Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Structure ; 32(2): 217-227.e3, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38052206

ABSTRACT

E-cadherins (Ecads) are a crucial cell-cell adhesion protein with tumor suppression properties. Ecad adhesion can be enhanced by the monoclonal antibody 66E8, which has potential applications in inhibiting cancer metastasis. However, the biophysical mechanisms underlying 66E8-mediated adhesion strengthening are unknown. Here, we use molecular dynamics simulations, site-directed mutagenesis, and single-molecule atomic force microscopy experiments to demonstrate that 66E8 strengthens Ecad binding by stabilizing the primary Ecad adhesive conformation: the strand-swap dimer. By forming electrostatic interactions with Ecad, 66E8 stabilizes the swapped ß-strand and its hydrophobic pocket and impedes Ecad conformational changes, which are necessary for rupture of the strand-swap dimer. Our findings identify fundamental mechanistic principles for strengthening of Ecad binding using monoclonal antibodies.


Subject(s)
Cadherins , Molecular Dynamics Simulation , Cadherins/metabolism , Protein Binding , Cell Adhesion
2.
bioRxiv ; 2023 Jul 05.
Article in English | MEDLINE | ID: mdl-37461464

ABSTRACT

E-cadherins (Ecads) are a crucial cell-cell adhesion protein with tumor suppression properties. Ecad adhesion can be enhanced by the monoclonal antibody 66E8, which has potential applications in inhibiting cancer metastasis. However, the biophysical mechanisms underlying 66E8 mediated adhesion strengthening are unknown. Here, we use molecular dynamics simulations, site directed mutagenesis and single molecule atomic force microscopy experiments to demonstrate that 66E8 strengthens Ecad binding by stabilizing the primary Ecad adhesive conformation: the strand-swap dimer. By forming electrostatic interactions with Ecad, 66E8 stabilizes the swapped ß-strand and its hydrophobic pocket and impedes Ecad conformational changes, which are necessary for rupture of the strand-swap dimer. Our findings identify fundamental mechanistic principles for strengthening of Ecad binding using monoclonal antibodies.

3.
Mol Biol Cell ; 34(5): ar46, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36989029

ABSTRACT

Given the role of E-cadherin (E-cad) in holding epithelial cells together, an inverse relationship between E-cad levels and cell invasion during the epithelial-mesenchymal transition and cancer metastasis has been well recognized. Here we report that E-cad is necessary for the invasiveness of RasV12-transformed intestinal epithelial cells in Drosophila. E-cad/ß-catenin disassembles at adherens junctions and assembles at invasive protrusions--the actin- and cortactin-rich invadopodium-like protrusions associated with the breach of the extracellular matrix (ECM)--during dissemination of RasV12-transformed intestinal epithelial cells. Loss of E-cad impairs the elongation of invasive protrusions and attenuates the ability of RasV12-transformed cells to compromise the ECM. Notably, E-cad and cortactin affect each other's localization to invasive protrusions. Given the essential roles of cortactin in cell invasion, our observations indicate that E-cad plays a role in the invasiveness of RasV12-transformed intestinal epithelial cells by controlling cortactin localization to invasive protrusions. Thus our study demonstrates that E-cad is a component of invasive protrusions and provides molecular insights into the unconventional role of E-cad in cell dissemination in vivo.


Subject(s)
Cadherins , Cortactin , Animals , Cortactin/metabolism , Cadherins/metabolism , Epithelial Cells/metabolism , Actins/metabolism , Adherens Junctions/metabolism , Drosophila/metabolism
4.
PNAS Nexus ; 1(4): pgac163, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36157596

ABSTRACT

E-cadherin adhesion is regulated at the cell surface, a process that can be replicated by activating antibodies. We use cryo-electron microscopy (EM) and X-ray crystallography to examine functional states of the cadherin adhesive dimer. This dimer is mediated by N-terminal beta strand-swapping involving Trp2, and forms via a different transient X-dimer intermediate. X-dimers are observed in cryo-EM along with monomers and strand-swap dimers, indicating that X-dimers form stable interactions. A novel EC4-mediated dimer was also observed. Activating Fab binding caused no gross structural changes in E-cadherin monomers, but can facilitate strand swapping. Moreover, activating Fab binding is incompatible with the formation of the X-dimer. Both cryo-EM and X-ray crystallography reveal a distinctive twisted strand-swap dimer conformation caused by an outward shift in the N-terminal beta strand that may represent a strengthened state. Thus, regulation of adhesion involves changes in cadherin dimer configurations.

5.
Proc Natl Acad Sci U S A ; 119(32): e2204473119, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35921442

ABSTRACT

E-cadherin (Ecad) is an essential cell-cell adhesion protein with tumor suppression properties. The adhesive state of Ecad can be modified by the monoclonal antibody 19A11, which has potential applications in reducing cancer metastasis. Using X-ray crystallography, we determine the structure of 19A11 Fab bound to Ecad and show that the antibody binds to the first extracellular domain of Ecad near its primary adhesive motif: the strand-swap dimer interface. Molecular dynamics simulations and single-molecule atomic force microscopy demonstrate that 19A11 interacts with Ecad in two distinct modes: one that strengthens the strand-swap dimer and one that does not alter adhesion. We show that adhesion is strengthened by the formation of a salt bridge between 19A11 and Ecad, which in turn stabilizes the swapped ß-strand and its complementary binding pocket. Our results identify mechanistic principles for engineering antibodies to enhance Ecad adhesion.


Subject(s)
Antibodies, Monoclonal , Cadherins , Cell Adhesion , Antibodies, Monoclonal/chemistry , Cadherins/chemistry , Cadherins/immunology , Crystallography, X-Ray , Humans , Microscopy, Atomic Force , Molecular Dynamics Simulation , Protein Domains
6.
Protein Expr Purif ; 193: 106056, 2022 05.
Article in English | MEDLINE | ID: mdl-35063654

ABSTRACT

The dynamic regulation of epithelial adherens junctions relies on all components of the E-cadherin-catenin complex. Previously, the complexes have been partially reconstituted and composed only of α-catenin, ß-catenin, and the E-cadherin cytoplasmic domain. However, p120-catenin and the full-length E-cadherin including the extracellular, transmembrane, and intra-cellular domains are vital to the understanding of the relationship between extracellular adhesion and intracellular signaling. Here, we reconstitute the complete and full-length cadherin-catenin complex, including full-length E-cadherin, α-catenin, ß-catenin, and p120-catenin, into nanodiscs. We are able to observe the cadherin in nanodiscs by cryo-EM. We also reconstitute α-catenin, ß-catenin, and p120-catenin with the E-cadherin cytoplasmic tail alone in order to analyze the affinities of their binding interactions. We find that p120-catenin does not associate strongly with α- or ß-catenin and binds much more transiently to the cadherin cytoplasmic tail than does ß-catenin. Overall, this work creates many new possibilities for biochemical studies understanding transmembrane signaling of cadherins and the role of p120-catenin in adhesion activation.


Subject(s)
Cadherins , Catenins , Cadherins/genetics , Cadherins/metabolism , Catenins/genetics , Catenins/metabolism , Cell Adhesion/physiology , Cell Membrane/metabolism , Phosphoproteins , Signal Transduction , beta Catenin/genetics , beta Catenin/metabolism
7.
Tissue Barriers ; 9(4): 1940741, 2021 10 02.
Article in English | MEDLINE | ID: mdl-34402758

ABSTRACT

Deficits in gastrointestinal (GI) paracellular permeability has been implicated in etiology of Inflammatory Bowel Disease (IBD), and E-cadherin, a key component of the epithelial junctional complex, has been implicated in both barrier function and IBD. We have previously described antibodies against E-cadherin that activate cell adhesion, and in this study, we show that they increase transepithelial electrical resistance in epithelial cell monolayers in vitro. We therefore tested the hypothesis that adhesion activating E-cadherin mAbs will enhance epithelial barrier function in vivo and limit progression of inflammation in IBD. Activating mAbs to mouse E-cadherin were tested in different mouse models of IBD including the IL10-/- and adoptive T cell transfer models of colitis. Previously established histological and biomarker measures of inflammation were evaluated to monitor disease progression. Mouse E-cadherin activating mAb treatment reduced total colitis score, individual histological measures of inflammation, and other hallmarks of inflammation compared to control treatment. Activating mAbs also reduced the fecal accumulation lipocalin2 and albumin content, consistent with enhanced barrier function. Therefore, E-cadherin activation could be a potential strategy for limiting inflammation in UC.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Animals , Cadherins/metabolism , Colitis/metabolism , Colitis/pathology , Inflammation/metabolism , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/metabolism , Mice
8.
Am J Physiol Heart Circ Physiol ; 320(4): H1403-H1410, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33577432

ABSTRACT

Excessive vascular permeability occurs in inflammatory disease processes. Vascular endothelial cadherin (VE-cadherin) is an adhesion protein that controls vascular permeability. We identified monoclonal antibodies (mAbs) to human VE-cadherin that activate cell adhesion and inhibit the increased permeability of endothelial cell monolayers induced by thrombin receptor activator peptide-6 (TRAP-6). Two mAbs, 8A12c and 3A5a, reduce permeability, whereas an inhibitory mAb, 2E11d, enhances permeability. Activating mAbs also reduce permeability induced by tumor necrosis factor-α (TNF-α) and vascular endothelial cell growth factor (VEGF). The activating mAbs also stabilize the organization of the adherens junctions that are disrupted by TRAP-6, VEGF, or TNF-α. The activating mAbs act directly on the adhesive function of VE-cadherin because they did not block the accumulation of actin filaments stimulated by TRAP-6 and enhance physical cell-cell adhesion of VE-cadherin-expressing tissue culture cells. Therefore, VE-cadherin function can be regulated at the cell surface to control endothelial permeability.NEW & NOTEWORTHY Excessive vascular permeability is a serious complication of many inflammatory disease conditions. We have developed monoclonal antibodies that inhibit increases in endothelial monolayer permeability induced by several signaling factors by activating VE-cadherin mediated adhesion and stabilizing cell junctions. These antibodies and/or the mechanisms they reveal may lead to important therapeutics to treat vascular leakiness and inflammation.


Subject(s)
Adherens Junctions/drug effects , Antibodies, Monoclonal/pharmacology , Cadherins/agonists , Capillary Permeability/drug effects , Cell Adhesion/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Adherens Junctions/metabolism , Antigens, CD/metabolism , Cadherins/antagonists & inhibitors , Cadherins/metabolism , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Nocodazole/pharmacology , Oligopeptides/pharmacology , Receptors, Thrombin/agonists , Receptors, Thrombin/metabolism , Signal Transduction , Tubulin Modulators/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
9.
PLoS One ; 15(6): e0235337, 2020.
Article in English | MEDLINE | ID: mdl-32589661

ABSTRACT

p120-catenin is considered to be a tumor suppressor because it stabilizes E-cadherin levels at the cell surface. p120-catenin phosphorylation is increased in several types of cancer, but the role of phosphorylation in cancer is unknown. The phosphorylation state of p120-catenin is important in controlling E-cadherin homophilic binding strength which maintains epithelial junctions. Because decreased cell-cell adhesion is associated with increased cancer metastasis we hypothesize that p120-catenin phosphorylation at specific Serine and Threonine residues alters the E-cadherin binding strength between tumor cells and thereby affect the ability of tumor cells to leave the primary tumor and metastasize to distant sites. In this study we show that expression of the p120-catenin phosphorylation dead mutant, by converting six Serine and Threonine sites to Alanine, leads to enhanced E-cadherin adhesive binding strength in tumor cells. We observed a decrease in the ability of tumor cells expressing the p120-catenin phosphorylation mutant to migrate and invade using in-vitro models of cancer progression. Further, tumor cells expressing the phosphorylation mutant form of p120-catenin demonstrated a decrease in ability to metastasize to the lungs using an in-vivo orthotopic mammary fat pad injection model of breast cancer development and metastasis. This suggests that regulation of p120-catenin phosphorylation at the cell surface is important in mediating cell-adhesion, thereby impacting cancer progression and metastasis.


Subject(s)
Cadherins/metabolism , Catenins/metabolism , Cell Adhesion , Animals , Catenins/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Mice , Mutation , Neoplasm Invasiveness , Neoplasm Metastasis , Phosphorylation , Delta Catenin
10.
Proc Natl Acad Sci U S A ; 117(11): 5931-5937, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32127478

ABSTRACT

E-cadherin is a tumor suppressor protein, and the loss of its expression in association with the epithelial mesenchymal transition (EMT) occurs frequently during tumor metastasis. However, many metastases continue to express E-cadherin, and a full EMT is not always necessary for metastasis; also, positive roles for E-cadherin expression in metastasis have been reported. We hypothesize instead that changes in the functional activity of E-cadherin expressed on tumor cells in response to environmental factors is an important determinant of the ability of the tumor cells to metastasize. We find that E-cadherin expression persists in metastatic lung nodules and circulating tumor cells (CTCs) in two mouse models of mammary cancer: genetically modified MMTV-PyMT mice and orthotopically grafted 4T1 tumor cells. Importantly, monoclonal antibodies that bind to and activate E-cadherin at the cell surface reduce lung metastasis from endogenous genetically driven tumors and from tumor cell grafts. E-cadherin activation inhibits metastasis at multiple stages, including the accumulation of CTCs from the primary tumor and the extravasation of tumor cells from the vasculature. These activating mAbs increase cell adhesion and reduce cell invasion and migration in both cell culture and three-dimensional spheroids grown from primary tumors. Moreover, activating mAbs increased the frequency of apoptotic cells without affecting proliferation. Although the growth of the primary tumors was unaffected by activating mAbs, CTCs and tumor cells in metastatic nodules exhibited increased apoptosis. Thus, the functional state of E-cadherin is an important determinant of metastatic potential beyond whether the gene is expressed.


Subject(s)
Breast Neoplasms/metabolism , Cadherins/antagonists & inhibitors , Cadherins/metabolism , Lung Neoplasms/metabolism , Neoplasm Metastasis , Animals , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cadherins/genetics , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Disease Models, Animal , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplasm Metastasis/drug therapy , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Neoplastic Processes
11.
Proc Natl Acad Sci U S A ; 116(20): 9877-9882, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31043565

ABSTRACT

The Hippo pathway is involved in regulating contact inhibition of proliferation and organ size control and responds to various physical and biochemical stimuli. It is a kinase cascade that negatively regulates the activity of cotranscription factors YAP and TAZ, which interact with DNA binding transcription factors including TEAD and activate the expression of target genes. In this study, we show that the palmitoylation of TEAD, which controls the activity and stability of TEAD proteins, is actively regulated by cell density independent of Lats, the key kinase of the Hippo pathway. The expression of fatty acid synthase and acetyl-CoA carboxylase involved in de novo biosynthesis of palmitate is reduced by cell density in an Nf2/Merlin-dependent manner. Depalmitoylation of TEAD is mediated by depalmitoylases including APT2 and ABHD17A. Palmitoylation-deficient TEAD4 mutant is unstable and degraded by proteasome through the activity of the E3 ubiquitin ligase CHIP. These findings show that TEAD activity is tightly controlled through the regulation of palmitoylation and stability via the orchestration of FASN, depalmitoylases, and E3 ubiquitin ligase in response to cell contact.


Subject(s)
DNA-Binding Proteins/metabolism , Fatty Acid Synthase, Type I/metabolism , Lipoylation , Neurofibromin 2/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Humans , Muscle Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , TEA Domain Transcription Factors
12.
Oncogene ; 37(35): 4769-4780, 2018 08.
Article in English | MEDLINE | ID: mdl-29780167

ABSTRACT

E-cadherin is a key component of the adherens junctions that are integral in cell adhesion and maintaining epithelial phenotype of cells. Homophilic E-cadherin binding between cells is important in mediating contact inhibition of proliferation when cells reach confluence. Loss of E-cadherin expression results in loss of contact inhibition and is associated with increased cell motility and advanced stages of cancer. In this review we discuss the role of E-cadherin and its downstream signaling in regulation of contact inhibition and the development and progression of cancer.


Subject(s)
Cadherins/metabolism , Contact Inhibition/physiology , Neoplasms/metabolism , Neoplasms/pathology , Animals , Cell Adhesion/physiology , Cell Movement/physiology , Disease Progression , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Signal Transduction/physiology
13.
Mol Biol Cell ; 27(21): 3233-3244, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27582386

ABSTRACT

The loss of E-cadherin expression in association with the epithelial-mesenchymal transition (EMT) occurs frequently during tumor metastasis. However, metastases often retain E-cadherin expression, an EMT is not required for metastasis, and metastases can arise from clusters of tumor cells. We demonstrate that the regulation of the adhesive activity of E-cadherin present at the cell surface by an inside-out signaling mechanism is important in cancer. First, we find that the metastasis of an E-cadherin-expressing mammary cell line from the mammary gland to the lung depends on reduced E-cadherin adhesive function. An activating monoclonal antibody to E-cadherin that induces a high adhesive state significantly reduced the number of cells metastasized to the lung without affecting the growth in size of the primary tumor in the mammary gland. Second, we find that many cancer-associated germline missense mutations in the E-cadherin gene in patients with hereditary diffuse gastric cancer selectively affect the mechanism of inside-out cell surface regulation without inhibiting basic E-cadherin adhesion function. This suggests that genetic deficits in E-cadherin cell surface regulation contribute to cancer progression. Analysis of these mutations also provides insights into the molecular mechanisms underlying cadherin regulation at the cell surface.


Subject(s)
Cadherins/genetics , Cadherins/metabolism , Animals , Cadherins/physiology , Cell Adhesion/physiology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic/physiology , Humans , Mammary Glands, Human/metabolism , Mice , Mice, Inbred BALB C , Mutation, Missense , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Receptors, Cell Surface , Signal Transduction , Transcription Factors/metabolism
14.
Mamm Genome ; 27(11-12): 556-564, 2016 12.
Article in English | MEDLINE | ID: mdl-27601049

ABSTRACT

The Hippo-YAP pathway mediates organ size control, contact inhibition, and tumorigenesis. It is a kinase cascade that inhibits the nuclear localization and transcriptional activities of YAP and TAZ. E-cadherin, cell junctions, polarity proteins, and the merlin/NF2 tumor suppressor activate the pathway to inhibit YAP/TAZ activity, while growth factor signaling inhibits the pathway to activate YAP/TAZ in the nucleus. We examined its role in the development of mouse mammary glands and tumor formation using gland reconstitution by transplantation of genetically modified mammary stem cells (MaSCs). Knockdown of YAP and TAZ with shRNA in MaSCs did not inhibit gland reconstitution. In contrast, knockdown of ß-catenin blocked gland reconstitution, consistent with the known role of Wnt signaling in mammary gland development. However, we find that Hippo signaling is involved in mammary tumor formation. Expression of a constitutively active form of YAP caused rapid formation of large tumors. Moreover, knockdown of YAP/TAZ slowed the development of tumors in polyoma middle T transgenic mice, a well-studied mammary tumor model involving activation of several signaling pathways. YAP accumulated in nuclei of mammary glands in ErbB2/EGFR-transgenic mice, suggesting that EGFR signaling affects YAP in vivo similar to cell culture. ErbB2/EGFR-transgenic mice develop mammary tumors in 7-8 months, but surprisingly, MaSCs from these mice did not form tumors when transplanted into host mice. Nonetheless, expression of dominant-negative Lats, which inhibits Hippo signaling, leads to tumor formation in ErbB2-transgenic mice, suggesting that Hippo signaling is involved in EGFR-induced mammary tumorigenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Breast Neoplasms/genetics , Carcinogenesis/genetics , ErbB Receptors/genetics , Phosphoproteins/genetics , Receptor, ErbB-2/genetics , Acyltransferases , Animals , Breast Neoplasms/pathology , Cell Cycle Proteins , Female , Hippo Signaling Pathway , Humans , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mice , Mice, Transgenic , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , Transcription Factors/genetics , Wnt Signaling Pathway/genetics , YAP-Signaling Proteins , beta Catenin/genetics
15.
PLoS One ; 11(2): e0148574, 2016.
Article in English | MEDLINE | ID: mdl-26845024

ABSTRACT

Tight regulation of cadherin-mediated intercellular adhesions is critical to both tissue morphogenesis during development and tissue homeostasis in adults. Cell surface expression of the cadherin-catenin complex is often directly correlated with the level of adhesion, however, examples exist where cadherin appears to be inactive and cells are completely non-adhesive. The state of p120-catenin phosphorylation has been implicated in regulating the adhesive activity of E-cadherin but the mechanism is currently unclear. We have found that destabilization of the microtubule cytoskeleton, independent of microtubule plus-end dynamics, dephosphorylates p120-catenin and activates E-cadherin adhesion in Colo 205 cells. Through chemical screening, we have also identified several kinases as potential regulators of E-cadherin adhesive activity. Analysis of several p120-catenin phosphomutants suggests that gross dephosphorylation of p120-catenin rather than that of specific amino acids may trigger E-cadherin adhesion. Uncoupling p120-catenin binding to E-cadherin at the membrane causes constitutive adhesion in Colo 205 cells, further supporting an inhibitory role of phosphorylated p120-catenin on E-cadherin activity.


Subject(s)
Cadherins/metabolism , Catenins/metabolism , Colonic Neoplasms/metabolism , Microtubules/metabolism , Catenins/genetics , Cell Adhesion , Cell Line, Tumor , Colonic Neoplasms/genetics , Humans , Mutation , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Delta Catenin
16.
J Cell Biol ; 210(3): 503-15, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26216901

ABSTRACT

The Hippo pathway is involved in the regulation of contact inhibition of proliferation and responses to various physical and chemical stimuli. Recently, several upstream negative regulators of Hippo signaling, including epidermal growth factor receptor ligands and lysophosphatidic acid, have been identified. We show that fibronectin adhesion stimulation of focal adhesion kinase (FAK)-Src signaling is another upstream negative regulator of the Hippo pathway. Inhibition of FAK or Src in MCF-10A cells plated at low cell density prevented the activation of Yes-associated protein (YAP) in a large tumor suppressor homologue (Lats)-dependent manner. Attachment of serum-starved MCF-10A cells to fibronectin, but not poly-d-lysine or laminin, induced YAP nuclear accumulation via the FAK-Src-phosphatidylinositol 4,5-bisphosphate 3-kinase (PI3K) signaling pathway. Attenuation of FAK, Src, PI3K, or PDK1 activity blocked YAP nuclear accumulation stimulated by adhesion to fibronectin. This negative regulation of the Hippo pathway by fibronectin adhesion signaling can, at least in part, explain the effects of cell spreading on YAP nuclear localization and represents a Lats-dependent component of the response to cell adhesion.


Subject(s)
Fibronectins/metabolism , Focal Adhesion Kinase 1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , src-Family Kinases/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Cell Line , Cell Proliferation/physiology , Contact Inhibition/physiology , Focal Adhesion Kinase 1/genetics , Hippo Signaling Pathway , Humans , Laminin/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphoproteins/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Polylysine/metabolism , Protein Serine-Threonine Kinases/genetics , RNA Interference , RNA, Small Interfering , Signal Transduction , Transcription Factors , YAP-Signaling Proteins , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
17.
J Biol Chem ; 290(35): 21749-61, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26175155

ABSTRACT

Cadherins are transmembrane adhesion proteins that maintain intercellular cohesion in all tissues, and their rapid regulation is essential for organized tissue remodeling. Despite some evidence that cadherin adhesion might be allosterically regulated, testing of this has been hindered by the difficulty of quantifying altered E-cadherin binding affinity caused by perturbations outside the ectodomain binding site. Here, measured kinetics of cadherin-mediated intercellular adhesion demonstrated quantitatively that treatment with activating, anti-E-cadherin antibodies or the dephosphorylation of a cytoplasmic binding partner, p120(ctn), increased the homophilic binding affinity of E-cadherin. Results obtained with Colo 205 cells, which express inactive E-cadherin and do not aggregate, demonstrated that four treatments, which induced Colo 205 aggregation and p120(ctn) dephosphorylation, triggered quantitatively similar increases in E-cadherin affinity. Several processes can alter cell aggregation, but these results directly demonstrated the allosteric regulation of cell surface E-cadherin by p120(ctn) dephosphorylation.


Subject(s)
Cadherins/metabolism , Allosteric Regulation/drug effects , Animals , Antibodies, Neutralizing/pharmacology , Catenins/metabolism , Cell Adhesion/drug effects , Dogs , Humans , Imaging, Three-Dimensional , Kinetics , Lithium Chloride/pharmacology , MCF-7 Cells , Mice , Models, Biological , Mutant Proteins/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Multimerization , Rats , Staurosporine/pharmacology , Delta Catenin
18.
Biochem Biophys Res Commun ; 459(3): 504-8, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25747715

ABSTRACT

Cadherin-6B induces bone morphogenetic protein (BMP) signaling to promote the epithelial mesenchymal transition (EMT) in the neural crest. We have previously found that knockdown of Cadherin-6B inhibits both BMP signaling and the emigration of the early pre-migratory neural crest cells from the dorsal neural tube. In this study, we found that inhibition of BMP signaling in the neural tube, mediated by the ectopic expression of Smad-6 or Noggin, decreased the size of the Islet-1-positive dorsal cell population. Knockdown or loss of function of Cadherin-6B suppressed the generation of Islet-1-expressing cells in the dorsal neural tube, but not the Lim-1/2 positive dorsal cell population. Our results thus indicate that Cadherin-6B is necessary for the generation of Islet-1-positive dorsal interneurons, as well as the initiation of pre-migratory neural crest cell emigration.


Subject(s)
Avian Proteins/metabolism , Cadherins/metabolism , Interneurons/metabolism , LIM-Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Avian Proteins/antagonists & inhibitors , Avian Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cadherins/antagonists & inhibitors , Cadherins/genetics , Carrier Proteins/metabolism , Cell Movement , Chick Embryo , Epithelial-Mesenchymal Transition , Gene Knockdown Techniques , Neural Crest/cytology , Neural Crest/embryology , Neural Crest/metabolism , Neural Tube/cytology , Neural Tube/embryology , Neural Tube/metabolism , Signal Transduction , Smad6 Protein/metabolism
19.
Tumour Biol ; 36(5): 3549-56, 2015 May.
Article in English | MEDLINE | ID: mdl-25542234

ABSTRACT

Epithelial-mesenchymal transition (EMT) of mammary epithelial cells is important in both normal morphogenesis of mammary glands and metastasis of breast cancer. Cadherin switching from E-cadherin to N-cadherin plays important roles in EMT. We found that cadherin switching is important in bone morphogenetic protein 4 (BMP4)-induced EMT in MCF-10A cells. BMP4 increased the phosphorylation of SMAD proteins in MCF-10A cells. Canonical BMP4 signaling decreased the expression of E-cadherin and disrupted the polarity of the tight junction protein ZO-1 in MCF-10A cells. However, the expression of N-cadherin and SNAI2 was up-regulated in BMP4-treated MCF-10A cells. MCF-10A cells that expressed N-cadherin migrated into type I collagen gels in response to BMP4 when evaluated using three-dimensional culture assays. Thus, active canonical BMP4 signaling is important for the migration and EMT of mammary epithelial cells. Moreover, the decrease in E-cadherin and/or increase in N-cadherin may be required for BMP4-induced migration and EMT.


Subject(s)
Bone Morphogenetic Protein 4/genetics , Breast Neoplasms/genetics , Cadherins/biosynthesis , Epithelial-Mesenchymal Transition/genetics , Breast Neoplasms/pathology , Cadherins/genetics , Cell Line, Tumor , Cell Movement/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Mammary Glands, Human/pathology , Morphogenesis/genetics , Neoplasm Metastasis , Zonula Occludens-1 Protein/genetics
20.
J Cell Sci ; 127(Pt 4): 709-17, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24532814

ABSTRACT

The Hippo-YAP pathway mediates the control of cell proliferation by contact inhibition as well as other attributes of the physical state of cells in tissues. Several mechanisms sense the spatial and physical organization of cells, and function through distinct upstream modules to stimulate Hippo-YAP signaling: adherens junction or cadherin-catenin complexes, epithelial polarity and tight junction complexes, the FAT-Dachsous morphogen pathway, as well as cell shape, actomyosin or mechanotransduction. Soluble extracellular factors also regulate Hippo pathway signaling, often inhibiting its activity. Indeed, the Hippo pathway mediates a reciprocal relationship between contact inhibition and mitogenic signaling. As a result, cells at the edges of a colony, a wound in a tissue or a tumor are more sensitive to ambient levels of growth factors and more likely to proliferate, migrate or differentiate through a YAP and/or TAZ-dependent process. Thus, the Hippo-YAP pathway senses and responds to the physical organization of cells in tissues and coordinates these physical cues with classic growth-factor-mediated signaling pathways. This Commentary is focused on the biological significance of Hippo-YAP signaling and how upstream regulatory modules of the pathway interact to produce biological outcomes.


Subject(s)
Cell Proliferation , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factors/metabolism , Animals , Cell Adhesion , Cell Communication , Cell Cycle Proteins , Cell Polarity , Hippo Signaling Pathway , Humans , Intercellular Signaling Peptides and Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...