Recently, we demonstrated that this receptor is essential for the renal uptake and conversion of 25-OH vitamin D3 to 1,25-(OH)2 vitamin D3, a central step in vitamin D and bone metabolism. Unfortunately, the perinatal lethality of the conventional megalin knockout mouse model precluded the detailed analysis of the significance of megalin for calcium homeostasis and bone turnover in vivo. Here, we have generated a new mouse model with conditional inactivation of the megalin gene in the kidney by using Cre recombinase. Animals with a renal-specific receptor gene defect were viable and fertile. However, lack of receptor expression in the kidney results in plasma vitamin D deficiency, in hypocalcemia and in severe bone disease, characterized by a decrease in bone mineral content, an increase in osteoid surfaces, and a lack of mineralizing activity. These features are consistent with osteomalacia (softening of the bones) as a consequence of hypovitaminosis D and demonstrate the crucial importance of the megalin pathway for systemic calcium homeostasis and bone metabolism.


Subject(s)
Hypocalcemia/metabolism , Kidney/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Osteomalacia/metabolism , Animals , Apolipoproteins E/genetics , Genotype , Humans , Hypocalcemia/genetics , Integrases/genetics , Integrases/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Osteomalacia/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Vitamin D/metabolism , Vitamin D-Binding Protein/blood , Vitamin D-Binding Protein/urine
SELECTION OF CITATIONS
SEARCH DETAIL
...