Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
2.
Development ; 145(5)2018 03 01.
Article in English | MEDLINE | ID: mdl-29437830

ABSTRACT

Human cleft lip with or without cleft palate (CL/P) is a common craniofacial abnormality caused by impaired fusion of the facial prominences. We have previously reported that, in the mouse embryo, epithelial apoptosis mediates fusion at the seam where the prominences coalesce. Here, we show that apoptosis alone is not sufficient to remove the epithelial layers. We observed morphological changes in the seam epithelia, intermingling of cells of epithelial descent into the mesenchyme and molecular signatures of epithelial-mesenchymal transition (EMT). Utilizing mouse lines with cephalic epithelium-specific Pbx loss exhibiting CL/P, we demonstrate that these cellular behaviors are Pbx dependent, as is the transcriptional regulation of the EMT driver Snail1. Furthermore, in the embryo, the majority of epithelial cells expressing high levels of Snail1 do not undergo apoptosis. Pbx1 loss- and gain-of-function in a tractable epithelial culture system revealed that Pbx1 is both necessary and sufficient for EMT induction. This study establishes that Pbx-dependent EMT programs mediate murine upper lip/primary palate morphogenesis and fusion via regulation of Snail1. Of note, the EMT signatures observed in the embryo are mirrored in the epithelial culture system.


Subject(s)
Body Patterning/genetics , Epithelial-Mesenchymal Transition/genetics , Face/embryology , Morphogenesis/genetics , Nose/embryology , Pre-B-Cell Leukemia Transcription Factor 1/physiology , Snail Family Transcription Factors/genetics , Animals , Apoptosis/genetics , Cells, Cultured , Cleft Lip/embryology , Cleft Lip/genetics , Cleft Palate/embryology , Cleft Palate/genetics , Embryo, Mammalian , Face/abnormalities , Gene Expression Regulation, Developmental , Lip/embryology , Mice , Mice, Transgenic , Palate/embryology , Pre-B-Cell Leukemia Transcription Factor 1/genetics
3.
Cell Signal ; 29: 41-51, 2017 01.
Article in English | MEDLINE | ID: mdl-27511963

ABSTRACT

Recent evidence suggests that mammary cells expressing R-spondin receptor and Wnt pathway regulator Lgr5, regarded as a stem cell marker in multiple tissues, might represent mammary stem cells (MaSCs). Whether L gr5 marks a multipotent subpopulation of Lin-CD24low/medCD49fhigh MaSCs remains controversial. To some extent the differing results reflect different assays used to assess properties of stemness, including lineage tracing in vivo, mammosphere culture, and mammary fat pad transplantation assays. To address this issue directly, we isolated Lgr5+ cells from mammary glands of Lgr5-lacZ mice and established organoids based on principles adapted from studies of Wnt-driven Lgr5+ cell populations in other organs. Mammary organoids were grown from single Lgr5+ mammary cells in Matrigel, the substratum of choice for intestinal organoids, and in a growth factor cocktail containing EGF, Wnt3a and R-spondin, designed to optimally activate the endogenous Wnt signaling program of stem cells. Colonies derived from single Lgr5+ cells manifest at least four distinct cell populations: Lgr5+ and Lgr5- basal cells and c-Kit+ and c-Kit- luminal cells that spontaneously organize into a ductal structure with basal cells around the periphery and luminal cells lining an interior cavity, reminiscent of normal mammary duct structure. Lgr5+ cell-derived organoids were sustainable during prolonged passaging. In contrast, although Lgr5- cells expand into primary colonies, colony-forming efficiency immediately dissipated upon passaging. Furthermore, reproductive hormones induce epithelial cell proliferation resulting in marked increases in lumen diameter accompanied by squamous transdifferentiation. We propose this estrogen-responsive, self-organizing duct-like structure derived from single murine Lgr5+ mammary cells represents a "mini-breast" organoid.


Subject(s)
Estrogens/pharmacology , Mammary Glands, Animal/metabolism , Organoids/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Differentiation/drug effects , Colony-Forming Units Assay , Female , Mammary Glands, Animal/cytology , Mice, Inbred C57BL , Organoids/cytology , Organoids/drug effects
4.
J Biol Chem ; 291(47): 24390-24405, 2016 Nov 18.
Article in English | MEDLINE | ID: mdl-27733685

ABSTRACT

Signaling mechanisms that regulate mammary stem/progenitor cell (MaSC) self-renewal are essential for developmental changes that occur in the mammary gland during pregnancy, lactation, and involution. We observed that equine MaSCs (eMaSCs) maintain their growth potential in culture for an indefinite period, whereas canine MaSCs (cMaSCs) lose their growth potential in long term cultures. We then used this system to investigate the role of microvesicles (MVs) in promoting self-renewal properties. We found that Wnt3a and Wnt1 were expressed at higher levels in MVs isolated from eMaSCs compared with those from cMaSCs. Furthermore, eMaSC-MVs were able to induce Wnt/ß-catenin signaling in different target cells, including cMaSCs. Interestingly, the induction of Wnt/ß-catenin signaling in cMaSCs was prolonged when using eMaSC-MVs compared with recombinant Wnt proteins, indicating that MVs are not only important for transport of Wnt proteins, but they also enhance their signaling activity. Finally, we demonstrate that the eMaSC-MVs-mediated activation of the Wnt/ß-catenin signaling pathway in cMaSCs significantly improves the ability of cMaSCs to grow as mammospheres and, importantly, that this effect is abolished when eMaSC-MVs are treated with Wnt ligand inhibitors. This suggests that this novel form of intercellular communication plays an important role in self-renewal.


Subject(s)
Cell-Derived Microparticles/metabolism , Mammary Glands, Human/metabolism , Stem Cells/metabolism , Wnt Signaling Pathway/physiology , Wnt1 Protein/metabolism , Wnt3A Protein/metabolism , Animals , Dogs , Female , Horses , Humans , Mammary Glands, Human/cytology , Pregnancy , Stem Cells/cytology
5.
PLoS Genet ; 12(8): e1006217, 2016 08.
Article in English | MEDLINE | ID: mdl-27500936

ABSTRACT

Ribosome biogenesis is essential for cell growth and proliferation and is commonly elevated in cancer. Accordingly, numerous oncogene and tumor suppressor signaling pathways target rRNA synthesis. In breast cancer, non-canonical Wnt signaling by Wnt5a has been reported to antagonize tumor growth. Here, we show that Wnt5a rapidly represses rDNA gene transcription in breast cancer cells and generates a chromatin state with reduced transcription of rDNA by RNA polymerase I (Pol I). These effects were specifically dependent on Dishevelled1 (DVL1), which accumulates in nucleolar organizer regions (NORs) and binds to rDNA regions of the chromosome. Upon DVL1 binding, the Pol I transcription activator and deacetylase Sirtuin 7 (SIRT7) releases from rDNA loci, concomitant with disassembly of Pol I transcription machinery at the rDNA promoter. These findings reveal that Wnt5a signals through DVL1 to suppress rRNA transcription. This provides a novel mechanism for how Wnt5a exerts tumor suppressive effects and why disruption of Wnt5a signaling enhances mammary tumor growth in vivo.


Subject(s)
Breast Neoplasms/genetics , Dishevelled Proteins/genetics , RNA Polymerase I/genetics , Transcription, Genetic , Wnt-5a Protein/genetics , Breast Neoplasms/pathology , Chromatin/genetics , DNA, Ribosomal/genetics , Dishevelled Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Nucleolus Organizer Region/genetics , Promoter Regions, Genetic , Protein Binding , RNA, Ribosomal/genetics , Sirtuins/genetics , Wnt Signaling Pathway/genetics , Wnt-5a Protein/metabolism
6.
PLoS One ; 9(7): e101800, 2014.
Article in English | MEDLINE | ID: mdl-25019931

ABSTRACT

The characterization of mammary stem cells, and signals that regulate their behavior, is of central importance in understanding developmental changes in the mammary gland and possibly for targeting stem-like cells in breast cancer. The canonical Wnt/ß-catenin pathway is a signaling mechanism associated with maintenance of self-renewing stem cells in many tissues, including mammary epithelium, and can be oncogenic when deregulated. Wnt1 and Wnt3a are examples of ligands that activate the canonical pathway. Other Wnt ligands, such as Wnt5a, typically signal via non-canonical, ß-catenin-independent, pathways that in some cases can antagonize canonical signaling. Since the role of non-canonical Wnt signaling in stem cell regulation is not well characterized, we set out to investigate this using mammosphere formation assays that reflect and quantify stem cell properties. Ex vivo mammosphere cultures were established from both wild-type and Wnt1 transgenic mice and were analyzed in response to manipulation of both canonical and non-canonical Wnt signaling. An increased level of mammosphere formation was observed in cultures derived from MMTV-Wnt1 versus wild-type animals, and this was blocked by treatment with Dkk1, a selective inhibitor of canonical Wnt signaling. Consistent with this, we found that a single dose of recombinant Wnt3a was sufficient to increase mammosphere formation in wild-type cultures. Surprisingly, we found that Wnt5a also increased mammosphere formation in these assays. We confirmed that this was not caused by an increase in canonical Wnt/ß-catenin signaling but was instead mediated by non-canonical Wnt signals requiring the receptor tyrosine kinase Ror2 and activity of the Jun N-terminal kinase, JNK. We conclude that both canonical and non-canonical Wnt signals have positive effects promoting stem cell activity in mammosphere assays and that they do so via independent signaling mechanisms.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Mammary Glands, Animal/cytology , Stem Cells/physiology , Wnt Signaling Pathway/physiology , Animals , Cells, Cultured , Female , Gene Expression Regulation, Developmental/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Stem Cells/drug effects , Wnt Signaling Pathway/drug effects , Wnt1 Protein/genetics , Wnt3A Protein/pharmacology
7.
Proc Natl Acad Sci U S A ; 110(29): 11851-6, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23818587

ABSTRACT

Peptidylarginine deiminase 4 (PAD4) is a Ca(2+)-dependent enzyme that converts arginine and methylarginine residues to citrulline, with histone proteins being among its best-described substrates to date. However, the biological function of this posttranslational modification, either in histones or in nonhistone proteins, is poorly understood. Here, we show that PAD4 recognizes, binds, and citrullinates glycogen synthase kinase-3ß (GSK3ß), both in vitro and in vivo. Among other functions, GSK3ß is a key regulator of transcription factors involved in tumor progression, and its dysregulation has been associated with progression of human cancers. We demonstrate that silencing of PAD4 in breast cancer cells leads to a striking reduction of nuclear GSK3ß protein levels, increased TGF-ß signaling, induction of epithelial-to-mesenchymal transition, and production of more invasive tumors in xenograft assays. Moreover, in breast cancer patients, reduction of PAD4 and nuclear GSK3ß is associated with increased tumor invasiveness. We propose that PAD4-mediated citrullination of GSK3ß is a unique posttranslational modification that regulates its nuclear localization and thereby plays a critical role in maintaining an epithelial phenotype. We demonstrate a dynamic and previously unappreciated interplay between histone-modifying enzymes, citrullination of nonhistone proteins, and epithelial-to-mesenchymal transition.


Subject(s)
Citrulline/metabolism , Epithelial-Mesenchymal Transition/physiology , Glycogen Synthase Kinase 3/metabolism , Hydrolases/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Calcium Ionophores , Fluorescent Antibody Technique , Gene Knockdown Techniques , Glycogen Synthase Kinase 3 beta , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , MCF-7 Cells , Mass Spectrometry , Microscopy, Interference , Mutagenesis, Site-Directed , Protein-Arginine Deiminase Type 4 , Protein-Arginine Deiminases , Real-Time Polymerase Chain Reaction , Statistics, Nonparametric
8.
Hum Mol Genet ; 22(21): 4267-81, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23773994

ABSTRACT

Low-density lipoprotein receptor related protein 6 (Lrp6) mutational effects on neurulation were examined using gain (Crooked tail, Lrp6(Cd)) and loss (Lrp6(-)) of function mouse lines. Two features often associated with canonical Wnt signaling, dorsal-ventral patterning and proliferation, were no different from wild-type (WT) in the Lrp6(Cd/Cd) neural tube. Lrp6(-/-) embryos showed reduced proliferation and subtle patterning changes in the neural folds. Cell polarity defects in both Lrp6(Cd/Cd) and Lrp6(-/-) cranial folds were indicated by cell shape, centrosome displacement and failure of F-actin and GTP-RhoA accumulation at the apical surface. Mouse embryonic fibroblasts (MEFs) derived from Lrp6(Cd/Cd) or Lrp6(-/-) embryos exhibited elevated and decreased RhoA basal activity levels, respectively. While ligand-independent activation of canonical Wnt signaling, bypassing Lrp-Frizzled receptors, did not activate RhoA, non-canonical Wnt5a stimulation of RhoA activity was impaired in Lrp6(-/-) MEFs. RhoA inhibition exacerbated NTDs in cultured Lrp6 knockout embryos compared with WT littermates. In contrast, a ROCK inhibitor rescued Lrp6(Cd/Cd) embryos from NTDs. Lrp6 co-immunoprecipitated with Disheveled-associated activator of morphogenesis 1 (DAAM1), a formin promoting GEF activity in Wnt signaling. Biochemical and cell biological data revealed intracellular accumulation of Lrp6(Cd) protein where interaction with DAAM1 could account for observed elevated RhoA activity. Conversely, null mutation that eliminates Lrp6 interaction with DAAM1 led to lower basal RhoA activity in Lrp6(-/-) embryos. These results indicate that Lrp6 mediates not only canonical Wnt signaling, but can also modulate non-canonical pathways involving RhoA-dependent mechanisms to impact neurulation, possibly through intracellular complexes with DAAM1.


Subject(s)
Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/physiology , Neural Tube/embryology , Wnt Proteins/metabolism , Wnt Signaling Pathway , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Alleles , Animals , Cell Polarity , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Mice , Mice, Transgenic , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , NIH 3T3 Cells , Neural Crest/metabolism , Neural Tube/physiology , Neurulation/genetics , Pregnancy , Wnt Proteins/genetics , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
9.
J Biol Chem ; 288(13): 9428-37, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23396967

ABSTRACT

Dishevelled (Dvl) proteins are intracellular effectors of Wnt signaling that have essential roles in both canonical and noncanonical Wnt pathways. It has long been known that Wnts stimulate Dvl phosphorylation, but relatively little is known about its functional significance. We have previously reported that both Wnt3a and Wnt5a induce Dvl2 phosphorylation that is associated with an electrophoretic mobility shift and loss of recognition by monoclonal antibody 10B5. In the present study, we mapped the 10B5 epitope to a 16-amino acid segment of human Dvl2 (residues 594-609) that contains four Ser/Thr residues. Alanine substitution of these residues (P4m) eliminated the mobility shift induced by either Wnt3a or Wnt5a. The Dvl2 P4m mutant showed a modest increase in canonical Wnt/ß-catenin signaling activity relative to wild type. Consistent with this finding, Dvl2 4Pm preferentially localized to cytoplasmic puncta. In contrast to wild-type Dvl2, however, the P4m mutant was unable to rescue Wnt3a-dependent neurite outgrowth in TC-32 cells following suppression of endogenous Dvl2/3. Earlier work has implicated casein kinase 1δ/ε as responsible for the Dvl mobility shift, and a CK1δ in vitro kinase assay confirmed that Ser(594), Thr(595), and Ser(597) of Dvl2 are CK1 targets. Alanine substitution of these three residues was sufficient to abrogate the Wnt-dependent mobility shift. Thus, we have identified a cluster of Ser/Thr residues in the C-terminal domain of Dvl2 that are Wnt-induced phosphorylation (WIP) sites. Our results indicate that phosphorylation at the WIP sites reduces Dvl accumulation in puncta and attenuates ß-catenin signaling, whereas it enables noncanonical signaling that is required for neurite outgrowth.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Gene Expression Regulation , Phosphoproteins/physiology , Wnt Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Alanine/chemistry , Animals , Culture Media, Conditioned , Dishevelled Proteins , HEK293 Cells , Humans , Mutation , Phosphoproteins/metabolism , Phosphorylation , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/metabolism , Rats , Serine/chemistry , Signal Transduction , Threonine/chemistry , Wnt-5a Protein , Wnt3A Protein/metabolism
10.
J Biol Chem ; 288(13): 9438-46, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23396968

ABSTRACT

Previously we reported that Wnt3a-dependent neurite outgrowth in Ewing sarcoma family tumor cell lines was mediated by Frizzled3, Dishevelled (Dvl), and c-Jun N-terminal kinase (Endo, Y., Beauchamp, E., Woods, D., Taylor, W. G., Toretsky, J. A., Uren, A., and Rubin, J. S. (2008) Mol. Cell. Biol. 28, 2368-2379). Subsequently, we observed that Dvl2/3 phosphorylation correlated with neurite outgrowth and that casein kinase 1δ, one of the enzymes that mediate Wnt3a-dependent Dvl phosphorylation, was required for neurite extension (Greer, Y. E., and Rubin, J. S. (2011) J. Cell Biol. 192, 993-1004). However, the functional relevance of Dvl phosphorylation in neurite outgrowth was not established. Dvl1 has been shown by others to be important for axon specification in hippocampal neurons via an interaction with atypical PKCζ, but the role of Dvl phosphorylation was not evaluated. Here we report that Ewing sarcoma family tumor cells express PKCι but not PKCζ. Wnt3a stimulated PKCι activation and caused a punctate distribution of pPKCι in the neurites and cytoplasm, with a particularly intense signal at the centrosome. Knockdown of PKCι expression with siRNA reagents blocked neurite formation in response to Wnt3a. Aurothiomalate, a specific inhibitor of PKCι/Par6 binding, also suppressed neurite extension. Wnt3a enhanced the co-immunoprecipitation of endogenous PKCι and Dvl2. Although FLAG-tagged wild-type Dvl2 immunoprecipitated with PKCι, a phosphorylation-deficient Dvl2 derivative did not. This derivative also was unable to rescue neurite outgrowth when endogenous Dvl2/3 was suppressed by siRNA (González-Sancho, J. M., Greer, Y. E., Abrahams, C. L., Takigawa, Y., Baljinnyam, B., Lee, K. H., Lee, K. S., Rubin, J. S., and Brown, A. M. (2013) J. Biol. Chem. 288, 9428-9437). Taken together, these results suggest that site-specific Dvl2 phosphorylation is required for Dvl2 association with PKCι. This interaction is likely to be one of the mechanisms essential for Wnt3a-dependent neurite outgrowth.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Neurites/metabolism , Phosphoproteins/metabolism , Protein Kinase C/physiology , Wnt3A Protein/metabolism , Casein Kinase I/metabolism , Dishevelled Proteins , HEK293 Cells , Hippocampus/metabolism , Humans , Isoenzymes/metabolism , Microscopy, Fluorescence/methods , Models, Biological , Neurons/metabolism , Phosphorylation , Protein Kinase C/metabolism , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Signal Transduction
11.
Biochem Biophys Res Commun ; 417(1): 223-30, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22142846

ABSTRACT

Wnt proteins that signal via the canonical Wnt/ß-catenin pathway directly regulate osteoblast differentiation. In contrast, most studies of Wnt-related effects on osteoclasts involve indirect changes. While investigating bone mineral density loss in the setting of human immunodeficiency virus (HIV) infection and its treatment with the protease inhibitor ritonavir (RTV), we observed that RTV decreased nuclear localization of ß-catenin, critical to canonical Wnt signaling, in primary human and murine osteoclast precursors. This occurred in parallel with upregulation of Wnt5a and Wnt5b transcripts. These Wnts typically stimulate noncanonical Wnt signaling, and this can antagonize the canonical Wnt pathway in many cell types, dependent upon Wnt receptor usage. We now document RTV-mediated upregulation of Wnt5a/b protein in osteoclast precursors. Recombinant Wnt5b and retrovirus-mediated expression of Wnt5a enhanced osteoclast differentiation from human and murine monocytic precursors, processes facilitated by RTV. In contrast, canonical Wnt signaling mediated by Wnt3a suppressed osteoclastogenesis. Both RTV and Wnt5b inhibited canonical, ß-catenin/T cell factor-based Wnt reporter activation in osteoclast precursors. RTV- and Wnt5-induced osteoclast differentiation were dependent upon the receptor-like tyrosine kinase Ryk, suggesting that Ryk may act as a Wnt5a/b receptor in this context. This is the first demonstration of a direct role for Wnt signaling pathways and Ryk in regulation of osteoclast differentiation, and its modulation by a clinically important drug, ritonavir. These studies also reveal a potential role for noncanonical Wnt5a/b signaling in acceleration of bone mineral density loss in HIV-infected individuals, and illuminate a potential means of influencing such processes in disease states that involve enhanced osteoclast activity.


Subject(s)
Bone Density/drug effects , HIV Protease Inhibitors/adverse effects , Osteoclasts/drug effects , Ritonavir/adverse effects , Wnt Proteins/metabolism , Animals , Cell Differentiation/drug effects , Cell Line , HEK293 Cells , Humans , Mice , Osteoclasts/metabolism , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Tyrosine Kinase-like Orphan Receptors/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Wnt Proteins/genetics , Wnt-5a Protein
12.
Proc Natl Acad Sci U S A ; 108(15): 5954-63, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21393571

ABSTRACT

Misregulated ß-catenin responsive transcription (CRT) has been implicated in the genesis of various malignancies, including colorectal carcinomas, and it is a key therapeutic target in combating various cancers. Despite significant effort, successful clinical implementation of CRT inhibitory therapeutics remains a challenging goal. This is, in part, because of the challenge of identifying inhibitory compounds that specifically modulate the nuclear transcriptional activity of ß-catenin while not affecting its cytoskeletal function in stabilizing adherens junctions at the cell membrane. Here, we report an RNAi-based modifier screening strategy for the identification of CRT inhibitors. Our data provide support for the specificity of these inhibitory compounds in antagonizing the transcriptional function of nuclear ß-catenin. We show that these inhibitors efficiently block Wnt/ß-catenin-induced target genes and phenotypes in various mammalian and cancer cell lines. Importantly, these Wnt inhibitors are specifically cytotoxic to human colon tumor biopsy cultures as well as colon cancer cell lines that exhibit deregulated Wnt signaling.


Subject(s)
Antineoplastic Agents/chemistry , Drug Discovery/methods , RNA Interference , Transcription, Genetic/drug effects , Wnt Proteins/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , Animals , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila melanogaster , Drug Screening Assays, Antitumor , Genes, Reporter , High-Throughput Screening Assays , Humans , Mice , Oxazoles/chemistry , Oxazoles/isolation & purification , Oxazoles/therapeutic use , Small Molecule Libraries , Wnt Proteins/genetics , Wnt1 Protein/antagonists & inhibitors , Wnt1 Protein/genetics , Xenograft Model Antitumor Assays , beta Catenin/genetics
13.
Breast Cancer Res ; 12(5): 313, 2010.
Article in English | MEDLINE | ID: mdl-20887643

ABSTRACT

The likely roles of Wnt signaling in regulating mammary stem cell behavior have been much discussed, in part because they may underlie the oncogenic effects of Wnt signaling in mammary tissue. Two recent papers add important data to this field. One tests directly the effects of purified Wnt protein on mouse mammary stem cells in culture and finds a specific increase in the proportion of cells with self-renewing stem cell phenotypes. The second identifies a novel target gene of canonical Wnt signaling that may be expressed in stem cells and is induced in both mouse and human mammary tumors associated with Wnt pathway activation.


Subject(s)
Breast Neoplasms/metabolism , Mammary Neoplasms, Animal/metabolism , Neoplastic Stem Cells/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Animals , Cell Cycle Proteins , Cell Line, Transformed , Female , Humans , Mice , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors , Wnt Proteins/pharmacology
14.
Curr Drug Targets ; 9(11): 1013-24, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18991612

ABSTRACT

Hepatocellular carcinoma (HCC) is a major cause of cancer death worldwide. As in many other types of cancer, aberrant activation of the canonical Wnt/beta-catenin signaling pathway is an important contributor to tumorigenesis. In HCC this frequently occurs through mutations in the N-terminal region of beta-catenin that stabilize the protein and permit an elevated level of constitutive transcriptional activation by beta-catenin/TCF complexes. In this article we review the abundant evidence that Wnt/beta-catenin signaling contributes to liver carcinogenesis. We also discuss what is known about the roles of Wnt signaling in liver development, regeneration, and stem cell behavior, in an effort to understand the mechanisms by which activation of the canonical Wnt pathway promotes tumor formation in this organ. The Wnt/beta-catenin pathway presents itself as an attractive target for developing novel rational therapies for HCC, a disease for which few successful treatment strategies are currently available.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Liver Neoplasms/physiopathology , Signal Transduction/physiology , Wnt Proteins/physiology , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Models, Biological , Signal Transduction/drug effects , Wnt Proteins/metabolism , beta Catenin/metabolism , beta Catenin/physiology
15.
Biochem Biophys Res Commun ; 356(3): 648-54, 2007 May 11.
Article in English | MEDLINE | ID: mdl-17376403

ABSTRACT

Low-density lipoprotein receptor-related protein 6 (LRP6) is a component of cell-surface receptors for Wnt proteins and Wnt is known to promote recruitment of Axin by LRP6 thereby inhibiting beta-catenin's degradation. We show here that growth factor receptor-bound protein10 (GRB10), a multi-modular adaptor protein that is known to associate with several transmembrane tyrosine kinase receptors, binds to the intracellular portion of LRP6 and negatively regulates Wnt signaling. GRB10 overexpression suppressed Wnt3a-, and LRP6-induced but not beta-catenin-induced TCF-dependent-reporter activities in HEK293T cells, suggesting that GRB10 functions upstream of beta-catenin. Actually, GRB10 overexpression attenuated the Wnt3a-induced accumulation of beta-catenin. In addition, RNAi-mediated down-regulation of endogenous GRB10 stimulated Wnt3a-induced reporter activities, indicating that GRB10 is indeed a novel negative regulator of the Wnt signaling pathway. The finding that GRB10 interferes with the binding of Axin to LRP6 indicated a possible molecular mechanism by which the overexpression of GRB10 suppresses Wnt signaling.


Subject(s)
GRB10 Adaptor Protein/metabolism , Receptors, LDL/metabolism , Wnt Proteins/physiology , Animals , Axin Protein , Binding Sites , Humans , Low Density Lipoprotein Receptor-Related Protein-6 , Mice , RNA Interference , Rats , Repressor Proteins/antagonists & inhibitors , Signal Transduction/drug effects , beta Catenin/antagonists & inhibitors , src Homology Domains/physiology
16.
Mol Biol Cell ; 17(12): 5163-72, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17035633

ABSTRACT

Wnts are lipid-modified secreted glycoproteins that regulate diverse biological processes. We report that Wnt5a, which functions in noncanonical Wnt signaling, has activity on endothelial cells. Wnt5a is endogenously expressed in human primary endothelial cells and is expressed in murine vasculature at several sites in mouse embryos and tissues. Expression of exogenous Wnt5a in human endothelial cells promoted angiogenesis. Wnt5a induced noncanonical Wnt signaling in endothelial cells, as measured by Dishevelled and ERK1/2 phosphorylation, and inhibition of canonical Wnt signaling, a known property of Wnt5a. Wnt5a induced endothelial cell proliferation and enhanced cell survival under serum-deprived conditions. The Wnt5a-mediated proliferation was blocked by Frizzled-4 extracellular domain. Wnt5a expression enhanced capillary-like network formation, whereas reduction of Wnt5a expression decreased network formation. Reduced Wnt5a expression inhibited endothelial cell migration. Screening for Wnt5a-regulated genes in cultured endothelial cells identified several encoding angiogenic regulators, including matrix metalloproteinase-1, an interstitial collagenase, and Tie-2, a receptor for angiopoietins. Thus, Wnt5a acts through noncanonical Wnt signaling to promote angiogenesis.


Subject(s)
Endothelial Cells/cytology , Matrix Metalloproteinase 1/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, TIE-2/metabolism , Signal Transduction , Wnt Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blood Vessels/metabolism , Cell Movement , Cell Proliferation , Cell Survival , Dishevelled Proteins , Extracellular Signal-Regulated MAP Kinases/metabolism , Frizzled Receptors/genetics , Gene Expression Regulation , Humans , Matrix Metalloproteinase 1/genetics , Mice , Microarray Analysis , Phosphoproteins/metabolism , Phosphorylation , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, TIE-2/genetics , Wnt Proteins/genetics , Wnt-5a Protein
17.
Cancer Res ; 65(21): 10113-9, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16267038

ABSTRACT

The inducible prostaglandin synthase cyclooxygenase-2 (Cox-2) is overexpressed in approximately 40% of human breast cancers and at higher frequencies in preinvasive ductal carcinoma in situ (DCIS). Cox-2 expression is particularly associated with overexpression of human epidermal growth factor receptor 2 (HER2/neu). To definitively interrogate the role of Cox-2 in mammary neoplasia, we have used a genetic approach, crossing Cox-2-deficient mice with a HER2/neu transgenic strain, MMTV/NDL. At 20 weeks of age, mammary glands from virgin MMTV/NDL females contained multiple focal tumors, or mammary intraepithelial neoplasias, which histologically resembled human DCIS. Mammary tumor multiplicity and prostaglandin E2 (PGE2) levels were significantly decreased in Cox-2 heterozygous and knockout animals relative to Cox-2 wild-type controls. Notably, the proportion of larger tumors was decreased in Cox-2-deficient mice. HER2/neu-induced mammary hyperplasia was also substantially reduced in Cox-2 null mice. Additionally, mammary glands from Cox-2 knockout mice exhibited a striking reduction in vascularization, and expression of proangiogenic genes was correspondingly reduced. Decreased vascularization was observed both in dysplastic and normal-appearing regions of Cox-2-null mammary glands. Our data provide the first genetic evidence that Cox-2 contributes to HER2/neu-induced mammary tumorigenesis. This finding may help to explain the reduced risk of breast cancer associated with regular use of nonsteroidal anti-inflammatory drugs.


Subject(s)
Cyclooxygenase 2/deficiency , Mammary Neoplasms, Experimental/genetics , Receptor, ErbB-2/genetics , Animals , Cyclooxygenase 2/genetics , Female , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/genetics
18.
Genome Biol ; 6(9): 231, 2005.
Article in English | MEDLINE | ID: mdl-16168092

ABSTRACT

The canonical Wnt signaling pathway is highly conserved in evolution, widely used throughout animal development, and frequently hyperactive in cancer. Although Wnt signaling has been the subject of extensive genetic analysis in the past, some 200 genes have now been identified as candidate modulators of this pathway by a recent study using high-throughput RNAi screening.


Subject(s)
RNA Interference , Signal Transduction , Wnt Proteins/metabolism , Animals , Drosophila melanogaster/metabolism , Humans , beta Catenin/metabolism
19.
Dev Biol ; 274(2): 334-47, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15385163

ABSTRACT

Pattern formation and growth must be tightly coupled during embryonic development. In vertebrates, however, little is known of the molecules that serve to link these two processes. Here we show that bone morphogenetic proteins (BMP) coordinate the acquisition of pattern information and the stimulation of proliferation in the embryonic spinal neural tube. We have blocked BMP and transforming growth factor-beta superfamily (TGFbeta) function in the chick embryo using Noggin, a BMP antagonist, and siRNA against Smad4. We show that BMPs/TGFbetas are necessary to regulate pattern formation and the specification of neural progenitor populations in the dorsal neural tube. BMPs also serve to establish discrete expression domains of Wnt ligands, receptors, and antagonists along the dorsal-ventral axis of the neural tube. Using the extracellular domain of Frizzled 8 to block Wnt signaling and Wnt3a ligand misexpression to activate WNT signaling, we demonstrate that the Wnt pathway acts mitogenically to expand the populations of neuronal progenitor cells specified by BMP. Thus, BMPs, acting through WNTs, couple patterning and growth to generate dorsal neuronal fates in the appropriate proportions within the neural tube.


Subject(s)
Body Patterning , Bone Morphogenetic Proteins/metabolism , Central Nervous System/embryology , Proto-Oncogene Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Bone Morphogenetic Proteins/genetics , Carrier Proteins , Cell Division , Central Nervous System/cytology , Central Nervous System/metabolism , Chick Embryo , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Morphogenesis , Neurons/cytology , Neurons/physiology , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/physiology , Smad4 Protein , Spinal Cord/cytology , Spinal Cord/embryology , Spinal Cord/metabolism , Stem Cells/cytology , Stem Cells/physiology , Trans-Activators/genetics , Trans-Activators/metabolism , Transforming Growth Factor beta/genetics , Wnt Proteins
20.
J Mammary Gland Biol Neoplasia ; 9(2): 119-31, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15300008

ABSTRACT

Secreted proteins of the Wnt family play widespread roles in the regulation of embryonic development, and aberrant activation of the canonical Wnt/beta-catenin pathway is one of the most frequent signaling abnormalities known in human cancer. While the consequences of Wnt signaling in development are diverse at the cellular level, they are often concerned with cell fate determination. Recent data also indicate that Wnt proteins influence the self-renewal of stem cells in certain tissues. In the mammary gland, Wnt signals are strongly implicated in initial development of the mammary rudiments, and in the ductal branching and alveolar morphogenesis that occurs during pregnancy. Transgenic expression of Wnt1 or Wnt10b in the mouse mammary gland leads to lobuloalveolar hyperplasia with a major risk of progression to carcinoma. Recent evidence suggests that this phenotype is associated with expansion of a multipotent progenitor cell population. In human breast cancer, evidence of beta-catenin accumulation implies that the canonical Wnt signaling pathway is active in over 50% of carcinomas. However, specific mutations that might account for this activation of signaling have not yet been identified.


Subject(s)
Breast Neoplasms/etiology , Breast/embryology , Intercellular Signaling Peptides and Proteins/physiology , Mammary Glands, Animal/embryology , Mammary Neoplasms, Animal/etiology , Cell Differentiation , Cell Movement , Cell Polarity , Signal Transduction , Stem Cells/cytology , Wnt Proteins , Wnt1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...