Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Am J Pathol ; 185(11): 2907-22, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26429739

ABSTRACT

Cripto-1, a member of the epidermal growth factor-Cripto-1/FRL-1/Cryptic family, is critical for early embryonic development. Together with its ligand Nodal, Cripto-1 has been found to be associated with the undifferentiated status of mouse and human embryonic stem cells. Several studies have clearly shown that Cripto-1 is involved in regulating branching morphogenesis and epithelial-mesenchymal transition of the mammary gland both in vitro and in vivo and together with the cofactor GRP78 is critical for the maintenance of mammary stem cells ex vivo. Our previous studies showed that mammary-specific overexpression of human Cripto-1 exhibited dramatic morphological alterations in nulliparous mice mammary glands. The present study shows a novel mechanism for Cripto-1 regulation of mammary gland development through direct effects on progesterone receptor expression and pathways regulated by progesterone in the mammary gland. We demonstrate a strict temporal regulation of mouse Cripto-1 (mCripto-1) expression that occurs during mammary gland development and a stage-specific function of mCripto-1 signaling during mammary gland development. Our data suggest that Cripto-1, like the progesterone receptor, is not required for the initial ductal growth but is essential for subsequent side branching and alveologenesis during the initial stages of pregnancy. Dissection of the mechanism by which this occurs indicates that mCripto-1 activates receptor activator NF-κB/receptor activator NF-κB ligand, and NF-κB signaling pathways.


Subject(s)
Epidermal Growth Factor/metabolism , Membrane Glycoproteins/metabolism , NF-kappa B p50 Subunit/metabolism , Neoplasm Proteins/metabolism , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptors, Progesterone/metabolism , Signal Transduction , Animals , Cell Proliferation , Endoplasmic Reticulum Chaperone BiP , Epidermal Growth Factor/genetics , Epithelial Cells , Epithelial-Mesenchymal Transition , Female , Humans , Mammary Glands, Animal/cytology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Models, Biological , NF-kappa B p50 Subunit/genetics , Neoplasm Proteins/genetics , Organ Specificity , Pregnancy , RANK Ligand/genetics , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptors, Progesterone/genetics
2.
J Clin Invest ; 124(7): 3003-15, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24911146

ABSTRACT

The majority of non-small cell lung cancer (NSCLC) patients harbor EGFR-activating mutations that can be therapeutically targeted by EGFR tyrosine kinase inhibitors (EGFR-TKI), such as erlotinib and gefitinib. Unfortunately, a subset of patients with EGFR mutations are refractory to EGFR-TKIs. Resistance to EGFR inhibitors reportedly involves SRC activation and induction of epithelial-to-mesenchymal transition (EMT). Here, we have demonstrated that overexpression of CRIPTO1, an EGF-CFC protein family member, renders EGFR-TKI-sensitive and EGFR-mutated NSCLC cells resistant to erlotinib in culture and in murine xenograft models. Furthermore, tumors from NSCLC patients with EGFR-activating mutations that were intrinsically resistant to EGFR-TKIs expressed higher levels of CRIPTO1 compared with tumors from patients that were sensitive to EGFR-TKIs. Primary NSCLC cells derived from a patient with EGFR-mutated NSCLC that was intrinsically erlotinib resistant were CRIPTO1 positive, but gained erlotinib sensitivity upon loss of CRIPTO1 expression during culture. CRIPTO1 activated SRC and ZEB1 to promote EMT via microRNA-205 (miR-205) downregulation. While miR-205 depletion induced erlotinib resistance, miR-205 overexpression inhibited CRIPTO1-dependent ZEB1 and SRC activation, restoring erlotinib sensitivity. CRIPTO1-induced erlotinib resistance was directly mediated through SRC but not ZEB1; therefore, cotargeting EGFR and SRC synergistically attenuated growth of erlotinib-resistant, CRIPTO1-positive, EGFR-mutated NSCLC cells in vitro and in vivo, suggesting that this combination may overcome intrinsic EGFR-inhibitor resistance in patients with CRIPTO1-positive, EGFR-mutated NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , GPI-Linked Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation , Neoplasm Proteins/genetics , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition , Erlotinib Hydrochloride , GPI-Linked Proteins/metabolism , Gefitinib , Genes, erbB-1 , Homeodomain Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/metabolism , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Transcription Factors/metabolism , Xenograft Model Antitumor Assays , Zinc Finger E-box-Binding Homeobox 1 , src-Family Kinases/metabolism
3.
Open J Clin Diagn ; 3(2): 37-51, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23956953

ABSTRACT

The gut hormone apelin is a major therapeutic focus for several diseases involving inflammation and aberrant cell growth. We investigated whether apelin-36 contained alternative bioactive peptides associated with normal physiology or disease. Amino acid sequence analysis of apelin-36 identified an amidation motif consistent with the formation of a secondary bioactive peptide (SCNH2). SCNH2 is proven to be mitogenic and chemotactic in normal/malignant cells and augments angiogenesis via a PTX-resistant/CT-X-sensitive G protein-coupled receptor (GPCR). Notably, SCNH2 is substantially more potent and sensitive than apelin-13 and vascular endothelial growth factor-A. Endogenous SCNH2 is highly expressed in human tumors and placenta and in mouse embryonic tissues. Our findings demonstrate that SCNH2 is a new apelinergic member with critical pluripotent roles in angiogenesis related diseases and embryogenesis via a non-APJ GPCR.

4.
Cell Signal ; 25(1): 178-89, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23022962

ABSTRACT

Cripto-1 is implicated in multiple cellular events, including cell proliferation, motility and angiogenesis, through the activation of an intricate network of signaling pathways. A crosstalk between Cripto-1 and the canonical Wnt/ß-catenin signaling pathway has been previously described. In fact, Cripto-1 is a downstream target gene of the canonical Wnt/ß-catenin signaling pathway in the embryo and in colon cancer cells and T-cell factor (Tcf)/lymphoid enhancer factor binding sites have been identified in the promoter and the first intronic region of the mouse and human Cripto-1 genes. We now demonstrate that Cripto-1 modulates signaling through the canonical Wnt/ß-catenin/Tcf pathway by binding to the Wnt co-receptors low-density lipoprotein receptor-related protein (LRP) 5 and LRP6, which facilitates Wnt3a binding to LRP5 and LRP6. Cripto-1 functionally enhances Wnt3a signaling through cytoplasmic stabilization of ß-catenin and elevated ß-catenin/Tcf transcriptional activation. Conversely, Wnt3a further increases Cripto-1 stimulation of migration, invasion and colony formation in soft agar of HC11 mouse mammary epithelial cells, indicating that Cripto-1 and the canonical Wnt/ß-catenin signaling co-operate in regulating motility and in vitro transformation of mammary epithelial cells.


Subject(s)
GPI-Linked Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Neoplasm Proteins/metabolism , Wnt Signaling Pathway , Animals , Cell Line , Cell Movement , GPI-Linked Proteins/chemistry , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Mice , Neoplasm Proteins/chemistry , Protein Binding , Protein Structure, Tertiary , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transcriptional Activation , Wnt3A Protein/metabolism , beta Catenin/metabolism
5.
J Cell Physiol ; 228(6): 1174-88, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23129342

ABSTRACT

Human Cripto-1 (CR-1) plays an important role in regulating embryonic development while also regulating various stages of tumor progression. However, mechanisms that regulate CR-1 expression during embryogenesis and tumorigenesis are still not well defined. In the present study, we investigated the effects of two nuclear receptors, liver receptor homolog (LRH)-1 and germ cell nuclear factor receptor (GCNF) and epigenetic modifications on CR-1 gene expression in NTERA-2 human embryonal carcinoma cells and in breast cancer cells. CR-1 expression in NTERA-2 cells was positively regulated by LRH-1 through direct binding to a DR0 element within the CR-1 promoter, while GCNF strongly suppressed CR-1 expression in these cells. In addition, the CR-1 promoter was unmethylated in NTERA-2 cells, while T47D, ZR75-1, and MCF7 breast cancer cells showed high levels of CR-1 promoter methylation and low CR-1 mRNA and protein expression. Treatment of breast cancer cells with a demethylating agent and histone deacetylase inhibitors reduced methylation of the CR-1 promoter and reactivated CR-1 mRNA and protein expression in these cells, promoting migration and invasion of breast cancer cells. Analysis of a breast cancer tissue array revealed that CR-1 was highly expressed in the majority of human breast tumors, suggesting that CR-1 expression in breast cancer cell lines might not be representative of in vivo expression. Collectively, these findings offer some insight into the transcriptional regulation of CR-1 gene expression and its critical role in the pathogenesis of human cancer.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Embryonal/metabolism , DNA Methylation , GPI-Linked Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Nuclear Receptor Subfamily 6, Group A, Member 1/metabolism , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/metabolism , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/pathology , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/pathology , Cell Movement , DNA Methylation/drug effects , DNA Modification Methylases/antagonists & inhibitors , DNA Modification Methylases/metabolism , Decitabine , Dose-Response Relationship, Drug , Embryonal Carcinoma Stem Cells/metabolism , Embryonal Carcinoma Stem Cells/pathology , Female , GPI-Linked Proteins/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Genes, Reporter , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Intercellular Signaling Peptides and Proteins/genetics , Luciferases/biosynthesis , Luciferases/genetics , MCF-7 Cells , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Nuclear Receptor Subfamily 6, Group A, Member 1/genetics , RNA Interference , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Time Factors , Tissue Array Analysis , Transcription, Genetic , Transfection , Tretinoin/pharmacology , Valproic Acid/pharmacology
6.
Am J Pathol ; 180(6): 2188-200, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22542493

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is a critical multistep process that converts epithelial cells to more motile and invasive mesenchymal cells, contributing to body patterning and morphogenesis during embryonic development. In addition, both epithelial plasticity and increased motility and invasiveness are essential for the branching morphogenesis that occurs during development of the mammary gland and during tumor formation, allowing cancer cells to escape from the primary tumor. Cripto-1, a member of the epidermal growth factor-Cripto-1/FRL-1/Cryptic (EGF/CFC) gene family, together with the transforming growth factor (TGF)-ß family ligand Nodal, regulates both cell movement and EMT during embryonic development. During postnatal development, Cripto-1 regulates the branching morphogenesis of the mouse mammary gland and enhances both the invasive and migratory properties of mammary epithelial cells in vitro. Furthermore, transgenic mouse models have shown that Cripto-1 promotes the formation of mammary tumors that display properties of EMT, including the down-regulation of the cell surface adherens junctional protein E-cadherin and the up-regulation of mesenchymal markers, such as vimentin, N-cadherin, and Snail. Interestingly, Cripto-1 is enriched in a subpopulation of embryonal, melanoma, prostate, and pancreatic cancer cells that possess stem-like characteristics. Therefore, Cripto-1 may play a role during developmental EMT, and it may also be involved in the reprogramming of differentiated tumor cells into cancer stem cells through the induction of an EMT program.


Subject(s)
Breast Neoplasms/metabolism , Cell Transformation, Neoplastic/metabolism , Embryonic Development/physiology , Epithelial-Mesenchymal Transition/physiology , GPI-Linked Proteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Mammary Neoplasms, Experimental/metabolism , Neoplasm Proteins/physiology , Animals , Cell Transformation, Neoplastic/pathology , Female , GPI-Linked Proteins/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mammary Glands, Animal/embryology , Mammary Glands, Animal/growth & development , Mice , Neoplasm Proteins/genetics , Signal Transduction/physiology
7.
Growth Factors ; 30(1): 13-21, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22149969

ABSTRACT

Over the past few decades, our understanding of the embryonic gene Cripto-1 has considerably advanced through biochemical, cell biology, and animal studies. Cripto-1 performs key functions during embryonic development, while it dramatically disappears in adult tissues, except possibly in adult tissue stem cells. Cripto-1 is re-expressed in human tumors promoting cell proliferation, migration, invasion, epithelial to mesenchymal transition, and tumor angiogenesis. This diversity of biological effects is dependent upon interaction of Cripto-1 with an extensive array of signaling molecules. In fact, Cripto-1 modulates signaling of transforming growth factor-ß family members, including Nodal, GDF-1/-3, Activin, and TGF-ß1, activates c-src/MAPK/Protein Kinase B (AKT) pathway in a Glypican-1 and GRP78-dependent manner, and cross-talks with erbB4, Wnt/ß-catenin, Notch, Caveolin-1, and Apelin/putative receptor protein related to Angiotensin-type I receptor (APJ) pathways. This article provides an updated survey of the various signaling pathways modulated by Cripto-1 with a focus on mechanistic insights in our understanding of the biological function of Cripto-1 in eukaryotic cells.


Subject(s)
Embryonic Development/drug effects , GPI-Linked Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/pharmacology , Neoplasms/physiopathology , Signal Transduction/drug effects , Animals , Cricetinae , Endoplasmic Reticulum Chaperone BiP , GPI-Linked Proteins/metabolism , Gene Expression Regulation , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Neoplasm Proteins/metabolism , Neoplasms/metabolism
8.
Expert Opin Ther Pat ; 20(12): 1739-49, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21073352

ABSTRACT

IMPORTANCE OF THE FIELD: Emerging evidence has clearly implicated an inappropriate activation of embryonic regulatory genes during cell transformation in adult tissues. An example of such a case is the embryonic gene Cripto-1. Cripto-1 is critical for embryonic development and is considered a marker of undifferentiated embryonic stem cells. Critpo-1 is expressed at low levels in adult tissues, but is re-expressed at a high frequency in a number of different types of human carcinomas, therefore, representing an attractive therapeutic target in cancer. AREA COVERED IN THIS REVIEW: This review surveys different approaches that have been used to target Cripto-1 in cancer as reflected by the relevant patent literature as well as peer-reviewed publications. Potential involvement and targeting of Cripto-1 in neurodegenerative and degenerative muscle diseases are also discussed. WHAT THE READER WILL GAIN: The reader will gain an overview of different mAbs, vaccines or oligonucleotides antisense targeting Cripto-1. A humanized anti-Cripto-1 antibody is currently being tested in a Phase I clinical trial in cancer patients. TAKE HOME MESSAGE: Targeting Cripto-1 in human tumors has the potential to eliminate not only differentiated cancer cells but also destroy an undifferentiated subpopulation of cancer cells with stem-like characteristics that support tumor initiation and self-renewal.


Subject(s)
Drug Delivery Systems , Epidermal Growth Factor/immunology , Membrane Glycoproteins/immunology , Neoplasm Proteins/immunology , Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Cancer Vaccines/immunology , GPI-Linked Proteins , Gene Expression Regulation, Neoplastic , Humans , Intercellular Signaling Peptides and Proteins , Neoplasms/immunology , Neoplasms/physiopathology , Oligonucleotides, Antisense/pharmacology , Patents as Topic
9.
Am J Pathol ; 177(2): 532-40, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20616345

ABSTRACT

Cripto-1 is critical for early embryonic development and, together with its ligand Nodal, has been found to be associated with the undifferentiated status of mouse and human embryonic stem cells. Like other embryonic genes, Cripto-1 performs important roles in the formation and progression of several types of human tumors, stimulating cell proliferation, migration, epithelial to mesenchymal transition, and tumor angiogenesis. Several studies have demonstrated that cell fate regulation during embryonic development and cell transformation during oncogenesis share common signaling pathways, suggesting that uncontrolled activation of embryonic signaling pathways might drive cell transformation and tumor progression in adult tissues. Here we review our current understanding of how Cripto-1 controls stem cell biology and how it integrates with other major embryonic signaling pathways. Because many cancers are thought to derive from a subpopulation of cancer stem-like cells, which may re-express embryonic genes, Cripto-1 signaling may drive tumor growth through the generation or expansion of tumor initiating cells bearing stem-like characteristics. Therefore, the Cripto-1/Nodal signaling may represent an attractive target for treatment in cancer, leading to the elimination of undifferentiated stem-like tumor initiating cells.


Subject(s)
Disease Progression , Epidermal Growth Factor/metabolism , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms/pathology , Stem Cells/physiology , Animals , Embryonic Development , Epidermal Growth Factor/genetics , Epithelial-Mesenchymal Transition , GPI-Linked Proteins , Humans , Hypoxia , Intercellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Neoplasm Proteins/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nodal Protein/genetics , Nodal Protein/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction/physiology , Wnt Proteins/genetics , Wnt Proteins/metabolism
10.
Future Oncol ; 6(7): 1127-42, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20624125

ABSTRACT

Several studies have shown that cell fate regulation during embryonic development and oncogenic transformation share common regulatory mechanisms and signaling pathways. Indeed, an embryonic gene member of the EGF-Cripto-1/FRL1/Cryptic family, Cripto-1, has been implicated in embryogenesis and in carcinogenesis. Cripto-1 together with the TGF-beta ligand Nodal is a key regulator of embryonic development and is a marker of undifferentiated human and mouse embryonic stem cells. While Cripto-1 expression is very low in normal adult tissues, Cripto-1 is re-expressed at high levels in several different human tumors, modulating cancer cell proliferation, migration, epithelial-to-mesenchymal transition and stimulating tumor angiogenesis. Therefore, inhibition of Cripto-1 expression using blocking antibodies or antisense expression vectors might be a useful modality not only to target fully differentiated cancer cells but also to target a subpopulation of tumor cells with stem-like characteristics.


Subject(s)
Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasms/physiopathology , Amino Acid Sequence , Animals , Embryo, Mammalian , Embryonic Development/genetics , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Gene Expression Regulation, Developmental , Humans , Mammary Glands, Human/growth & development , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Molecular Sequence Data , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism
11.
Stem Cells ; 28(8): 1303-14, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20549704

ABSTRACT

Deregulation of stem cells is associated with the generation and progression of malignant tumors. In addition, genes that are associated with early embryogenesis are frequently expressed in cancer. Cripto-1 (CR-1), a glycosylphosphatidylinositol-linked glycoprotein, is expressed during early embryogenesis and in various human carcinomas. We demonstrated that human embryonal carcinoma (EC) cells are heterogeneous for CR-1 expression and consist of two distinct subpopulations: a CR-1(High) and a CR-1(Low) population. By segregating CR-1(High) and CR-1(Low) populations of NTERA2/D1 EC cells by fluorescence-activated cell sorting, we demonstrated that CR-1(High) cells were more tumorigenic than CR-1(Low) cells by an in vitro tumor sphere assay and by in vivo xenograft formation. The CR-1(High) population was enriched in mRNA expression for the pluripotent embryonic stem (ES) cell genes Oct4, Sox2, and Nanog. CR-1 expression in NTERA2/D1 cells was regulated by a Smad2/3-dependent autocrine loop, by the ES cell-related transcription factors Oct4/Nanog, and partially by the DNA methylation status of the promoter region. These results demonstrate that CR-1 expression is enriched in an undifferentiated, tumorigenic subpopulation and is regulated by key regulators of pluripotent stem cells.


Subject(s)
Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/metabolism , Epidermal Growth Factor/metabolism , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Animals , Blotting, Western , Cell Line , Chromatin Immunoprecipitation , DNA Methylation , Epidermal Growth Factor/genetics , Flow Cytometry , Fluorescent Antibody Technique , GPI-Linked Proteins , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Mice , Mice, Nude , Nanog Homeobox Protein , Neoplasm Proteins/genetics , Neoplasm Transplantation , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction
12.
J Cell Biol ; 187(3): 343-53, 2009 Nov 02.
Article in English | MEDLINE | ID: mdl-19948478

ABSTRACT

Nodal and Notch signaling pathways play essential roles in vertebrate development. Through a yeast two-hybrid screening, we identified Notch3 as a candidate binding partner of the Nodal coreceptor Cripto-1. Coimmunoprecipitation analysis confirmed the binding of Cripto-1 with all four mammalian Notch receptors. Deletion analyses revealed that the binding of Cripto-1 and Notch1 is mediated by the Cripto-1/FRL-1/Cryptic domain of Cripto-1 and the C-terminal region of epidermal growth factor-like repeats of Notch1. Binding of Cripto-1 to Notch1 occurred mainly in the endoplasmic reticulum-Golgi network. Cripto-1 expression resulted in the recruitment of Notch1 protein into lipid raft microdomains and enhancement of the furin-like protein convertase-mediated proteolytic maturation of Notch1 (S1 cleavage). Enhanced S1 cleavage resulted in the sensitization to ligand-induced activation of Notch signaling. In addition, knockdown of Cripto-1 expression in human and mouse embryonal carcinoma cells desensitized the ligand-induced Notch signaling activation. These results suggest a novel role of Cripto-1 in facilitating the posttranslational maturation of Notch receptors.


Subject(s)
Epidermal Growth Factor/metabolism , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Receptor, Notch1/metabolism , Signal Transduction , Amino Acid Motifs , Animals , Binding Sites , CHO Cells , COS Cells , Cells, Cultured , Chlorocebus aethiops , Cricetinae , Cricetulus , Epidermal Growth Factor/chemistry , Extracellular Matrix Proteins/metabolism , GPI-Linked Proteins , Gene Library , Humans , Intercellular Signaling Peptides and Proteins , Membrane Glycoproteins/chemistry , Membrane Microdomains/metabolism , Mice , Neoplasm Proteins/chemistry , Protein Interaction Mapping , Receptor, Notch1/chemistry , Receptor, Notch3 , Receptors, Notch/chemistry , Receptors, Notch/metabolism , Two-Hybrid System Techniques
13.
Am J Pathol ; 175(5): 2146-58, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19834060

ABSTRACT

Cripto-1 is a membrane-bound protein that is highly expressed in embryonic stem cells and in human tumors. In the present study, we investigated the effect of low levels of oxygen, which occurs naturally in rapidly growing tissues, on Cripto-1 expression in mouse embryonic stem (mES) cells and in human embryonal carcinoma cells. During hypoxia, Cripto-1 expression levels were significantly elevated in mES cells and in Ntera-2 or NCCIT human embryonal carcinoma cells, as compared with cells growing with normal oxygen levels. The transcription factor hypoxia-inducible factor-1alpha directly regulated Cripto-1 expression by binding to hypoxia-responsive elements within the promoter of mouse and human Cripto-1 genes in mES and NCCIT cells, respectively. Furthermore, hypoxia modulated differentiation of mES cells by enhancing formation of beating cardiomyocytes as compared with mES cells that were differentiated under normoxia. However, hypoxia failed to induce differentiation of mES cells into cardiomyocytes in the absence of Cripto-1 expression, demonstrating that Cripto-1 is required for hypoxia to fully differentiate mES cells into cardiomyocytes. Finally, cardiac tissue samples derived from patients who had suffered ischemic heart disease showed a dramatic increase in Cripto-1 expression as compared with nonischemic heart tissue samples, suggesting that hypoxia may also regulate Cripto-1 in vivo.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Epidermal Growth Factor/metabolism , Heart , Hypoxia/metabolism , Membrane Glycoproteins/metabolism , Myocytes, Cardiac/physiology , Neoplasm Proteins/metabolism , Animals , Biomarkers/metabolism , Cell Line , Embryonic Stem Cells/cytology , Epidermal Growth Factor/genetics , Heart/anatomy & histology , Heart/embryology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Myocardium/cytology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Neoplasm Proteins/genetics , Promoter Regions, Genetic , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Response Elements , Signal Transduction/physiology , Swine
14.
Expert Opin Ther Pat ; 19(2): 141-4, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19441915

ABSTRACT

Human Cripto-1 is a cell membrane protein that has been shown to be overexpressed in different types of human tumors. Because Cripto-1 is expressed at low levels in normal tissues, it represents a promising candidate for therapeutic intervention in cancer. The present patent describes a novel approach to target Cripto-1 in cancer using a vaccine. Immunization with Cripto-1 modified by addition of a foreign peptide to overcome T-cell tolerance for self-proteins has the potential to generate an antibody-based immune response that ultimately will block Cripto-1 activity in cancer cells. Although targeting Cripto-1 with a vaccine in cancer patients is promising, several experimental and clinical studies need to be done to validate this approach.


Subject(s)
Cancer Vaccines/immunology , Epidermal Growth Factor/immunology , Membrane Glycoproteins/immunology , Neoplasm Proteins/immunology , Animals , Drug Delivery Systems , GPI-Linked Proteins , Gene Expression Regulation, Neoplastic/immunology , Humans , Intercellular Signaling Peptides and Proteins , Neoplasms/drug therapy , Neoplasms/immunology , Patents as Topic , Self Tolerance/immunology , T-Lymphocytes/immunology
15.
Cancer Res ; 69(5): 1717-21, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19223540

ABSTRACT

Pluripotent cells within embryonal carcinoma (EC) can differentiate in vivo or in vitro on treatment with specific agents. Differentiating EC cells express lower levels of stem cell-related genes, such as Cripto-1. We show that migration of human EC cells (NTERA/2 and NCCIT) can be reduced following treatment with the guidance molecule Netrin-1. Moreover, Netrin-1 treatment increased the levels of beta-III tubulin, glial filament acidic protein, Nestin, and gamma-aminobutyric acid and reduced the expressions of Cripto-1, Nanog, and Oct4 in EC cells. These Netrin-1-induced effects in the EC cells were mediated via binding of Netrin-1 to the Neogenin receptor and activation of SHP-2, resulting in increased levels of inactive phosphorylated c-src((Y527)). These results suggest that Netrin-1 can induce neuroectodermal-like differentiation of human EC cells by affecting c-src signaling via SHP-2 activation and regulation of Nanog, Oct4, and Cripto-1 expressions.


Subject(s)
Embryonal Carcinoma Stem Cells/drug effects , Nerve Growth Factors/pharmacology , Tumor Suppressor Proteins/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Embryonal Carcinoma Stem Cells/pathology , Epidermal Growth Factor/analysis , GPI-Linked Proteins , Humans , Intercellular Signaling Peptides and Proteins , Membrane Glycoproteins/analysis , Neoplasm Proteins/analysis , Netrin-1 , Octamer Transcription Factor-3/analysis , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism
16.
FEBS Lett ; 582(29): 3997-4002, 2008 Dec 10.
Article in English | MEDLINE | ID: mdl-19013461

ABSTRACT

Cripto-1, a co-receptor for Nodal, can activate Nodal-dependent and Nodal-independent signaling pathways. In this study we have investigated whether Cripto-1 mutants, that fail to activate a Nodal-dependent signaling pathway, are capable to activate a Nodal-independent signaling pathway in mammary epithelial cells. Cripto-1 mutants expressed in EpH4 mouse mammary epithelial cells are fully functional in regard to activation of a Nodal-independent signaling pathway, leading to phosphorylation of mitogen-activated protein kinase (MAPK) and Akt and to enhanced proliferation and motility of these cells, suggesting that Cripto-1 mutants with impaired Nodal signaling are still active in a Nodal-independent signaling pathway.


Subject(s)
Epidermal Growth Factor/metabolism , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Nodal Protein/metabolism , Animals , COS Cells , Cell Line , Cell Movement , Cell Proliferation , Chlorocebus aethiops , Epidermal Growth Factor/genetics , Epithelial Cells/metabolism , Glypicans/metabolism , Mammary Glands, Animal , Membrane Glycoproteins/genetics , Mice , Neoplasm Proteins/genetics , Signal Transduction
17.
Biochim Biophys Acta ; 1778(12): 2671-81, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18930707

ABSTRACT

Epidermal Growth Factor-Cripto-1/FRL-1/Cryptic (EGF-CFC) proteins, including human Cripto-1 (hCFC2/hCR-1) and human Cryptic (hCFC1), are membrane-associated Nodal co-receptors, which have critical roles in vertebrate development. Most of the EGF-CFC proteins have been experimentally proven or predicted to be glycosylphosphatidylinositol (GPI)-anchored proteins. However, unlike other EGF-CFC proteins, hCFC1 does not exhibit a typical GPI-signal sequence, containing a 32-amino acid hydrophilic extension in its COOH-terminal end. Here we experimentally demonstrate that the COOH-terminal sequence of hCFC1 functions as a GPI-anchoring signal. Moreover, addition of a hydrophilic epitope tag of 55-amino acids (V5-His) after the GPI signal of hCR-1 interfered with generation of a GPI-anchored form of hCR-1. In contrast, addition of the same epitope tag to the end of GPI signal of hCFC1 did not affect the GPI-attachment of hCFC1. The COOH-terminal signal of hCFC1 could produce two different forms of the protein; a GPI-anchored form and an unprocessed form which was more prone to be secreted into the conditioned medium. The hydrophilic extension of hCFC1 negatively regulates the activity of hCFC1 as a Nodal co-receptor. These results demonstrate the presence of endogenous GPI-signal sequence with a hydrophilic extension, which can generate both GPI-anchored and soluble forms of the protein.


Subject(s)
Epidermal Growth Factor/metabolism , Glycosylphosphatidylinositols/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Nodal Protein/metabolism , Protein Sorting Signals/physiology , Amino Acid Sequence , Cell Line , Epidermal Growth Factor/genetics , GPI-Linked Proteins , Genes, Reporter , Glycosylphosphatidylinositols/genetics , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Kidney/cytology , Luciferases/metabolism , Membrane Glycoproteins/genetics , Molecular Sequence Data , Neoplasm Proteins/genetics , Nodal Protein/genetics , Protein Sorting Signals/genetics , Transfection
18.
Cell Signal ; 20(9): 1632-41, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18595660

ABSTRACT

Both canonical Wnt/beta-catenin and TGFbeta/Smad signaling pathways coordinately regulate pattern formation during embryogenesis as well as tumor progression. Evidence of cross-talk between these two pathways has been reported. Here we demonstrated that the Activin-like kinase 4 (Alk4)/Smad2 pathway facilitates the transcriptional activity of the oncogenic Wnt/beta-catenin/Tcf4 pathway through a novel Smad4-independent mechanism. Upon activation, Smad2 physically interacted with Tcf4, beta-catenin and the co-activator p300 to enhance transcriptional activity of beta-catenin/Tcf4 through the histone acetyltransferase activity of p300. Transactivation by Smad2 was independent of a Smad-binding element (SBE) and Smad4. Indeed, the enhancement of beta-catenin/Tcf4 transcriptional activity by activated Smad2 was negatively regulated by the presence of Smad4. Moreover, a tumor-derived missense mutant of Smad2, lacking the ability to bind to Smad4 was still able to enhance the Tcf4 transcriptional reporter in the presence of beta-catenin and Tcf4. Our findings suggest that Smad2 may function as an activator of canonical Wnt/beta-catenin/Tcf4 signaling through a SBE/Smad4-independent pathway.


Subject(s)
Histone Acetyltransferases/metabolism , Signal Transduction , Smad2 Protein/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , p300-CBP Transcription Factors/metabolism , Activin Receptors, Type I/metabolism , Cell Line , Humans , Models, Genetic , Protein Binding , Smad2 Protein/genetics , Smad4 Protein/metabolism , TCF Transcription Factors/metabolism , Transcription Factor 7-Like 2 Protein , Transcription, Genetic , Transcriptional Activation
19.
J Cell Physiol ; 216(3): 824-34, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18425773

ABSTRACT

Netrin-1 has been shown to regulate the function of the EGF-like protein Cripto-1 (Cr-1) and affect mammary gland development. Since Cr-1 is a target gene of Nanog and Oct4, we investigated the relationship between Netrin-1 and Cr-1, Nanog and Oct4 during different stages of development in the mouse mammary gland. Results from histological analysis show that exogenous Netrin-1 was able to induce formation of alveolar-like structures within the mammary gland terminal end buds of virgin transgenic Cripto-1 mice and enhance mammary gland alveologenesis in early pregnant FVB/N mice. Results from immunostaining and Western blot analysis show that Netrin-1, Nanog and Oct4 are expressed in the mouse embryonic mammary anlage epithelium while Cripto-1 is predominantly expressed outside this structure in the surrounding mesenchyme. We find that in lactating mammary glands of postnatal FVB/N mice, Netrin-1 expression is highest while Cripto-1 and Nanog levels are lowest indicating that Netrin-1 may perform a role in the mammary gland during lactation. HC-11 mouse mammary epithelial cells stimulated with lactogenic hormones and exogenous soluble Netrin-1 showed increased beta-casein expression as compared to control thus supporting the potential role for Netrin-1 during functional differentiation of mouse mammary epithelial cells. Finally, mouse ES cells treated with exogenous soluble Netrin-1 showed reduced levels of Nanog and Cripto-1 and higher levels of beta-III tubulin during differentiation. These results suggest that Netrin-1 may facilitate functional differentiation of mammary epithelial cells and possibly affect the expression of Nanog and/or Cripto-1 in multipotent cells that may reside in the mammary gland.


Subject(s)
Epidermal Growth Factor/metabolism , Homeodomain Proteins/metabolism , Mammary Glands, Animal/growth & development , Membrane Glycoproteins/metabolism , Morphogenesis/physiology , Neoplasm Proteins/metabolism , Nerve Growth Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Caseins/metabolism , Cell Differentiation , Cells, Cultured , Dexamethasone/metabolism , Embryonic Stem Cells/cytology , Epidermal Growth Factor/genetics , Female , Gene Expression Regulation, Developmental , Glucocorticoids/metabolism , Homeodomain Proteins/genetics , Insulin/metabolism , Lactation , Male , Mammary Glands, Animal/anatomy & histology , Mammary Glands, Animal/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Nanog Homeobox Protein , Neoplasm Proteins/genetics , Nerve Growth Factors/genetics , Netrin-1 , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pregnancy , Prolactin/metabolism , Tumor Suppressor Proteins/genetics
20.
Am J Pathol ; 172(2): 345-57, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18202186

ABSTRACT

Human and mouse Cripto-1 (CR-1/Cr-1) proteins play an important role in mammary gland development and tumorigenesis. In this study, we examined the relationship between Cripto-1 and caveolin-1 (Cav-1), a membrane protein that acts as a tumor suppressor in the mammary gland. Cripto-1 was found to interact with Cav-1 in COS7 cells and mammary epithelial cells. Using EpH4 mouse mammary epithelial cells expressing Cr-1 (EpH4 Cr-1) or Cr-1 and Cav-1 (EpH4 Cr-1/Cav-1), we demonstrate that Cav-1 expression markedly reduced the ability of Cr-1 to enhance migration, invasion, and formation of branching structures in EpH4 Cr-1/Cav-1 cells as compared to EpH4 Cr-1 cells. Furthermore, coexpression of Cav-1 together with Cr-1 in EpH4 Cr-1/Cav-1 cells inhibited Cr-1-mediated activation of c-src and mitogen-activated protein kinase signaling pathways. Conversely, primary mammary epithelial cells isolated from Cav-1 null(-/-)/mouse mammary tumor virus-CR-1 transgenic animals showed enhanced motility and activation of mitogen-activated protein kinase and c-src as compared to Cav-1(+/-)/CR-1 mammary cells. Finally, mammary tumors derived from mouse mammary tumor virus-CR-1 mice showed a dramatic reduction of Cav-1 expression as compared to mammary tissue from normal FVB/N mice, suggesting that in vivo Cav-1 is down-regulated during the process of CR-1-mediated mammary tumorigenesis.


Subject(s)
Caveolin 1/metabolism , Epidermal Growth Factor/metabolism , Epithelial Cells/metabolism , Mammary Glands, Animal/metabolism , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , COS Cells , CSK Tyrosine-Protein Kinase , Cell Movement/physiology , Chlorocebus aethiops , Enzyme Activation/physiology , Epithelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Mammary Glands, Animal/pathology , Mice , Mice, Transgenic , Microscopy, Confocal , Neoplasm Invasiveness/physiopathology , Protein-Tyrosine Kinases/metabolism , Transfection , src-Family Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...