Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
J Cell Biol ; 152(1): 87-96, 2001 Jan 08.
Article in English | MEDLINE | ID: mdl-11149923

ABSTRACT

Wnt signaling plays a critical role in development and oncogenesis. Although significant progress has been made in understanding the downstream signaling cascade of Wnt signaling, little is known regarding Wnt signaling modification of the cell death machinery. Given that numerous oncogenes transform cells by providing cell survival function, we hypothesized that Wnt signaling may inhibit apoptosis. Here, we report that cells expressing Wnt-1 were resistant to cancer therapy-mediated apoptosis. Wnt-1 signaling inhibited the cytochrome c release and the subsequent caspase-9 activation induced by chemotherapeutic drugs, including both vincristine and vinblastine. Furthermore, we found that Wnt-1-mediated cell survival was dependent on the activation of beta-catenin/T cell factor (Tcf) transcription. Inhibition of beta-catenin/Tcf transcription by expression of the dominant-negative mutant of Tcf-4 blocked Wnt-1-mediated cell survival and rendered cells sensitive to apoptotic stimuli. These results provide the first demonstration that Wnt-1 inhibits cancer therapy-mediated apoptosis and suggests that Wnt-1 may exhibit its oncogenic potential through a mechanism of anti-apoptosis.


Subject(s)
Apoptosis , Cytoskeletal Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction/physiology , Trans-Activators , Transcription Factors/metabolism , Transcriptional Activation , Zebrafish Proteins , Animals , Caspase 9 , Caspases/metabolism , Cell Line , Cell Survival , Colorectal Neoplasms , Cytochrome c Group/metabolism , Enzyme Activation , Humans , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Rats , TCF Transcription Factors , Transcription Factor 7-Like 2 Protein , Transcription, Genetic , Tumor Cells, Cultured , Wnt Proteins , Wnt1 Protein , beta Catenin
2.
Br J Cancer ; 99(8): 1204-9, 2008 Oct 21.
Article in English | MEDLINE | ID: mdl-18827808

ABSTRACT

Tumour angiogenesis has become an important target for antitumour therapy, with most current therapies aimed at blocking the VEGF pathway. However, not all tumours are responsive to VEGF blockers, and some tumours that are responsive initially may become resistant during the course of treatment, thus there is a need to explore other angiogenesis signalling pathways. Recently, the Delta-Notch pathway, and particularly the ligand Delta-like 4 (Dll4), was identified as a new target in tumour angiogenesis. An important feature in angiogenesis is the manifold ways in which the VEGF and Delta-Notch pathways interact. The emerging picture is that the VEGF pathway acts as a potent upstream activating stimulus for angiogenesis, whereas Delta-Notch helps to guide cell fate decisions that appropriately shape the activation. Here we review the two signalling pathways and what is currently known about the ways in which they interact during tumour angiogenesis.


Subject(s)
Membrane Proteins/metabolism , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Humans , Intracellular Signaling Peptides and Proteins
3.
Oncogene ; 25(24): 3436-44, 2006 Jun 08.
Article in English | MEDLINE | ID: mdl-16474850

ABSTRACT

beta-Catenin, a component of the Wnt signaling pathway, is a coactivator of human androgen receptor (hAR) transcriptional activity. Here, we show that Wnt signaling also influences androgen-mediated signaling through its ability to regulate hAR mRNA and protein in prostate cancer (PCa) cells. Three functional LEF-1/TCF binding sites lie within the promoter of the hAR gene as shown by CHIP assays that captured beta-catenin-bound chromatin from Wnt-activated LNCaP cells. Chimeric reporter vectors that use the hAR gene promoter to drive luciferase expression confirmed that these LEF-1/TCF binding elements are able to confer robust upregulation of luciferase expression when stimulated by Wnt-1 or by transfection with beta-catenin and that dominant-negative TCF or mutations within the dominant TCF-binding element abrogated the response. Semi-quantitative and real time RT-PCR assays confirmed that Wnt activation upregulates hAR mRNA in PCa cells. In contrast, hAR protein expression was strongly suppressed by Wnt activation. The reduction of hAR protein is consistent with evidence that Wnt signaling increased phosphorylation of Akt and its downstream target, MDM2 that promotes degradation of hAR protein through a proteasomal pathway. These data indicate that the hAR gene is a direct target of LEF-1/TCF transcriptional regulation in PCa cells but also show that the expression of the hAR protein is suppressed by a degradation pathway regulated by cross-talk of Wnt to Akt that is likely mediated by Wnt-directed degradation of the B regulatory subunit of protein phosphatase, PP2A.


Subject(s)
Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/metabolism , Receptors, Androgen/biosynthesis , Receptors, Androgen/genetics , Wnt Proteins/metabolism , Binding Sites , Cell Line, Tumor , Humans , Male , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Prostatic Neoplasms/genetics , Protein Phosphatase 2 , Proto-Oncogene Proteins c-mdm2/metabolism , Receptors, Androgen/metabolism , Signal Transduction
4.
Mol Cell Biol ; 18(5): 2474-85, 1998 May.
Article in English | MEDLINE | ID: mdl-9566868

ABSTRACT

Genetic evidence suggests that regulation of beta-catenin and regulation of Tcf/Lef family transcription factors are downstream events of the Wnt signal transduction pathway. However, a direct link between Wnt activity and Tcf/Lef transcriptional activation has yet to be established. In this study, we show that Wnt-1 induces a growth response in a cultured mammalian cell line, Rat-1 fibroblasts. Wnt-1 induces serum-independent cellular proliferation of Rat-1 fibroblasts and changes in morphology. Rat-1 cells stably expressing Wnt-1 (Rat-1/Wnt-1) show a constitutive up-regulation of cytosolic beta-catenin, while membrane-associated beta-catenin remains unaffected. Induction of cytosolic beta-catenin in Rat-1/Wnt-1 cells is correlated with activation of a Tcf-responsive transcriptional element. We thus provide evidence that Wnt-1 induces Tcf/Lef transcriptional activation in a mammalian system. Expression of a mutant beta-catenin (beta-CatS37A) in Rat-1 cells does not result in a proliferative response or a detectable change in the cytosolic beta-catenin protein level. However, beta-CatS37A expression in Rat-1 cells results in strong Tcf/Lef transcriptional activation, comparable to that seen in Wnt-1-expressing cells. These results suggest that Wnt-1 induction of cytosolic beta-catenin may have functions in addition to Tcf/Lef transcriptional activation.


Subject(s)
Cytoskeletal Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators , Transcription Factors/biosynthesis , Zebrafish Proteins , Animals , Cell Cycle , Cells, Cultured , Culture Media, Serum-Free , Fibroblasts/cytology , Gene Expression Regulation, Neoplastic , Rats , Signal Transduction , Transcription Factors/genetics , Transcription, Genetic , Wnt Proteins , Wnt1 Protein , beta Catenin
5.
Mol Cell Biol ; 6(12): 4493-8, 1986 Dec.
Article in English | MEDLINE | ID: mdl-3796608

ABSTRACT

The VAI RNA of adenovirus is a small, RNA polymerase III-transcribed species required for the efficient translation of host cell and viral mRNAs late after infection. VAI RNA prevented activation of the interferon-induced P1/eIF-2 alpha kinase. In its absence the kinase was activated, eIF-2 alpha was phosphorylated, and translational initiation was inhibited. H5dl331 (dl331), a mutant which cannot express VAI RNA, grew poorly in 293 cells but generated wild-type yields in KB cells. The growth phenotype of the mutant appeared to correlate with the kinetics of kinase induction and activation. Active kinase appeared more rapidly in cell extracts prepared from infected 293 cells, in which dl331 grew poorly, than in extracts of KB cells, in which the mutant grew well. However, when kinase was induced in KB cells by interferon treatment and then activated subsequent to dl331 infection, viral protein synthesis was less severely inhibited than in interferon-treated 293 cells. Thus, activated kinase per se is insufficient to severely inhibit dl331 protein synthesis in KB cells.


Subject(s)
Adenoviruses, Human/genetics , Interferon Type I/pharmacology , Mutation , Adenoviruses, Human/drug effects , Adenoviruses, Human/growth & development , Cell Line , Eukaryotic Initiation Factor-2 , HeLa Cells/microbiology , Humans , KB Cells/microbiology , Kidney/embryology , Kinetics , Peptide Initiation Factors/metabolism , Protein Biosynthesis , Proteins/metabolism , RNA Polymerase III/metabolism
6.
Mol Cell Biol ; 12(2): 784-90, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1531088

ABSTRACT

The mouse Wnt-1 gene, a target for insertional activation in mouse mammary tumor virus-induced mammary tumors, encodes poorly secreted, cysteine-rich glycoproteins required for proper central nervous system development. We have been analyzing the biosynthesis of Wnt-1 proteins in several cell lines that express Wnt-1 cDNA from heterologous promoters. A protein of 78 kDa was found to be associated with the intracellular forms of Wnt-1 proteins in mammalian and avian cells by using multiple antisera against Wnt-1 proteins. We have identified p78 as the binding protein BiP with anti-BiP antisera and by its release from Wnt-1 immunoprecipitates upon incubation with MgCl2 and ATP. Experiments with a Wnt-1 mutant that lacks the sequence encoding the signal peptide indicates that Wnt-1 proteins must enter the secretory pathway in order to interact with BiP. We demonstrate that Wnt-1 proteins are associated with BiP in cells in which active Wnt-1 proteins are produced, such as a cultured mammary epithelial cell line and Wnt-1 transgenic mouse mammary tumor cells. The association of Wnt-1 proteins with BiP may be a factor in determining the efficiency of secretion of Wnt-1 gene products.


Subject(s)
Fungal Proteins/metabolism , HSP70 Heat-Shock Proteins , Heat-Shock Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Zebrafish Proteins , Adenosine Triphosphate/metabolism , Cell Line, Transformed , Precipitin Tests , Protein Sorting Signals/metabolism , Wnt Proteins , Wnt1 Protein
7.
Mol Cell Biol ; 21(21): 7403-15, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11585921

ABSTRACT

Notch receptors and their ligands play important roles in both normal animal development and pathogenesis. We show here that the F-box/WD40 repeat protein SEL-10 negatively regulates Notch receptor activity by targeting the intracellular domain of Notch receptors for ubiquitin-mediated protein degradation. Blocking of endogenous SEL-10 activity was done by expression of a dominant-negative form containing only the WD40 repeats. In the case of Notch1, this block leads to an increase in Notch signaling stimulated by either an activated form of the Notch1 receptor or Jagged1-induced signaling through Notch1. Expression of dominant-negative SEL-10 leads to stabilization of the intracellular domain of Notch1. The Notch4 intracellular domain bound to SEL-10, but its activity was not increased as a result of dominant-negative SEL-10 expression. SEL-10 bound Notch4 via the WD40 repeats and bound preferentially to a phosphorylated form of Notch4 in cells. We mapped the region of Notch4 essential for SEL-10 binding to the C-terminal region downstream of the ankyrin repeats. When this C-terminal fragment of Notch4 was expressed in cells, it was highly labile but could be stabilized by the expression of dominant-negative SEL-10. Ubiquitination of Notch1 and Notch4 intracellular domains in vitro was dependent on SEL-10. Although SEL-10 interacts with the intracellular domains of both Notch1 and Notch4, these proteins respond differently to interference with SEL-10 function. Thus, SEL-10 functions to promote the ubiquitination of Notch proteins; however, the fates of these proteins may differ.


Subject(s)
Caenorhabditis elegans Proteins , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , Helminth Proteins/metabolism , Helminth Proteins/physiology , Membrane Proteins/metabolism , Signal Transduction , Ubiquitin/metabolism , Animals , Blotting, Western , Cell Line , Cysteine Endopeptidases , Dose-Response Relationship, Drug , Gene Deletion , Genes, Dominant , Genetic Vectors , Humans , Insecta , Ligands , Luciferases/metabolism , Models, Genetic , Multienzyme Complexes/antagonists & inhibitors , Phosphorylation , Plasmids/metabolism , Precipitin Tests , Promoter Regions, Genetic , Proteasome Endopeptidase Complex , Protein Binding , Protein Structure, Tertiary , Proteins/metabolism , Receptors, Notch
8.
Mol Biol Cell ; 3(5): 521-33, 1992 May.
Article in English | MEDLINE | ID: mdl-1535241

ABSTRACT

The proto-oncogene Wnt-1 encodes a cysteine-rich, secretory glycoprotein implicated in virus-induced mouse mammary cancer and intercellular signaling during vertebrate neural development. To attempt to correlate structural motifs of Wnt-1 protein with its function, 12 mutations were introduced singly and in several combinations into the coding sequence of Wnt-1 cDNA by site-directed mutagenesis. Mutant alleles in a retroviral vector were tested for their ability to transform the mouse mammary epithelial cell line C57MG in two ways: by direct infection of C57MG cells and by infection of NIH3T3 cells that serve as donors of Wnt-1 protein to adjacent C57MG cells in a secretion-dependent (paracrine) assay. In addition, the synthesis and secretion of mutant proteins were monitored in multiple cell types by immunological assays. Deletion of the signal peptide demonstrated that transformation in both direct and paracrine assays depends upon entry of Wnt-1 protein into the endoplasmic reticulum. Changes in potential proteolytic processing sites (two basic dipeptides and a probable signal peptidase cleavage site) did not adversely impair biological activity or protein processing and uncovered a second site for cleavage by signal peptidase. Replacement of each of the four asparagine-linked glycosylation sites did not affect transforming activity at normal temperatures, but one glycosylation site mutant was found to be temperature-sensitive for transformation. An allele encoding a protein that lacks all four glycosylation sites was also transformation competent. In two of four cases, substitution of serine for a cysteine residue impaired transforming activity at the usual temperature, and transformation was temperature sensitive in a third case, implying that at least some of the highly conserved cysteine residues are important for Wnt-1 function.


Subject(s)
Proto-Oncogene Proteins/physiology , Transformation, Genetic/physiology , Zebrafish Proteins , Alleles , Animals , Base Sequence , Cell Line , Cysteine/genetics , Cysteine/physiology , DNA Mutational Analysis , Glycosylation , Hot Temperature , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Transformation, Genetic/genetics , Wnt Proteins , Wnt1 Protein
9.
Oncogene ; 18(1): 149-56, 1999 Jan 07.
Article in English | MEDLINE | ID: mdl-9926929

ABSTRACT

Wnt-1 induces morphological transformation of C57MG mammary epithelial cells and accumulation of cytosolic beta-catenin whereas Wnt-5a has no effect. To identify regions within the 370 amino acid Wnt-1 protein required for these functions we tested eleven chimeric genes that contained variable amounts of Wnt-1 and Wnt-5a sequence. Transformation and beta-catenin regulation in C57MG cells is controlled by amino acids that lie within 186 residues of the amino terminus of Wnt-1. Small substitutions between residues 186 and 292 reduced Wnt-1 activity. Replacement of the carboxy terminal 79 amino acids of Wnt-1 by Wnt-5a did not affect function. These results were supported by transient expression assays in 293 cells wherein beta-catenin accumulated in the cytoplasm in response to ectopic Wnt-1 expression. In 293 cells, a larger region of the amino-terminus of Wnt-1 was found to be required for beta-catenin regulation. Nonfunctional chimeras that contained at least 99 amino terminal Wnt-1 residues inhibited Wnt-1 stimulation of 293 cells. One of these chimeras inhibited both Wnt-1 and Wnt-3 activity suggesting that Wnt-1 and Wnt-3 interact with a common signaling component.


Subject(s)
Cell Transformation, Neoplastic , Proto-Oncogene Proteins/genetics , Trans-Activators , Zebrafish Proteins , Amino Acid Sequence , Animals , Binding Sites , Cell Line, Transformed , Cytoskeletal Proteins/metabolism , Cytosol/metabolism , Gene Expression , Humans , Mice , Molecular Sequence Data , Proteins/genetics , Recombinant Fusion Proteins , Sequence Homology, Amino Acid , Wnt Proteins , Wnt-5a Protein , Wnt1 Protein , Wnt3 Protein , beta Catenin
10.
Oncogene ; 18(44): 5959-66, 1999 Oct 28.
Article in English | MEDLINE | ID: mdl-10557084

ABSTRACT

The human homologue of fz1 (Hfz1) was cloned from a cDNA library. Hfz1 was shown to couple to Wnt signal transduction pathways by its ability to enhance Wnt induced TCF dependent transcription in both autocrine and paracrine modes. Enhanced TCF dependent signaling was dose dependent with respect to both Wnt-3A and Hfz1. Moreover, Hfz1 deletion mutants with truncated carboxy termini showed markedly reduced capacity to enhance Wnt signal transduction. Specificity was demonstrated with respect to signal transduction by different Wnts. While Wnt-3a, -3, -1 and to a lesser extent Wnt-2 cooperated with Hfz1 in the paracrine assay for TCF dependent signaling, neither Wnt-4, -5a, -5b, -6, -7a nor -7b did so, despite similar levels of expression. However, coimmunoprecipitation of Hfz1 with both Wnt-3a and Wnt-5a indicated that TCF dependent signaling in response to Wnts is not determined solely by their ability to bind the receptor. All of these findings provide strong evidence that Hfz1 is a functional partner for certain Wnts in inducing TCF dependent transcription.


Subject(s)
DNA-Binding Proteins/metabolism , Proteins/metabolism , Receptors, Neurotransmitter/genetics , Receptors, Neurotransmitter/metabolism , Transcription Factors/metabolism , Zebrafish Proteins , Cell Line , Cell Transformation, Neoplastic , Cloning, Molecular , DNA-Binding Proteins/genetics , Frizzled Receptors , Humans , Lymphoid Enhancer-Binding Factor 1 , Molecular Biology/methods , Molecular Sequence Data , Mutation , Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled , Sensitivity and Specificity , Sequence Analysis , Signal Transduction , Transcription Factors/genetics , Transcription, Genetic , Transfection , Wnt Proteins , Wnt-5a Protein , Wnt2 Protein , Wnt3 Protein , Wnt3A Protein , Wnt4 Protein
11.
J Clin Endocrinol Metab ; 86(2): 768-72, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11158044

ABSTRACT

Indirect evidence in the nonhuman primate and human suggests that angiogenesis and regulators of angiogenesis such as vascular endothelial growth factor (VEGF) may play an active role in cyclic folliculogenesis. Indeed, the follicle selected for maturation and ovulation possesses a denser microvascular network, and VEGF messenger ribonucleic acid and its protein have been identified in granulosa cells of the developing follicle during the mid- and late follicular phases, with a more intense signal in the mature follicle. The objective of this study was to obtain direct evidence in the nonhuman primate for an active role of VEGF in follicular growth and maturation by studying the effect of VEGF-blocking antibodies in this process. After documenting two normal ovulatory cycles, female rhesus monkeys (n = 7) received iv injections of anti-VEGF antibodies (0.5 mg) twice on successive days in the late follicular phase. Three monkeys also received nonspecific goat IgG (0.5 mg) twice on successive days in the late follicular phase. Daily measurements of estradiol, progesterone, LH, and FSH were obtained during the two control cycles, the anti-VEGF treatment and posttreatment cycles, and the IgG treatment cycle. Anti-VEGF antibody administration significantly lengthened the follicular phase in six of seven monkeys to 17.8 +/- 1.7 vs. 10.0 +/- 0.7 and 9.8 +/- 0.6 in control cycles and 10.7 +/- 0.3 days (mean +/- SE) in IgG-treated cycles. The expected late follicular phase rise in estradiol, as documented in the control cycles (day 0, 96.1 +/- 6.0; day 1, 125.5 +/- 20.0; day 2, 165.5 +/- 24.9; day 3, 183.8 +/- 11.0 pg/mL), was interrupted by anti-VEGF antibody treatment (99.3 +/- 5.0, day 0, preinjection control) to 63.3 +/- 12.2 (day 1), 48.5 +/- 8.7 (day 2), and 57.6 +/- 9.0 (day 3). Mean FSH levels were significantly increased by day 2 of anti-VEGF antibody treatment. After a variable delay, estradiol concentrations increased to reach a preovulatory peak in all anti-VEGF-treated animals, followed by ovulation, normal luteal function, and a normal posttreatment cycle. The data clearly demonstrate that short-term inhibition of angiogenesis with an anti-VEGF-blocking antibody during the later growth phase of the dominant follicle interferes with normal follicular development. Persistence of estradiol secretion and delayed resumption of its rise also suggest recovery of the follicle. We conclude that the angiogenic regulator VEGF is a crucial component in the process of follicular growth in the primate.


Subject(s)
Antibodies/pharmacology , Endothelial Growth Factors/immunology , Follicular Phase/physiology , Lymphokines/immunology , Ovarian Follicle/physiology , Animals , Endothelial Growth Factors/physiology , Female , Follicle Stimulating Hormone/blood , Humans , Immunoglobulin G/pharmacology , Luteinizing Hormone/blood , Lymphokines/physiology , Macaca mulatta , Neovascularization, Physiologic , Time Factors , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
12.
Methods Enzymol ; 207: 423-31, 1992.
Article in English | MEDLINE | ID: mdl-1528123

ABSTRACT

The utility of baculovirus as a vehicle for protein expression for both soluble and integral membrane proteins has been proved repeatedly. Our results suggest that baculovirus also holds promise as a means for expressing ion channel proteins. Because Sf9 cells are especially well suited to electrophysiology and because the construction of recombinant viruses that carry cloned genes is easy, baculovirus may also prove valuable for detailed functional studies of ion channels.


Subject(s)
Membrane Proteins/biosynthesis , Potassium Channels/genetics , Baculoviridae/genetics , Electric Conductivity , Genetic Vectors/genetics , Humans , Microelectrodes , Recombination, Genetic , Transfection
13.
Biotechniques ; 28(4): 702-8, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10769748

ABSTRACT

We have developed a retroviral vector that incorporates unique features of some previously described vectors. This vector includes: 3' long terminal repeats (LTRs) of the self-inactivating class; a 5' LTR that is a hybrid of the cytomegalovirus (CMV) enhancer and the mouse sarcoma virus promoter; an internal CMV immediate early region promoter to drive expression of the transduced gene and the neomycin phosphotransferase selectable marker; an expanded multiple cloning site and an internal ribosome entry site. An SV40 ori was introduced into the vector backbone to promote high copy number replication in packaging cell lines that express the SV40 large T antigen. We demonstrate that these retroviral constructs, designated Q vectors, can be used in applications where high viral titers and high level stable or transient gene expression are desirable.


Subject(s)
Gene Transfer Techniques , Genes/genetics , Genetic Vectors/genetics , Retroviridae/genetics , Zebrafish Proteins , Animals , Antigens, Viral, Tumor/metabolism , Cell Line , Cytomegalovirus/genetics , Epithelial Cells/cytology , Epithelial Cells/virology , Female , Fibroblasts/cytology , Fibroblasts/virology , Genes, Reporter/genetics , Genetic Vectors/chemistry , Mammary Glands, Animal/cytology , Mammary Glands, Animal/virology , Mice , Plasmids/chemistry , Plasmids/genetics , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Rats , Retroviridae/growth & development , Sarcoma Viruses, Murine/genetics , Transfection , Wnt Proteins , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
14.
Adv Exp Med Biol ; 480: 175-84, 2000.
Article in English | MEDLINE | ID: mdl-10959425

ABSTRACT

We have investigated whether repression of the putative tumor suppressor gene BARD1 or expression of the Notch4(int-3) oncogene in non-tumorigenic mammary epithelial cells affects their in vitro morphogenetic properties. Bard1 (Brca1-associated ring domain) is a protein interacting with Brca1 and thought to be involved in Brca1-mediated tumor suppression. To investigate the potential role of Bard1 in mammary gland development, we repressed its expression in TAC-2 cells, a murine mammary epithelial cell line which, when grown in three-dimensional collagen gels, forms branching ducts in response to hepatocyte growth factor (HGF) and alveolar-like cysts in response to hydrocortisone. Whereas Bard1 repression did not markedly modify the tubulogenic response of TAC-2 cells to HGF, it dramatically altered cyst development, resulting in the formation of compact cell aggregates devoid of central lumen. In addition, when grown to post-confluence in two-dimensional cultures, Bard1-suppressed TAC-2 cells overcame contact-inhibition of cell proliferation and formed multiple cell layers. The Notch4(int-3) oncogene, which codes for a constitutively activated form of the Notch4 receptor, has been reported to induce undifferentiated carcinomas when expressed in the mammary gland. The potential effect of activated Notch4 on mammary gland morphogenesis was investigated by retroviral expression of the oncogene in TAC-2 cells. Notch4(int-3) expression was found to significantly reduce HGF-induced tubulogenesis and to markedly inhibit hydrocortisone-induced cyst formation. In addition, Notch4(int-3) expressing TAC-2 cells formed multilayers in post-confluent cultures and exhibited an invasive behavior when grown on the surface of collagen gels. Taken together, these results indicate that both repression of Bard1 and expression of Notch4(int-3) disrupt cyst morphogenesis and induce an invasive phenotype in TAC-2 mammary epithelial cells.


Subject(s)
Breast , Carrier Proteins/physiology , Proto-Oncogene Proteins/physiology , Receptors, Cell Surface , Tumor Suppressor Proteins , Ubiquitin-Protein Ligases , Breast/embryology , Breast/physiology , Cell Line , Female , Gene Expression Regulation, Developmental/physiology , Genes, Tumor Suppressor , Humans , Morphogenesis/physiology , Receptor, Notch4 , Receptors, Notch
15.
Oncogene ; 29(6): 789-801, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-19901963

ABSTRACT

The Capillary Morphogenesis Gene 2 (CMG2) gene encodes an Anthrax toxin receptor (ANTXR2), but the normal physiological function is not known. ANTXR2/CMG2 was originally identified as a result of up-regulation during capillary morphogenesis of endothelial cells (ECs) cultured in vitro. We explored the hypothesis that key steps of the angiogenic process are either dependent or are influenced by ANTXR2/CMG2 activity. We describe the expression pattern of ANTXR2/CMG2 in several murine tissues and in normal breast and breast tumors. Endothelial expression was found in all of the tissues analyzed, in cultured ECs and in breast tumor vessels; however, ANTXR2/CMG2 expression was not restricted to this cell type. To assess potential angiogenic function, we used RNA interference to achieve significant reduction of ANTXR2/CMG2 expression in cultured human umbilical venous endothelial cells (HUVECs). Reduced ANTXR2/CMG2 expression resulted in significant inhibition of proliferation and reduced capacity of ECs to form capillary-like networks in vitro, whereas overexpression of ANTXR2/CMG2 in HUVEC increased proliferation and capillary-like network formation. Little change in migration of ECs was observed on knockdown or overexpression. We conclude that ANTXR2/CMG2 functions to promote endothelial proliferation and morphogenesis during sprouting angiogenesis, consistent with the endothelial expression of ANTXR2/CMG2 in several vascular beds.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/pathology , Gene Expression Regulation , Morphogenesis , Neoplasms/blood supply , Neoplasms/genetics , Receptors, Peptide/metabolism , Animals , Breast/blood supply , Breast/cytology , Breast/metabolism , Breast/pathology , Capillaries/cytology , Capillaries/growth & development , Capillaries/pathology , Cell Line , Cell Movement/genetics , Cell Proliferation , Endothelial Cells/metabolism , Endothelium/growth & development , Gene Knockdown Techniques , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Morphogenesis/genetics , Neoplasms/metabolism , Neovascularization, Physiologic/genetics , Receptors, Peptide/deficiency , Receptors, Peptide/genetics
17.
Oncogene ; 27(38): 5132-7, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18758482

ABSTRACT

The Notch signaling pathway is fundamental to proper cardiovascular development and is now recognized as an important player in tumor angiogenesis. Two key Notch ligands have been implicated in tumor angiogenesis, Delta-like 4 and Jagged1. We introduce the proteins and how they work in normal developing vasculature and then discuss differing models describing the action of these Notch ligands in tumor angiogenesis. Endothelial Dll4 expression activates Notch resulting in restriction of new sprout development; for instance, in growing retinal vessels. In agreement with this activity, inhibition of Dll4-mediated Notch signaling in tumors results in hypersprouting of nonfunctional vasculature. This Dll4 inhibition may paradoxically lead to increased angiogenesis but poor tumor growth because the newly growing vessels are not functional. In contrast, Jagged1 has been described as a Notch ligand expressed in tumor cells that can have a positive influence on tumor angiogenesis, possibly by activating Notch on tumor endothelium. A novel Notch inhibitor, the Notch1 decoy, which blocks both Dll4 and Jagged1 has been recently shown to restrict tumor vessel growth. We discuss these models and speculate on therapeutic approaches.


Subject(s)
Neoplasm Proteins/physiology , Neoplasms/blood supply , Neovascularization, Pathologic/physiopathology , Receptors, Notch/physiology , Animals , Arteries/cytology , Arteries/embryology , Calcium-Binding Proteins/physiology , Cell Communication/physiology , Cell Differentiation/physiology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Gene Expression Regulation, Neoplastic/physiology , Humans , Intercellular Signaling Peptides and Proteins/physiology , Intracellular Signaling Peptides and Proteins , Jagged-1 Protein , Membrane Proteins/physiology , Mice , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Neoplasms/physiopathology , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/physiopathology , Neovascularization, Physiologic/physiology , Serrate-Jagged Proteins , Signal Transduction/physiology
18.
Bioessays ; 22(10): 902-10, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10984716

ABSTRACT

Reproductive tissues respond to steroid hormones and thus are particularly vulnerable to the effects of exogenous steroid 'mimic' compounds (endocrine disrupters). One such endocrine disrupter, diethylstilbestrol (DES), is linked to gynecological cancers and changes in uterine structure that reduce or completely abrogate reproductive competence. Until recently, little was known about the identity of target genes and signaling pathways involved in pathologies linked to endocrine disrupters such as DES. We outline genetic, cellular and molecular roles for patterning genes, with emphasis on homeobox and Wnt genes. There is evidence that changes in the expression of Wnt and homeogenes underlie many of the defects induced by DES. Data obtained from murine systems will likely apply to a broad spectrum of gynecological pathologies involving abnormal cell behaviors ranging from fibroids to malignant tumors. Knowledge garnered from modern molecular genetics should lead to progress in the emerging field of molecular gynecology.


Subject(s)
Genes, Homeobox , Genitalia, Female/physiology , Proto-Oncogene Proteins/genetics , Zebrafish Proteins , Animals , Female , Humans , Morphogenesis , Proteins/physiology , Signal Transduction , Wnt Proteins
19.
Genes Dev ; 7(11): 2181-93, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8224845

ABSTRACT

The product of the Wnt-1 proto-oncogene is a cysteine-rich glycoprotein that plays a crucial role in the development of the vertebrate central nervous system. Wnt-1 protein is secreted but remains associated with the cell surface and extracellular matrix. The function of Wnt-1 in several different biological settings can be carried out by cells that receive the Wnt signal from adjacent cells. Ectopic expression of Wnt-1 in certain mammary gland cell lines, such as C57MG, causes morphological transformation; C57MG cells can also be transformed by a paracrine mechanism when mixed with other cell types secreting Wnt-1 protein. To ask whether Wnt-1 protein can function while bound to the cell of origin, a variety of cell types were programmed to produce chimeric proteins containing the complete sequence of mature Wnt-1 protein fused to part or all of the transmembrane protein CD4 or CD8. The chimeras were found at the cell surface of transfected cells and did not appear to be proteolytically processed. In autocrine and paracrine transformation assays with C57MG cells and in an axis induction assay in Xenopus laevis embryos, the Wnt-1/CD4 or CD8 fusions retained significant activity, as did a secreted chimera containing the CD8 extracellular domain but lacking the transmembrane domain. However, a chimera lacking a spacer between the Wnt-1 and the transmembrane domains was weakly active and only in autocrine transformation. These results show that tethering Wnt-1 to the cell surface still allows Wnt-1-mediated cell-to-cell signaling.


Subject(s)
Embryo, Nonmammalian/physiology , Proto-Oncogene Proteins/metabolism , Proto-Oncogenes , Zebrafish Proteins , Animals , Base Sequence , CD4 Antigens/biosynthesis , CD8 Antigens/biosynthesis , Cell Division , Cell Line , Cell Membrane/metabolism , Codon , Embryo, Nonmammalian/cytology , Fluorescent Antibody Technique , Gene Expression , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/biosynthesis , Rats , Recombinant Fusion Proteins/biosynthesis , Transfection , Wnt Proteins , Wnt1 Protein , Xenopus Proteins , Xenopus laevis/embryology
20.
Int J Cancer ; 86(5): 652-9, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10797286

ABSTRACT

The protein encoded by the Notch4 gene is a member of the Notch/lin-12 family of transmembrane receptor proteins, which have been shown to control cell fate determination and cell differentiation in a wide variety of organisms. Expression of Notch4(int-3), a truncated form of Notch4 having most of its extracellular domain deleted, as a transgene in mice induces the formation of poorly differentiated mammary carcinomas. To establish whether Notch4(int-3) has the capacity of subverting normal epithelial architecture, we assessed the effect of Notch4(int-3) expression on the in vitro morphogenetic properties of TAC-2 mammary epithelial cells. When grown in three-dimensional collagen gels in the presence of hydrocortisone, both wild-type and LacZ-transfected TAC-2 cells formed alveolar-like structures composed of polarized epithelial cells surrounding a central lumen. In contrast, TAC-2 cells programmed to express Notch4(int-3) formed compact cell aggregates devoid of tissue-specific organization. In addition, when grown on the surface of a collagen gel, Notch4(int-3)-expressing TAC-2 cells invaded the underlying matrix, whereas TAC-2 LacZ cells remained strictly confined to the gel surface. Expression of Notch4(int-3) in TAC-2 cells also disrupted contact-inhibition of cell proliferation, resulting in cell multilayering. Our results suggest that the ability of Notch4(int-3) to subvert normal epithelial morphogenesis and to promote invasion of the extracellular matrix contributes significantly to its tumorigenic potential.


Subject(s)
Cell Transformation, Neoplastic , Mammary Glands, Animal/cytology , Proto-Oncogene Proteins/physiology , Receptors, Cell Surface , Animals , Cell Division/genetics , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/physiology , Female , Mammary Glands, Animal/pathology , Mice , Neoplasm Invasiveness , Phenotype , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Receptor, Notch4 , Receptors, Notch
SELECTION OF CITATIONS
SEARCH DETAIL