201
|
Alcaide P, Newton G, Auerbach S, Sehrawat S, Mayadas TN, Golan DE, Yacono P, Vincent P, Kowalczyk A, Luscinskas FW. p120-Catenin regulates leukocyte transmigration through an effect on VE-cadherin phosphorylation. Blood 2008; 112:2770-9. [PMID: 18641366 PMCID: PMC2556612 DOI: 10.1182/blood-2008-03-147181] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Accepted: 07/07/2008] [Indexed: 12/13/2022] Open
Abstract
Vascular endothelial-cadherin (VE-cad) is localized to adherens junctions at endothelial cell borders and forms a complex with alpha-, beta-, gamma-, and p120-catenins (p120). We previously showed that the VE-cad complex disassociates to form short-lived "gaps" during leukocyte transendothelial migration (TEM); however, whether these gaps are required for leukocyte TEM is not clear. Recently p120 has been shown to control VE-cad surface expression through endocytosis. We hypothesized that p120 regulates VE-cad surface expression, which would in turn have functional consequences for leukocyte transmigration. Here we show that endothelial cells transduced with an adenovirus expressing p120GFP fusion protein significantly increase VE-cad expression. Moreover, endothelial junctions with high p120GFP expression largely prevent VE-cad gap formation and neutrophil leukocyte TEM; if TEM occurs, the length of time required is prolonged. We find no evidence that VE-cad endocytosis plays a role in VE-cad gap formation and instead show that this process is regulated by changes in VE-cad phosphorylation. In fact, a nonphosphorylatable VE-cad mutant prevented TEM. In summary, our studies provide compelling evidence that VE-cad gap formation is required for leukocyte transmigration and identify p120 as a critical intracellular mediator of this process through its regulation of VE-cad expression at junctions.
Collapse
Affiliation(s)
- Pilar Alcaide
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
202
|
Abu-Elneel K, Ochiishi T, Medina M, Remedi M, Gastaldi L, Caceres A, Kosik KS. A delta-catenin signaling pathway leading to dendritic protrusions. J Biol Chem 2008; 283:32781-91. [PMID: 18809680 DOI: 10.1074/jbc.m804688200] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Delta-catenin is a synaptic adherens junction protein pivotally positioned to serve as a signaling sensor and integrator. Expression of delta-catenin induces filopodia-like protrusions in neurons. Here we show that the small GTPases of the Rho family act coordinately as downstream effectors of delta-catenin. A dominant negative Rac prevented delta-catenin-induced protrusions, and Cdc42 activity was dramatically increased by delta-catenin expression. A kinase dead LIMK (LIM kinase) and a mutant Cofilin also prevented delta-catenin-induced protrusions. To link the effects of delta-catenin to a physiological pathway, we noted that (S)-3,5-dihydroxyphenylglycine (DHPG) activation of metabotropic glutamate receptors induced dendritic protrusions that are very similar to those induced by delta-catenin. Furthermore, delta-catenin RNA-mediated interference can block the induction of dendritic protrusions by DHPG. Interestingly, DHPG dissociated PSD-95 and N-cadherin from the delta-catenin complex, increased the association of delta-catenin with Cortactin, and induced the phosphorylation of delta-catenin within the sites that bind to these protein partners.
Collapse
Affiliation(s)
- Kawther Abu-Elneel
- Neuroscience Research Institute, and Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, California 93106, USA
| | | | | | | | | | | | | |
Collapse
|
203
|
Reintsch WE, Mandato CA, McCrea PD, Fagotto F. Inhibition of cell adhesion by xARVCF indicates a regulatory function at the plasma membrane. Dev Dyn 2008; 237:2328-41. [PMID: 18729204 DOI: 10.1002/dvdy.21651] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The cytoplasmic tail of cadherins is thought to regulate the strength and dynamics of cell-cell adhesion. Part of its regulatory activity has been attributed to a membrane-proximal region, the juxtamembrane domain (JMD), and its interaction with members of the p120 catenin subfamily. We show that titration of xARVCF, a member of this family, to the plasma membrane disrupts adhesion in the early embryo. Adhesion can be restored by coexpression of constitutively active Rac, suggesting that intracellular signaling is the primary cause in the loss of adhesion phenotype. Our observations suggest that the recruitment of p120 type catenins to the plasma membrane by the cadherin cytoplasmic tail may create protein complexes, which actively modulate the adhesion "status" of embryonic cells.
Collapse
|
204
|
Guillemot L, Paschoud S, Jond L, Foglia A, Citi S. Paracingulin regulates the activity of Rac1 and RhoA GTPases by recruiting Tiam1 and GEF-H1 to epithelial junctions. Mol Biol Cell 2008; 19:4442-53. [PMID: 18653465 DOI: 10.1091/mbc.e08-06-0558] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Small GTPases control key cellular events, including formation of cell-cell junctions and gene expression, and are regulated by activating and inhibiting factors. Here, we characterize the junctional protein paracingulin as a novel regulator of the activity of two small GTPases, Rac1 and RhoA, through the functional interaction with their respective activators, Tiam1 and GEF-H1. In confluent epithelial monolayers, paracingulin depletion leads to increased RhoA activity and increased expression of mRNA for the tight junction protein claudin-2. During tight junction assembly by the calcium-switch, Rac1 shows two transient peaks of activity, at earlier (10-20 min) and later (3-8 h) time points. Paracingulin depletion reduces such peaks of Rac1 activation in a Tiam1-dependent manner, resulting in a delay in junction formation. Paracingulin physically interacts with GEF-H1 and Tiam1 in vivo and in vitro, and it is required for their efficient recruitment to junctions, based on immunofluorescence and biochemical experiments. Our results provide the first description of a junctional protein that interacts with GEFs for both Rac1 and RhoA, and identify a novel molecular mechanism whereby Rac1 is activated during junction formation.
Collapse
Affiliation(s)
- Laurent Guillemot
- Department of Molecular Biology, University of Geneva, CH-1211 Geneva, Switzerland
| | | | | | | | | |
Collapse
|
205
|
Ogden SR, Wroblewski LE, Weydig C, Romero-Gallo J, O'Brien DP, Israel DA, Krishna US, Fingleton B, Reynolds AB, Wessler S, Peek RM. p120 and Kaiso regulate Helicobacter pylori-induced expression of matrix metalloproteinase-7. Mol Biol Cell 2008; 19:4110-21. [PMID: 18653469 DOI: 10.1091/mbc.e08-03-0283] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Helicobacter pylori is the strongest known risk factor for gastric adenocarcinoma, yet only a fraction of infected persons develop cancer. One H. pylori constituent that augments disease risk is the cytotoxin-associated gene (cag) pathogenicity island, which encodes a secretion system that translocates bacterial effector molecules into host cells. Matrix metalloproteinase (MMP)-7, a member of a family of enzymes with tumor-initiating properties, is overexpressed in premalignant and malignant gastric lesions, and H. pylori cag(+) strains selectively increase MMP-7 protein levels in gastric epithelial cells in vitro and in vivo. We now report that H. pylori-mediated mmp-7 induction is transcriptionally regulated via aberrant activation of p120-catenin (p120), a component of adherens junctions. H. pylori increases mmp-7 mRNA levels in a cag- and p120-dependent manner and induces translocation of p120 to the nucleus in vitro and in a novel ex vivo gastric gland culture system. Nuclear translocation of p120 in response to H. pylori relieves Kaiso-mediated transcriptional repression of mmp-7, which is implicated in tumorigenesis. These results indicate that selective and coordinated induction of mmp-7 expression by H. pylori cag(+) isolates may explain in part the augmentation in gastric cancer risk associated with these strains.
Collapse
Affiliation(s)
- Seth R Ogden
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232-2279, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
206
|
Wheelock MJ, Shintani Y, Maeda M, Fukumoto Y, Johnson KR. Cadherin switching. J Cell Sci 2008; 121:727-35. [PMID: 18322269 DOI: 10.1242/jcs.000455] [Citation(s) in RCA: 644] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The cadherin molecules at adherens junctions have multiple isoforms. Cadherin isoform switching (cadherin switching) occurs during normal developmental processes to allow cell types to segregate from one another. Tumor cells often recapitulate this activity and the result is an aggressive tumor cell that gains the ability to leave the site of the tumor and metastasize. At present, we understand some of the mechanisms that promote cadherin switching and some of the pathways downstream of this process that influence cell behavior. Specific cadherin family members influence growth-factor-receptor signaling and Rho GTPases to promote cell motility and invasion. In addition, p120-catenin probably plays multiple roles in cadherin switching, regulating Rho GTPases and stabilizing cadherins.
Collapse
Affiliation(s)
- Margaret J Wheelock
- University of Nebraska Medical Center, Department of Oral Biology and Eppley Cancer Center, Omaha, NE 68198-7696, USA.
| | | | | | | | | |
Collapse
|
207
|
Kiss A, Troyanovsky RB, Troyanovsky SM. p120-catenin is a key component of the cadherin-gamma-secretase supercomplex. Mol Biol Cell 2008; 19:4042-50. [PMID: 18632982 DOI: 10.1091/mbc.e08-04-0394] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
In this work, we show several previously unknown features of p120-catenin in a cadherin-catenin complex that are critical for our understanding of cadherin-based adhesion and signaling. We show that in human epithelial A-431 cells, nearly all p120 molecules engage in high-affinity interaction with E-cadherin-catenin complexes located at the cellular surface. p120 is positioned in proximity to alpha-catenin in the complex with cadherin. These findings suggest a functional cooperation between p120 and alpha-catenin in cadherin-based adhesion. A low level of cadherin-free p120 molecules, in contrast, could facilitate p120-dependent signaling. Finally, we present compelling evidence that p120 is a key linker cementing the E-cadherin-catenin complex with the transmembrane protease gamma-secretase. The cell-cell contact location of this supercomplex makes it an important candidate for conducting different signals that rely on gamma-secretase proteolytic activity.
Collapse
Affiliation(s)
- Alexi Kiss
- Division of Dermatology, Washington University Medical School, St. Louis, MO 63110, USA
| | | | | |
Collapse
|
208
|
Nyqvist D, Giampietro C, Dejana E. Deciphering the functional role of endothelial junctions by using in vivo models. EMBO Rep 2008; 9:742-7. [PMID: 18600233 DOI: 10.1038/embor.2008.123] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2008] [Accepted: 05/23/2008] [Indexed: 11/09/2022] Open
Abstract
Endothelial cell-to-cell junctions are vital for the formation and integrity of blood vessels. The main adhesive junctional complexes in endothelial cells, adherens junctions and tight junctions, are formed by transmembrane adhesive proteins that are linked to intracellular signalling partners and cytoskeletal-binding proteins. Gene inactivation and blocking antibodies in mouse models have revealed some of the functions of the individual junctional components in vivo, and are increasing our understanding of the functional role of endothelial cell junctions in angiogenesis and vascular homeostasis. Adherens-junction organization is required for correct vascular morphogenesis during embryo development. By contrast, the data available suggest that tight-junction proteins are not essential for vascular development but are necessary for endothelial barrier function.
Collapse
Affiliation(s)
- Daniel Nyqvist
- IFOM, FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | | | | |
Collapse
|
209
|
Huang RYJ, Wang SM, Hsieh CY, Wu JC. Lysophosphatidic acid induces ovarian cancer cell dispersal by activating Fyn kinase associated with p120-catenin. Int J Cancer 2008; 123:801-9. [DOI: 10.1002/ijc.23579] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
210
|
Yanagisawa M, Huveldt D, Kreinest P, Lohse CM, Cheville JC, Parker AS, Copland JA, Anastasiadis PZ. A p120 catenin isoform switch affects Rho activity, induces tumor cell invasion, and predicts metastatic disease. J Biol Chem 2008; 283:18344-54. [PMID: 18407999 DOI: 10.1074/jbc.m801192200] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
p120 catenin is a cadherin-associated protein that regulates Rho GTPases and promotes the invasiveness of E-cadherin-deficient cancer cells. Multiple p120 isoforms are expressed in cells via alternative splicing, and all of them are essential for HGF signaling to Rac1. However, only full-length p120 (isoform 1) promotes invasiveness. This selective ability of p120 isoform 1 is mediated by reduced RhoA activity, both under basal conditions and following HGF treatment. All p120 isoforms can bind RhoA in vitro, via a central RhoA binding site. However, only the cooperative binding of RhoA to the central p120 domain and to the alternatively spliced p120 N terminus stabilizes RhoA binding and inhibits RhoA activity. Consistent with this, increased expression of p120 isoform 1, when compared with other p120 isoforms, is predictive of renal tumor micrometastasis and systemic progression, following nephrectomy. Furthermore, ectopic expression of the RhoA-binding, N-terminal domain of p120 is sufficient to block the ability of p120 isoform 1 to inhibit RhoA and to promote invasiveness. The data indicate that the increased expression of p120 isoform 1 during tumor progression contributes to the invasive phenotype of cadherin-deficient carcinomas and that the N-terminal domain of p120 is a valid therapeutic target.
Collapse
|
211
|
Vaughan MH, Xia X, Wang X, Chronopoulou E, Gao GJ, Campos-Gonzalez R, Reynolds AB. Generation and characterization of a novel phospho-specific monoclonal antibody to p120-catenin serine 879. Hybridoma (Larchmt) 2008; 26:407-15. [PMID: 18158786 DOI: 10.1089/hyb.2007.0527] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
To better understand the mechanisms that regulate p120-catenin (p120) and E-cadherin function, we are systematically generating phospho-specific monoclonal antibodies (MAb) to the major p120 phosphorylation sites. p120 has emerged recently as a master regulator of E-cadherin stability and an important modulator of RhoGTPase activities. A number of phosphorylation sites have been identified, but none have as yet been linked to specific regulatory roles. Here, we describe a novel phospho-specific monoclonal antibody to the major PKC-induced p120 phosphorylation site, phospho-serine 879 (pS879). With a few exceptions, p120 MAb pS879 is remarkably specific for the phosphorylated S879 epitope and works effectively in common applications such as Western blot analysis, immunoprecipitation, and immunofluorescence. p120 MAb pS879 should facilitate efforts to identify the role of S879 phosphorylation and to map signaling pathways that modify p120 function through activation of PKC.
Collapse
Affiliation(s)
- Meredith H Vaughan
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232-6840, USA
| | | | | | | | | | | | | |
Collapse
|
212
|
Niessen CM, Gottardi CJ. Molecular components of the adherens junction. BIOCHIMICA ET BIOPHYSICA ACTA 2008; 1778:562-71. [PMID: 18206110 PMCID: PMC2276178 DOI: 10.1016/j.bbamem.2007.12.015] [Citation(s) in RCA: 191] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/25/2007] [Revised: 12/13/2007] [Accepted: 12/17/2007] [Indexed: 12/24/2022]
Abstract
Adherens junctions serve to couple individual cells into various arrangements required for tissue structure and function. The central structural components of adherens junctions are transmembrane adhesion receptors, and their associated actin-binding/regulatory proteins. The molecular machineries that organize these adhesion receptor complexes into higher order junction structures, and the functional consequences of this junctional organization will be discussed.
Collapse
Affiliation(s)
- Carien M. Niessen
- Center for Molecular Medicine Cologne, University of Cologne, Germany
- Department of Dermatology, University of Cologne, Germany
| | - Cara J. Gottardi
- Department of Medicine, Feinberg School of Medicine, Northwestern University, USA
| |
Collapse
|
213
|
Larson DE, Liberman Z, Cagan RL. Cellular behavior in the developing Drosophila pupal retina. Mech Dev 2008; 125:223-32. [PMID: 18166433 PMCID: PMC2965056 DOI: 10.1016/j.mod.2007.11.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Revised: 11/16/2007] [Accepted: 11/17/2007] [Indexed: 02/03/2023]
Abstract
Correct patterning of cells within an epithelium is key to establishing their normal function. However, the precise mechanisms by which individual cells arrive at their final developmental niche remains poorly understood. We developed an optimized system for imaging the developing Drosophila retina, an ideal tissue for the study of cell positioning. Using this technique, we characterized the cellular dynamics of developing wild-type pupal retinas. We also analyzed two mutants affecting eye patterning and demonstrate that cells mutant for Notch or Roughest signaling were aberrantly dynamic in their cell movements. Finally, we establish a role for the adherens junction regulator P120-Catenin in retinal patterning through its regulation of normal adherens junction integrity. Our results indicate a requirement for P120-Catenin in the developing retina, the first reported developmental function of this protein in the epithelia of lower metazoa. Based upon our live visualization of the P120-Catenin mutant as well as genetic data, we conclude that P120-Catenin is acting to stabilize E-cadherin and adherens junction integrity during eye development.
Collapse
Affiliation(s)
- David E. Larson
- Department of Molecular Biology and Pharmacology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8103, Saint Louis, MO 63110, USA
| | - Zoe Liberman
- Department of Molecular Biology and Pharmacology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8103, Saint Louis, MO 63110, USA
| | - Ross L. Cagan
- Department of Molecular Biology and Pharmacology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8103, Saint Louis, MO 63110, USA
| |
Collapse
|
214
|
Hartsock A, Nelson WJ. Adherens and tight junctions: structure, function and connections to the actin cytoskeleton. BIOCHIMICA ET BIOPHYSICA ACTA 2008; 1778:660-9. [PMID: 17854762 PMCID: PMC2682436 DOI: 10.1016/j.bbamem.2007.07.012] [Citation(s) in RCA: 1106] [Impact Index Per Article: 65.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/15/2007] [Revised: 07/12/2007] [Accepted: 07/19/2007] [Indexed: 02/07/2023]
Abstract
Adherens junctions and Tight junctions comprise two modes of cell-cell adhesion that provide different functions. Both junctional complexes are proposed to associate with the actin cytoskeleton, and formation and maturation of cell-cell contacts involves reorganization of the actin cytoskeleton. Adherens junctions initiate cell-cell contacts, and mediate the maturation and maintenance of the contact. Adherens junctions consist of the transmembrane protein E-cadherin, and intracellular components, p120-catenin, beta-catenin and alpha-catenin. Tight junctions regulate the paracellular pathway for the movement of ions and solutes in-between cells. Tight junctions consist of the transmembrane proteins occludin and claudin, and the cytoplasmic scaffolding proteins ZO-1, -2, and -3. This review discusses the binding interactions of the most studied proteins that occur within each of these two junctional complexes and possible modes of regulation of these interactions, and the different mechanisms that connect and regulate interactions with the actin cytoskeleton.
Collapse
Affiliation(s)
- Andrea Hartsock
- Department of Molecular and Cellular Physiology, Stanford University
| | - W. James Nelson
- Department of Molecular and Cellular Physiology, Stanford University
- Department of Biological Sciences, Stanford University
| |
Collapse
|
215
|
Schnack C, Danzer KM, Hengerer B, Gillardon F. Protein array analysis of oligomerization-induced changes in alpha-synuclein protein-protein interactions points to an interference with Cdc42 effector proteins. Neuroscience 2008; 154:1450-7. [PMID: 18541383 DOI: 10.1016/j.neuroscience.2008.02.049] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2007] [Revised: 02/15/2008] [Accepted: 02/19/2008] [Indexed: 12/31/2022]
Abstract
Aggregation of alpha-synuclein may contribute to neuropathology in Parkinson's disease patients and in transgenic animal models. Natively unfolded alpha-synuclein binds to various proteins and conformational changes due to alpha-synuclein misfolding may alter physiological interactions. In the present study, we used protein arrays spotted with 5000 recombinant human proteins for a large scale interaction analysis of monomeric versus oligomeric alpha-synuclein. Monomeric alpha-synuclein bound to arrayed cAMP regulated phosphoprotein 19 and binding appears to be disrupted by alpha-synuclein oligomerization. Incubation with recombinant alpha-synuclein oligomers lead to the identification of several GTPase activating proteins and Cdc42 effector proteins as binding partners. Protein database searches revealed a Cdc42/Rac interactive binding domain in some interactors. To demonstrate in vivo relevance, we analyzed brainstem protein extracts from alpha-synuclein(A30P) transgenic mice. Pull-down assays using beads conjugated with a Cdc42/Rac interactive binding domain lead to an enrichment of endogenous alpha-synuclein oligomers. Cdc42 effector proteins were also co-immunoprecipitated with alpha-synuclein from brainstem lysates and were colocalized with alpha-synuclein aggregates in brain sections by double immunostaining. By two-dimensional gel electrophoretic analysis of synaptosomal fractions from transgenic mouse brains we detected additional isoforms of septin 6, a downstream target of Cdc42 effector proteins. Small GTPases have recently been identified in a genetic modifier screen to suppress alpha-synuclein toxicity in yeast. Our data indicate that components of small GTPase signal transduction pathways may be directly targeted by alpha-synuclein oligomers which potentially leads to signaling deficits and neurodegeneration.
Collapse
Affiliation(s)
- C Schnack
- Boehringer Ingelheim Pharma GmbH & Co KG, CNS Research, Biberach an der Riss, Germany
| | | | | | | |
Collapse
|
216
|
Kim H, Han JR, Park J, Oh M, James SE, Chang S, Lu Q, Lee KY, Ki H, Song WJ, Kim K. Delta-catenin-induced dendritic morphogenesis. An essential role of p190RhoGEF interaction through Akt1-mediated phosphorylation. J Biol Chem 2008; 283:977-87. [PMID: 17993462 PMCID: PMC2265781 DOI: 10.1074/jbc.m707158200] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Delta-catenin was first identified through its interaction with Presenilin-1 and has been implicated in the regulation of dendrogenesis and cognitive function. However, the molecular mechanisms by which delta-catenin promotes dendritic morphogenesis were unclear. In this study, we demonstrated delta-catenin interaction with p190RhoGEF, and the importance of Akt1-mediated phosphorylation at Thr-454 residue of delta-catenin in this interaction. We have also found that delta-catenin overexpression decreased the binding between p190RhoGEF and RhoA, and significantly lowered the levels of GTP-RhoA but not those of GTP-Rac1 and -Cdc42. Delta-catenin T454A, a defective form in p190RhoGEF binding, did not decrease the binding between p190RhoGEF and RhoA. Delta-catenin T454A also did not lower GTP-RhoA levels and failed to induce dendrite-like process formation in NIH 3T3 fibroblasts. Furthermore, delta-catenin T454A significantly reduced the length and number of mature mushroom shaped spines in primary hippocampal neurons. These results highlight signaling events in the regulation of delta-catenin-induced dendrogenesis and spine morphogenesis.
Collapse
Affiliation(s)
- Hangun Kim
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Korea
| | - Jeong Ran Han
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Korea
| | - Jaejun Park
- Department of Life Science, Gwangju Institute of Science and Technology, Gwangju 500-712, Korea
| | - Minsoo Oh
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Korea
| | - Sarah E. James
- Department of Anatomy and Cell Biology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, U.S.A
| | - Sunghoe Chang
- Department of Life Science, Gwangju Institute of Science and Technology, Gwangju 500-712, Korea
| | - Qun Lu
- Department of Anatomy and Cell Biology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, U.S.A
| | - Kwang Youl Lee
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Korea
| | - Hyunkyoung Ki
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Korea
| | - Woo-Joo Song
- Graduate Program in Neuroscience and Institute for Brain Science and Technology, Inje University, Daejeon, Korea
| | - Kwonseop Kim
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Korea
| |
Collapse
|
217
|
Fukumoto Y, Shintani Y, Reynolds AB, Johnson KR, Wheelock MJ. The regulatory or phosphorylation domain of p120 catenin controls E-cadherin dynamics at the plasma membrane. Exp Cell Res 2008; 314:52-67. [PMID: 17719574 PMCID: PMC2211447 DOI: 10.1016/j.yexcr.2007.07.024] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2007] [Revised: 07/16/2007] [Accepted: 07/17/2007] [Indexed: 10/23/2022]
Abstract
In contrast to growth factor-stimulated tyrosine phosphorylation of p120, its relatively constitutive serine/threonine phosphorylation is not well understood. Here we examined the role of serine/threonine phosphorylation of p120 in cadherin function. Expression of cadherins in cadherin-null cells converted them to an epithelial phenotype, induced p120 phosphorylation and localized it to sites of cell contact. Detergent solubility and immunofluorescence confirmed that phosphorylated p120 was at the plasma membrane. E-cadherin constructs incapable of traveling to the plasma membrane did not induce serine/threonine phosphorylation of p120, nor did cadherins constructs incapable of binding p120. However, an E-cadherin cytoplasmic domain construct artificially targeted to the plasma membrane did induce serine/threonine phosphorylation of p120, suggesting phosphorylation occurs independently of signals from cadherin dimerization and trafficking through the ER/Golgi. Solubility assays following calcium switch showed that p120 isoform 3A was more effective at stabilizing E-cadherin at the plasma membrane relative to isoform 4A. Since the major phosphorylation domain of p120 is included in isoform 3A but not 4A, we tested p120 mutated in the known phosphorylation sites in this domain and found that it was even less effective at stabilizing E-cadherin. These data suggest that serine/threonine phosphorylation of p120 influences the dynamics of E-cadherin in junctions.
Collapse
Affiliation(s)
- Yuri Fukumoto
- University of Nebraska Medical Center, Eppley Institute for Research in Cancer and Allied Diseases, Omaha, Nebraska 68198-7696
| | - Yasushi Shintani
- University of Nebraska Medical Center, Department of Oral Biology, Omaha, Nebraska 68198-7696
| | - Albert B. Reynolds
- Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232
| | - Keith R. Johnson
- University of Nebraska Medical Center, Department of Oral Biology, Omaha, Nebraska 68198-7696
- University of Nebraska Medical Center, Eppley Institute for Research in Cancer and Allied Diseases, Omaha, Nebraska 68198-7696
- University of Nebraska Medical Center, Eppley Cancer Center, Omaha, Nebraska 68198-7696
| | - Margaret J. Wheelock
- University of Nebraska Medical Center, Department of Oral Biology, Omaha, Nebraska 68198-7696
- University of Nebraska Medical Center, Eppley Institute for Research in Cancer and Allied Diseases, Omaha, Nebraska 68198-7696
- University of Nebraska Medical Center, Eppley Cancer Center, Omaha, Nebraska 68198-7696
| |
Collapse
|
218
|
Kiryushko D, Bock E, Berezin V. Pharmacology of cell adhesion molecules of the nervous system. Curr Neuropharmacol 2007; 5:253-67. [PMID: 19305742 PMCID: PMC2644493 DOI: 10.2174/157015907782793658] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Revised: 04/27/2007] [Accepted: 07/17/2007] [Indexed: 12/15/2022] Open
Abstract
Cell adhesion molecules (CAMs) play a pivotal role in the development and maintenance of the nervous system under normal conditions. They also are involved in numerous pathological processes such as inflammation, degenerative disorders, and cancer, making them attractive targets for drug development. The majority of CAMs are signal transducing receptors. CAM-induced intracellular signalling is triggered via homophilic (CAM-CAM) and heterophilic (CAM - other counter-receptors) interactions, which both can be targeted pharmacologically. We here describe the progress in the CAM pharmacology focusing on cadherins and CAMs of the immunoglobulin (Ig) superfamily, such as NCAM and L1. Structural basis of CAM-mediated cell adhesion and CAM-induced signalling are outlined. Different pharmacological approaches to study functions of CAMs are presented including the use of specific antibodies, recombinant proteins, and synthetic peptides. We also discuss how unravelling of the 3D structure of CAMs provides novel pharmacological tools for dissection of CAM-induced signalling pathways and offers therapeutic opportunities for a range of neurological disorders.
Collapse
Affiliation(s)
- Darya Kiryushko
- Protein Laboratory, Department of Neuroscience and Pharmacology, Panum Institute Bld. 6.2, Blegdamsvej 3C, DK-2200, Copenhagen N, Denmark.
| | | | | |
Collapse
|
219
|
Schmandke A, Schmandke A, Strittmatter SM. ROCK and Rho: biochemistry and neuronal functions of Rho-associated protein kinases. Neuroscientist 2007; 13:454-69. [PMID: 17901255 PMCID: PMC2849133 DOI: 10.1177/1073858407303611] [Citation(s) in RCA: 137] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Rho-associated protein kinases (ROCKs) play key roles in mediating the control of the actin cytoskeleton by Rho family GTPases in response to extracellular signals. Such signaling pathways contribute to diverse neuronal functions from cell migration to axonal guidance to dendritic spine morphology to axonal regeneration to cell survival. In this review, the authors summarize biochemical knowledge of ROCK function and categorize neuronal ROCK-dependent signaling pathways. Further study of ROCK signal transduction mechanisms and specificities will enhance our understanding of brain development, plasticity, and repair. The ROCK pathway also provides a potential site for therapeutic intervention to promote neuronal regeneration and to limit degeneration.
Collapse
Affiliation(s)
- André Schmandke
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Neurology Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | |
Collapse
|
220
|
Ichii T, Takeichi M. p120-catenin regulates microtubule dynamics and cell migration in a cadherin-independent manner. Genes Cells 2007; 12:827-39. [PMID: 17584295 DOI: 10.1111/j.1365-2443.2007.01095.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
p120-catenin (p120) has been shown to be essential for cadherin stability. Here, we show that p120 is capable of regulating microtubule (MT) dynamics in a cadherin-independent manner. When p120 was depleted in cadherin-deficient Neuro-2a (N2a) cells, MT stability was reduced, as assessed by the nocodazole sensitivity of MTs. On the contrary, over-expression of p120 caused MTs to become resistant to nocodazole. Time-lapse recording of GFP-tagged EB1, a protein which binds the growing plus-ends of MTs, introduced into these cells demonstrated that the plus ends underwent more frequent catastrophe in p120-depleted cells. In addition, p120 knockdown up-regulated the motility of isolated cells, whereas it down-regulated the directional migration of cells from wound edges; and these migratory behaviors of cells were mimicked by nocodazole-induced MT depolymerization. These results suggest that p120 has the ability to regulate MT dynamics and that this activity, in turn, affects cell motility independently of the cadherin adhesion system.
Collapse
Affiliation(s)
- Tetsuo Ichii
- Graduate School of Biostudies, Kyoto University, Yoshida-Honmachi, Sakyo-ku, Kyoto, 606-8501, Japan
| | | |
Collapse
|
221
|
Liu WF, Nelson CM, Tan JL, Chen CS. Cadherins, RhoA, and Rac1 are differentially required for stretch-mediated proliferation in endothelial versus smooth muscle cells. Circ Res 2007; 101:e44-52. [PMID: 17712140 DOI: 10.1161/circresaha.107.158329] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Abnormal mechanical forces can trigger aberrant proliferation of endothelial and smooth muscle cells, as observed in the progression of vascular diseases such as atherosclerosis. It has been previously shown that cells can sense physical forces such as stretch through adhesions to the extracellular matrix. Here, we set out to examine whether cell-cell adhesions are also involved in transducing mechanical stretch into a proliferative response. We found that both endothelial and smooth muscle cells exhibited an increase in proliferation in response to stretch. Using micropatterning to isolate the role of cell-cell adhesion from cell-extracellular matrix adhesion, we demonstrate that endothelial cells required cell-cell contact and vascular endothelial cadherin engagement to transduce stretch into proliferative signals. In contrast, smooth muscle cells responded to stretch without contact to neighboring cells. We further show that stretch stimulated Rac1 activity in endothelial cells, whereas RhoA was activated by stretch in smooth muscle cells. Blocking Rac1 signaling by pharmacological or adenoviral reagents abrogated the proliferative response to stretch in endothelial cells but not in smooth muscle cells. Conversely, blocking RhoA completely inhibited the proliferative response in smooth muscle cells but not in endothelial cells. Together, these data suggest that vascular endothelial cadherin has an important role in mechanotransduction and that endothelial and smooth muscle cells use different mechanisms to respond to stretch.
Collapse
Affiliation(s)
- Wendy F Liu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | |
Collapse
|
222
|
Yuan BZ, Jefferson AM, Millecchia L, Popescu NC, Reynolds SH. Morphological changes and nuclear translocation of DLC1 tumor suppressor protein precede apoptosis in human non-small cell lung carcinoma cells. Exp Cell Res 2007; 313:3868-80. [PMID: 17888903 DOI: 10.1016/j.yexcr.2007.08.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2007] [Revised: 07/17/2007] [Accepted: 08/08/2007] [Indexed: 11/19/2022]
Abstract
We have previously shown that reactivation of DLC1, a RhoGAP containing tumor suppressor gene, inhibits tumorigenicity of human non-small cell lung carcinoma cells (NSCLC). After transfection of NSCLC cells with wild type (WT) DLC1, changes in cell morphology were observed. To determine whether such changes have functional implications, we generated several DLC1 mutants and examined their effects on cell morphology, proliferation, migration and apoptosis in a DLC1 deficient NSCLC cell line. We show that WT DLC1 caused actin cytoskeleton-based morphological alterations manifested as cytoplasmic extensions and membrane blebbings in most cells. Subsequently, a fraction of cells exhibiting DLC1 protein nuclear translocation (PNT) underwent caspase 3-dependent apoptosis. We also show that the RhoGAP domain is essential for the occurrence of morphological alterations, PNT and apoptosis, and the inhibition of cell migration. DLC1 PNT is dependent on a bipartite nuclear localizing sequence and most likely is regulated by a serine-rich domain at N-terminal part of the DLC1 protein. Also, we found that DLC1 functions in the cytoplasm as an inhibitor of tumor cell proliferation and migration, but in the nucleus as an inducer of apoptosis. Our analyses provide evidence for a possible link between morphological alterations, PNT and proapoptotic and anti-oncogenic activities of DLC1 in lung cancer.
Collapse
Affiliation(s)
- Bao-Zhu Yuan
- Laboratory of Molecular Genetics, Toxicology and Molecular Biology Branch, National Institute for Occupational Safety and Health, CDC, Morgantown, WV 26505, USA.
| | | | | | | | | |
Collapse
|
223
|
Boguslavsky S, Grosheva I, Landau E, Shtutman M, Cohen M, Arnold K, Feinstein E, Geiger B, Bershadsky A. p120 catenin regulates lamellipodial dynamics and cell adhesion in cooperation with cortactin. Proc Natl Acad Sci U S A 2007; 104:10882-7. [PMID: 17576929 PMCID: PMC1904144 DOI: 10.1073/pnas.0702731104] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The armadillo-family protein, p120 catenin (p120), binds to the juxtamembrane domain of classical cadherins and increases cell-cell junction stability. Overexpression of p120 modulates the activity of Rho family GTPases and augments cell migratory ability. Here we show that down-regulation of p120 in epithelial MCF-7 cells by siRNA leads to a striking decrease in lamellipodial persistence and focal adhesion formation. Similar alterations in lamellipodial activity were observed in MCF-7 cells treated with siRNA to cortactin, an activator of Arp2/3-dependent actin polymerization. We found that, in many cell types, p120 is colocalized with cortactin-containing actin structures not only at cell-cell junctions, but also at extrajunctional sites including membrane ruffles and actin-rich halos around endocytotic vesicles. p120 depletion led to dramatic loss of cortactin and its partner, Arp3, from the cell leading edges. Cortactin and p120 are shown to directly interact with each other via the cortactin N-terminal region. We propose that the mechanism underlying p120 functions at the leading edge involves its cooperation with cortactin.
Collapse
Affiliation(s)
- Shlomit Boguslavsky
- *Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Inna Grosheva
- *Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Elad Landau
- *Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Michael Shtutman
- Cancer Center, Ordway Research Institute, Inc., Albany, NY 12208; and
| | - Miriam Cohen
- *Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Katya Arnold
- *Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | - Benjamin Geiger
- *Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Alexander Bershadsky
- *Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
224
|
Tao Q, Nandadasa S, McCrea PD, Heasman J, Wylie C. G-protein-coupled signals control cortical actin assembly by controlling cadherin expression in the early Xenopus embryo. Development 2007; 134:2651-61. [PMID: 17567666 DOI: 10.1242/dev.002824] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
During embryonic development, each cell of a multicellular organ rudiment polymerizes its cytoskeletal elements in an amount and pattern that gives the whole cellular population its characteristic shape and mechanical properties. How does each cell know how to do this? We have used the Xenopus blastula as a model system to study this problem. Previous work has shown that the cortical actin network is required to maintain shape and rigidity of the whole embryo, and its assembly is coordinated throughout the embryo by signaling through G-protein-coupled receptors. In this paper, we show that the cortical actin network colocalizes with foci of cadherin expressed on the cell surface. We then show that cell-surface cadherin expression is both necessary and sufficient for cortical actin assembly and requires the associated catenin p120 for this function. Finally, we show that the previously identified G-protein-coupled receptors control cortical actin assembly by controlling the amount of cadherin expressed on the cell surface. This identifies a novel mechanism for control of cortical actin assembly during development that might be shared by many multicellular arrays.
Collapse
Affiliation(s)
- Qinghua Tao
- Children's Hospital Research Foundation, Division of Developmental Biology, and Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, OH 45219, USA
| | | | | | | | | |
Collapse
|
225
|
Hosking CR, Ulloa F, Hogan C, Ferber EC, Figueroa A, Gevaert K, Birchmeier W, Briscoe J, Fujita Y. The transcriptional repressor Glis2 is a novel binding partner for p120 catenin. Mol Biol Cell 2007; 18:1918-27. [PMID: 17344476 PMCID: PMC1855037 DOI: 10.1091/mbc.e06-10-0941] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2006] [Revised: 02/06/2007] [Accepted: 02/27/2007] [Indexed: 11/11/2022] Open
Abstract
In epithelial cells, p120 catenin (p120) localizes at cell-cell contacts and regulates adhesive function of the cadherin complex. In addition, p120 has been reported to localize in the nucleus, although the nuclear function of p120 is not fully understood. Here, we report the identification of Gli-similar 2 (Glis2) as a novel binding protein for p120. Glis2 is a Krüppel-like transcriptional repressor with homology to the Gli family, but its physiological function has not been well characterized. In this study, we show that coexpression of Glis2 and Src induces nuclear translocation of p120. Furthermore, p120 induces the C-terminal cleavage of Glis2, and this cleavage is further enhanced by Src. The cleaved form of Glis2 loses one of its five zinc finger domains, but it is still able to bind DNA. Functional studies in chick neural tube indicate that full-length Glis2 can affect neuronal differentiation, whereas the cleaved form requires coexpression of p120 to have a similar effect. These data indicate that p120 has additional novel functions in the nucleus together with Glis2.
Collapse
Affiliation(s)
- Catherine Rose Hosking
- *Medical Research Council Laboratory for Molecular Cell Biology and Cell Biology Unit, and Department of Biology, University College London, London WC1E 6BT, United Kingdom
| | - Fausto Ulloa
- Division of Developmental Neurobiology, National Institute for Medical Research, London NW7 1AA, United Kingdom
| | - Catherine Hogan
- *Medical Research Council Laboratory for Molecular Cell Biology and Cell Biology Unit, and Department of Biology, University College London, London WC1E 6BT, United Kingdom
| | - Emma C. Ferber
- *Medical Research Council Laboratory for Molecular Cell Biology and Cell Biology Unit, and Department of Biology, University College London, London WC1E 6BT, United Kingdom
| | - Angélica Figueroa
- *Medical Research Council Laboratory for Molecular Cell Biology and Cell Biology Unit, and Department of Biology, University College London, London WC1E 6BT, United Kingdom
| | - Kris Gevaert
- Department of Medical Protein Research, Proteome Analysis and Bioinformatics Unit, Flanders Interuniversity Institute for Biotechnology, Faculty of Medicine and Health Sciences, Ghent University, B9000 Gent, Belgium; and
| | | | - James Briscoe
- Division of Developmental Neurobiology, National Institute for Medical Research, London NW7 1AA, United Kingdom
| | - Yasuyuki Fujita
- *Medical Research Council Laboratory for Molecular Cell Biology and Cell Biology Unit, and Department of Biology, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
226
|
Sampson HW, Dearman AC, Akintola AD, Zimmer WE, Parrish AR. Immunohistochemical localization of cadherin and catenin adhesion molecules in the murine growth plate. J Histochem Cytochem 2007; 55:845-52. [PMID: 17438349 DOI: 10.1369/jhc.7a7184.2007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mouse tibial growth plates were examined for the presence of adhesion molecules using immunohistochemistry and RT-PCR. All of the components of the classical cadherin/catenin complex (cadherin, alpha-, beta-, and gamma-catenin), as well as a heavy presence of p120, were identified in the murine growth plate. All of the major cadherins (1-5, 11, 13, and 15) were, for the first time, identified and localized in the murine growth plate. We have demonstrated that most of the cadherins and catenins reside in the zone of hypertrophy. Only alpha-catenin and E-, P-, R-, and VE-cadherin were found in all regions of the growth plate. The results for T-cadherin were inconclusive.
Collapse
Affiliation(s)
- H Wayne Sampson
- Department of Systems Biology and Translational Medicine, Texas A & M Health Science Center College of Medicine, 336 Reynolds Building, College Station, TX 77843-1114, USA.
| | | | | | | | | |
Collapse
|
227
|
Abstract
The catenin p120 is involved in many processes, including cell-cell adhesion and cancer. Recent work explores whether p120 independently regulates two key binding partners, RhoGTPase and cadherin.
Collapse
Affiliation(s)
- Donald T Fox
- Department of Biology and Lineberger Comprehensive Cancer Center, University of North Carolina Chapel Hill, North Carolina 27599-3280, USA
| | | |
Collapse
|
228
|
Brusés JL. N-cadherin signaling in synapse formation and neuronal physiology. Mol Neurobiol 2007; 33:237-52. [PMID: 16954598 DOI: 10.1385/mn:33:3:237] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2005] [Revised: 11/30/1999] [Accepted: 02/22/2006] [Indexed: 11/11/2022]
Abstract
Neural cadherin (N-cadherin) is an adhesion receptor that is localized in abundance at neuronto- neuron synapses. N-cadherin contains an extracellular domain that binds to other cadherins on juxtaposed cell membranes, a single-pass transmembrane region, and a cytoplasmic tail that interacts with various proteins, including catenins, kinases, phosphatases, and presenilin 1. N-cadherin contributes to the structural and functional organization of the synaptic complex by ensuring the adhesion between synaptic membranes and organizing the underlying actin cytoskeleton. Additionally, recent findings have shown that N-cadherin may participate in synaptic physiology by regulating calcium influx through voltage-activated calcium currents. The diverse activities of N-cadherin stem from its ability to operate as both an adhesion molecule that links cytoskeletons across cell membranes and a ligand-activated homophilic receptor capable of initiating intracellular signaling. An important mechanism of cadherin signaling is the regulation of small Rho guanosine triphosphatase activity that affects cytoskeleton dynamics and calcium influx. Because both the regulation of cadherin adhesive activity and cadherin-mediated signaling are affected by the binding of molecules to the intracellular domain, changes in the composition of the N-cadherin complex are central to the regulation of cadherin-mediated functions. This article focuses on the roles that N-cadherin might play at the level of the synapse through its effect on adhesion and signaling in the proximity of the synaptic junction.
Collapse
Affiliation(s)
- Juan L Brusés
- Department of Anatomy and Cell Biology, Kansas University Medical Center, Kansas City, KS, USA.
| |
Collapse
|
229
|
Abstract
Given the complexity of signaling pathways in the cell, it is a mystery how these pathways communicate with one other. In this issue of Cell, Wildenberg et al. (2006) reveal that the key effector molecule p120-catenin can mediate crosstalk between the Rac and Rho signaling pathways.
Collapse
Affiliation(s)
- Carien M Niessen
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Joseph Stelzmannstrasse 9, 50931 Cologne.
| | | |
Collapse
|
230
|
Wildenberg GA, Dohn MR, Carnahan RH, Davis MA, Lobdell NA, Settleman J, Reynolds AB. p120-catenin and p190RhoGAP regulate cell-cell adhesion by coordinating antagonism between Rac and Rho. Cell 2007; 127:1027-39. [PMID: 17129786 DOI: 10.1016/j.cell.2006.09.046] [Citation(s) in RCA: 322] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2006] [Revised: 07/15/2006] [Accepted: 09/20/2006] [Indexed: 11/30/2022]
Abstract
Integration of receptor tyrosine kinase, integrin, and cadherin activities is crucial for normal cell growth, motility, and adhesion. Here, we describe roles for p120-catenin (p120) and p190RhoGAP that coordinate crosstalk between these systems and regulate cadherin function. Surprisingly, PDGFR-induced actin remodeling in NIH3T3 cells is blocked in the absence of p120, and the cells are partially transformed via constitutive activation of Rho. We have traced the mechanism to unexpected codependent roles for p120 and p190RhoGAP in regulating Rac-dependent antagonism of Rho. Receptor-induced Rac activity causes translocation of p190RhoGAP to adherens junctions (AJs), where it couples to the cadherin complex via interaction with p120. AJ formation is dependent on this p120-p190RhoGAP interaction and fails altogether if either of these proteins are compromised. We propose that Rac activation links diverse signaling systems to AJ assembly by controlling transient p190RhoGAP interactions with p120 and localized inhibition of Rho.
Collapse
Affiliation(s)
- Gregg A Wildenberg
- Department of Cancer Biology, 438 Preston Building, Vanderbilt University, Nashville, TN 37232, USA
| | | | | | | | | | | | | |
Collapse
|
231
|
Madhavan R, Zhao XT, Reynolds AB, Peng HB. Involvement of p120 catenin in myopodial assembly and nerve-muscle synapse formation. ACTA ACUST UNITED AC 2007; 66:1511-27. [PMID: 17031840 DOI: 10.1002/neu.20320] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
At developing neuromuscular junctions (NMJs), muscles initially contact motor axons by microprocesses, or myopodia, which are induced by nerves and nerve-secreted agrin, but it is unclear how myopodia are assembled and how they influence synaptic differentiation at the NMJ. Here, we report that treatment of cultured muscle cells with agrin transiently depleted p120 catenin (p120ctn) from cadherin junctions in situ, and increased the tyrosine phosphorylation and decreased the cadherin-association of p120ctn in cell extracts. Whereas ectopic expression of wild-type p120ctn in muscle generated myopodia in the absence of agrin, expression of a specific dominant-negative mutant form of p120ctn, which blocks filopodial assembly in nonmuscle cells, suppressed nerve- and agrin-induction of myopodia. Significantly, approaching neurites triggered reduced acetylcholine receptor (AChR) clustering along the edges of muscle cells expressing mutant p120ctn than of control cells, although the ability of the mutant cells to cluster AChRs was itself normal. Our results indicate a novel role of p120ctn in agrin-induced myopodial assembly and suggest that myopodia increase muscle-nerve contacts and muscle's access to neural agrin to promote NMJ formation.
Collapse
Affiliation(s)
- Raghavan Madhavan
- Department of Biology, Hong Kong University of Science and Technology, Kowloon, Hong Kong
| | | | | | | |
Collapse
|
232
|
Reynolds AB. p120-catenin: Past and present. BIOCHIMICA ET BIOPHYSICA ACTA 2007; 1773:2-7. [PMID: 17175391 PMCID: PMC2892545 DOI: 10.1016/j.bbamcr.2006.09.019] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/11/2006] [Accepted: 09/12/2006] [Indexed: 11/19/2022]
Abstract
p120-catenin was first described in 1989 as a Src substrate whose phosphorylation correlated with transformation. It was identified by cDNA cloning in 1992, and shown to interact with cadherins in 1994. Though enigmatic for some time, p120 has emerged as a master regulator of cadherin stability, and an important modulator of RhoGTPase activities. With the discovery of p120 family members and evidence for fundamental roles in cell biology and cancer, the field has expanded dramatically in recent years. As an introduction to this collection of reviews on p120 and its relatives, the editors have requested a personal commentary and historical perspective on the discovery of p120. The anecdotal parts have no particular purpose, but are mostly unpublished and perhaps of interest to some.
Collapse
Affiliation(s)
- Albert B Reynolds
- Department of Cancer Biology, Vanderbilt University, 438 Preston Building Nashville, TN 37232-684, USA.
| |
Collapse
|
233
|
Anastasiadis PZ. p120-ctn: A nexus for contextual signaling via Rho GTPases. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:34-46. [PMID: 17028013 DOI: 10.1016/j.bbamcr.2006.08.040] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2006] [Revised: 08/24/2006] [Accepted: 08/27/2006] [Indexed: 01/11/2023]
Abstract
p120 catenin (p120) is the prototypic member of a subfamily of armadillo repeat domain proteins involved in intercellular adhesion. Recent evidence indicates that p120 associates with classical cadherins and regulates their stability. Ectopic p120 expression results in a variety of morphological effects, and promotes cell migration. There is now strong evidence that p120 acts, at least in part, through regulation of Rho GTPases. The data suggest that p120 may act as a signaling nexus, conveying messages from the cellular micro- and macro-environment to the cell's interior. By regulating Rho GTPases in a context-dependent manner p120 can exert profound effects on cellular responses from synaptic plasticity to vesicle trafficking, as well as regulate the motile vs. sessile, and possibly the proliferative vs. quiescent phenotype of epithelial cells. Here, we review the new evidence on the relationship of p120 to Rho GTPases, and discuss potential roles for the p120-Rho connection in normal and malignant cells.
Collapse
Affiliation(s)
- Panos Z Anastasiadis
- Department Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Griffin Cancer Research Building, Rm. 307, 4500 San Pablo Road, Jacksonville, FL 32224, USA.
| |
Collapse
|
234
|
Zhuang G, Hunter S, Hwang Y, Chen J. Regulation of EphA2 Receptor Endocytosis by SHIP2 Lipid Phosphatase via Phosphatidylinositol 3-Kinase-dependent Rac1 Activation. J Biol Chem 2007; 282:2683-94. [PMID: 17135240 DOI: 10.1074/jbc.m608509200] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Endocytosis of Eph receptors is critical for a number of biological processes, including modulating axon growth cone collapse response and regulating cell surface levels of receptor in epithelial cells. In particular, ephrin-A ligand stimulation of tumor cells induces EphA2 receptor internalization and degradation, a process that has been explored as a means to reduce tumor malignancy. However, the mechanism and regulation of ligand-induced Eph receptor internalization are not well understood. Here we show that SHIP2 (Src homology 2 domain-containing phosphoinositide 5-phosphatase 2) is recruited to activated EphA2 via a heterotypic sterile alpha motif (SAM)-SAM domain interaction, leading to regulation of EphA2 internalization. Overexpression of SHIP2 inhibits EphA2 receptor endocytosis, whereas suppression of SHIP2 expression by small interfering RNA-mediated gene silencing promotes ligand-induced EphA2 internalization and degradation. SHIP2 regulates EphA2 endocytosis via phosphatidylinositol 3-kinase-dependent Rac1 activation. Phosphatidylinositol 3,4,5-trisphosphate levels are significantly elevated in SHIP2 knockdown cells, phosphatidylinositol 3-kinase inhibitor decreases phosphatidylinositol 3,4,5-trisphosphate levels and suppresses increased EphA2 endocytosis. Ephrin-A1 stimulation activates Rac1 GTPase, and the Rac1-GTP levels are further increased in SHIP2 knockdown cells. A dominant negative Rac1 GTPase effectively inhibited ephrin-A1-induced EphA2 endocytosis. Together, our findings provide evidence that recruitment of SHIP2 to EphA2 attenuates a positive signal to receptor endocytosis mediated by phosphatidylinositol 3-kinase and Rac1 GTPase.
Collapse
Affiliation(s)
- Guanglei Zhuang
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | | | | | |
Collapse
|
235
|
Xiao K, Oas RG, Chiasson CM, Kowalczyk AP. Role of p120-catenin in cadherin trafficking. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:8-16. [PMID: 16949165 DOI: 10.1016/j.bbamcr.2006.07.005] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2006] [Revised: 07/13/2006] [Accepted: 07/14/2006] [Indexed: 10/24/2022]
Abstract
p120-catenin (p120) has emerged over the past several years as an important regulatory component of the cadherin adhesive complex. A core function of p120 in mammalian cells is to stabilize cadherins at the cell membrane by modulating cadherin membrane trafficking and degradation. In this way, p120 levels act as a set point mechanism that tunes cell-cell adhesive interactions. The primary control point for this regulatory activity appears to be at the level of cadherin internalization from the plasma membrane, although p120 may also impact other aspects of cadherin trafficking and turnover. In the following review, the general mechanisms of cadherin trafficking are discussed, and models for how p120 may influence cadherin membrane dynamics are presented. In one model, p120 may function as a "cap" to bind the cadherin cytoplasmic tail and prevent cadherin interactions with endocytic membrane trafficking machinery. Alternatively, p120 may stabilize cell junctions or regulate membrane trafficking machinery through interactions with small GTPases such as Rho A, Rac and Cdc42. Through these mechanisms p120 exerts influence over a wide range of biological processes that are dependent upon tight regulation of cell surface cadherin levels.
Collapse
Affiliation(s)
- Kanyan Xiao
- Department of Cell Biology and Dermatology, Emory University School of Medicine, Whitehead Biomedical Research Building, 615 Michael Street, Atlanta, GA 30322, USA.
| | | | | | | |
Collapse
|
236
|
Castaño J, Solanas G, Casagolda D, Raurell I, Villagrasa P, Bustelo XR, García de Herreros A, Duñach M. Specific phosphorylation of p120-catenin regulatory domain differently modulates its binding to RhoA. Mol Cell Biol 2006; 27:1745-57. [PMID: 17194753 PMCID: PMC1820477 DOI: 10.1128/mcb.01974-06] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
p120-catenin is an adherens junction-associated protein that controls E-cadherin function and stability. p120-catenin also binds intracellular proteins, such as the small GTPase RhoA. In this paper, we identify the p120-catenin N-terminal regulatory domain as the docking site for RhoA. Moreover, we demonstrate that the binding of RhoA to p120-catenin is tightly controlled by the Src family-dependent phosphorylation of p120-catenin on tyrosine residues. The phosphorylation induced by Src and Fyn tyrosine kinases on p120-catenin induces opposite effects on RhoA binding. Fyn, by phosphorylating a residue located in the regulatory domain of p120-catenin (Tyr112), inhibits the interaction of this protein with RhoA. By contrast, the phosphorylation of Tyr217 and Tyr228 by Src promotes a better affinity of p120-catenin towards RhoA. In agreement with these biochemical data, results obtained in cell lines support the important role of these phosphorylation sites in the regulation of RhoA activity by p120-catenin. Taken together, these observations uncover a new regulatory mechanism acting on p120-catenin that contributes to the fine-tuned regulation of the RhoA pathways during specific signaling events.
Collapse
Affiliation(s)
- Julio Castaño
- Unitat de Biofísica, Departament Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
237
|
Ohno H, Uemura K, Shintani-Ishida K, Nakamura M, Inomata M, Yoshida KI. Ischemia promotes calpain-mediated degradation of p120-catenin in SH-SY5Y cells. Biochem Biophys Res Commun 2006; 353:547-52. [PMID: 17196166 DOI: 10.1016/j.bbrc.2006.12.061] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2006] [Accepted: 12/05/2006] [Indexed: 12/26/2022]
Abstract
p120-catenin contributes to the cadherin-mediated adhesion and aggregation of cells. mu-Calpain was activated and p120-catenin was degraded after 36 h of ischemia in differentiated SH-SY5Y cells. Calpain inhibitors Cbz-Val-Phe-H (MDL28170, 20 microM) and N-acetyl-leucyl-leucyl-norleucinal (ALLN, 20 microM) increased the levels of dephosphorylated p120-catenin, aggregation, and cell survival as detected by reduced LDH release in ischemic cells. However, a proteasome inhibitor lactacystin had no such effects. This is the first report of the calpain-mediated degradation of p120-catenin and an association between the level of dephosphorylated p120-catenin and cell aggregation in ischemic neuronal cells.
Collapse
Affiliation(s)
- Hiroshi Ohno
- Department of Forensic Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | | | | | | | | | | |
Collapse
|
238
|
Park JI, Ji H, Jun S, Gu D, Hikasa H, Li L, Sokol SY, McCrea PD. Frodo Links Dishevelled to the p120-Catenin/Kaiso Pathway: Distinct Catenin Subfamilies Promote Wnt Signals. Dev Cell 2006; 11:683-95. [PMID: 17084360 DOI: 10.1016/j.devcel.2006.09.022] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2006] [Revised: 08/14/2006] [Accepted: 09/26/2006] [Indexed: 12/13/2022]
Abstract
p120-catenin is an Arm repeat protein that interacts with varied components such as cadherin, small G proteins, kinases, and the Kaiso transcriptional repressor. Despite recent advances in understanding the roles that p120-catenin and Kaiso play in downstream modulation of Wnt/beta-catenin signaling, the identity of the upstream regulators of the p120-catenin/Kaiso pathway have remained unclear. Here, we find that p120-catenin binds Frodo, which itself interacts with the Wnt pathway protein Dishevelled (Dsh). In Xenopus laevis, we demonstrate that Wnt signals result in Frodo-mediated stabilization of p120-catenin, which, in turn, promotes Kaiso sequestration or removal from the nucleus. Our results point to Dsh and Frodo as upstream regulators of the p120-catenin/Kaiso signaling pathway. Importantly, this suggests that Wnt signals acting through Dsh regulate the stability of p120-catenin in addition to that of beta-catenin, and that each catenin promotes its respective signal in parallel to regulate distinct, as well as shared, direct downstream gene targets.
Collapse
Affiliation(s)
- Jae-il Park
- Department of Biochemistry and Molecular Biology and Program in Genes and Development, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
239
|
Abstract
Adherens junctions have been traditionally viewed as building blocks of tissue architecture. The foundations for this view began to change with the discovery that a central component of AJs, beta-catenin, can also function as a transcriptional cofactor in Wnt signaling. In recent years, conventional views have similarly been shaken about the other two major AJ catenins, alpha-catenin and p120-catenin. Catenins have emerged as molecular sensors that integrate cell-cell junctions and cytoskeletal dynamics with signaling pathways that govern morphogenesis, tissue homeostasis, and even intercellular communication between different cell types within a tissue. These findings reveal novel aspects of AJ function in normal tissues and offer insights into how changes in AJs and their associated proteins and cytoskeletal dynamics impact wound-repair and cancer.
Collapse
Affiliation(s)
- Mirna Perez-Moreno
- Howard Hughes Medical Institute, Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, New York 10021, USA
| | | |
Collapse
|
240
|
Kim SC, Sprung R, Chen Y, Xu Y, Ball H, Pei J, Cheng T, Kho Y, Xiao H, Xiao L, Grishin NV, White M, Yang XJ, Zhao Y. Substrate and functional diversity of lysine acetylation revealed by a proteomics survey. Mol Cell 2006; 23:607-18. [PMID: 16916647 DOI: 10.1016/j.molcel.2006.06.026] [Citation(s) in RCA: 1214] [Impact Index Per Article: 63.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2006] [Revised: 05/03/2006] [Accepted: 06/27/2006] [Indexed: 12/16/2022]
Abstract
Acetylation of proteins on lysine residues is a dynamic posttranslational modification that is known to play a key role in regulating transcription and other DNA-dependent nuclear processes. However, the extent of this modification in diverse cellular proteins remains largely unknown, presenting a major bottleneck for lysine-acetylation biology. Here we report the first proteomic survey of this modification, identifying 388 acetylation sites in 195 proteins among proteins derived from HeLa cells and mouse liver mitochondria. In addition to regulators of chromatin-based cellular processes, nonnuclear localized proteins with diverse functions were identified. Most strikingly, acetyllysine was found in more than 20% of mitochondrial proteins, including many longevity regulators and metabolism enzymes. Our study reveals previously unappreciated roles for lysine acetylation in the regulation of diverse cellular pathways outside of the nucleus. The combined data sets offer a rich source for further characterization of the contribution of this modification to cellular physiology and human diseases.
Collapse
Affiliation(s)
- Sung Chan Kim
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
241
|
Yanagisawa M, Anastasiadis PZ. p120 catenin is essential for mesenchymal cadherin-mediated regulation of cell motility and invasiveness. ACTA ACUST UNITED AC 2006; 174:1087-96. [PMID: 16982802 PMCID: PMC2064398 DOI: 10.1083/jcb.200605022] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
During epithelial tumor progression, the loss of E-cadherin expression and inappropriate expression of mesenchymal cadherins coincide with increased invasiveness. Reexpression experiments have established E-cadherin as an invasion suppressor. However, the mechanism by which E-cadherin suppresses invasiveness and the role of mesenchymal cadherins are poorly understood. We show that both p120 catenin and mesenchymal cadherins are required for the invasiveness of E-cadherin-deficient cells. p120 binding promotes the up-regulation of mesenchymal cadherins and the activation of Rac1, which are essential for cell migration and invasiveness. p120 also promotes invasiveness by inhibiting RhoA activity, independently of cadherin association. Furthermore, association of endogenous p120 with E-cadherin is required for E-cadherin-mediated suppression of invasiveness and is accompanied by a reduction in mesenchymal cadherin levels. The data indicate that p120 acts as a rheostat, promoting a sessile cellular phenotype when associated with E-cadherin or a motile phenotype when associated with mesenchymal cadherins.
Collapse
|
242
|
van Hengel J, van Roy F. Diverse functions of p120ctn in tumors. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2006; 1773:78-88. [PMID: 17030444 DOI: 10.1016/j.bbamcr.2006.08.033] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2006] [Revised: 08/22/2006] [Accepted: 08/23/2006] [Indexed: 01/11/2023]
Abstract
p120ctn is a member of the Armadillo protein family. It stabilizes the cadherin-catenin adhesion complex at the plasma membrane, but also has additional roles in the cytoplasm and nucleus. Extensive alternative mRNA splicing and multiple phosphorylation sites generate additional complexity. Evidence is emerging that complete loss, downregulation or mislocalization of p120ctn correlates with progression of different types of human tumors. It remains to be determined whether a causal relationship exists between specific isoform expression, subcellular localization or selective phosphorylation of p120ctn on the one hand and tumor prognosis on the other.
Collapse
Affiliation(s)
- Jolanda van Hengel
- Molecular Cell Biology Unit, Department for Molecular Biomedical Research, VIB-Ghent University, Technologiepark 927, B-9052 Gent (Zwijnaarde), Belgium
| | | |
Collapse
|
243
|
Hou JC, Shigematsu S, Crawford HC, Anastasiadis PZ, Pessin JE. Dual Regulation of Rho and Rac by p120 Catenin Controls Adipocyte Plasma Membrane Trafficking. J Biol Chem 2006; 281:23307-12. [PMID: 16754687 DOI: 10.1074/jbc.m603127200] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
During 3T3L1 adipogenesis there is a marked reduction in beta-catenin and N-cadherin expression with a relatively small decrease in p120 catenin protein levels. Cell fractionation demonstrated a predominant decrease in the particulate (membrane-bound) pool of p120 catenin with little effect on the soluble pool, resulting in a large redistribution from the plasma membrane to the cytosol. Reexpression of p120 catenin inhibited constitutive (transferrin receptor) and regulated mannose 6-phosphate receptor and GLUT4 trafficking to the plasma membrane. The inhibition of membrane trafficking was specific for p120 catenin function as this could be rescued by co-expression of N-cadherin. Moreover, overexpression of a p120 catenin deletion mutant (p120delta622-628) or splice variant (p120-4A), neither of which could regulate Rho or Rac activity, showed no significant effect. The inhibition of GLUT4 translocation was also observed upon the simultaneous expression of a constitutively active Rac mutant (Rac1/Val12) in combination with a dominant-interfering Rho mutant (RhoA/Asn19). This was recapitulated by expression of the Rho ADP-ribosylation factor (C3ADP) in combination with constitutively active Rac1/Val12. Moreover, siRNA-mediated knockdown of p120 catenin resulted in increased basal state accumulation of GLUT4 at the plasma membrane. Together, these data demonstrate that p120 catenin plays an important role in maintaining the basal tone of membrane protein trafficking in adipocytes through the dual regulation of Rho and Rac function and accounts for reports implicating Rho or Rac in the control of GLUT4 translocation.
Collapse
Affiliation(s)
- June C Hou
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York 11794-8651, USA
| | | | | | | | | |
Collapse
|
244
|
Xia X, Carnahan RH, Vaughan MH, Wildenberg GA, Reynolds AB. p120 serine and threonine phosphorylation is controlled by multiple ligand-receptor pathways but not cadherin ligation. Exp Cell Res 2006; 312:3336-48. [PMID: 16935280 DOI: 10.1016/j.yexcr.2006.07.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2006] [Accepted: 07/06/2006] [Indexed: 12/25/2022]
Abstract
p120-catenin (p120) regulates cadherin turnover and is required for cadherin stability. Extensive and dynamic phosphorylation on tyrosine, serine and threonine residues in the N-terminal regulatory domain has been postulated to regulate p120 function, possibly through modulation of the efficiency of p120/cadherin interaction. Here we have utilized novel phospho-specific monoclonal antibodies to four major p120 serine and threonine phosphorylation sites to monitor individual phosphorylation events and their consequences. Surprisingly, membrane-localization and not cadherin interaction is the main determinant in p120 serine and threonine phosphorylation and dephosphorylation. Furthermore, the phospho-status of these four residues had no obvious effect on p120's role in cadherin complex stabilization or cell-cell adhesion. Interestingly, dephosphorylation was dramatically induced by PKC activation, but PKC-independent pathways were also evident. The data suggest that p120 dephosphorylation at these sites is modulated by multiple cell surface receptors primarily through PKC-dependent pathways, but these changes do not seem to reduce p120/cadherin affinity.
Collapse
Affiliation(s)
- Xiaobo Xia
- Department of Cancer Biology, Vanderbilt University Medical Center, 771 Preston Research Building, Nashville, TN 37232, USA
| | | | | | | | | |
Collapse
|
245
|
Abstract
E-cadherin has been linked to the suppression of tumor growth and the inhibition of cell proliferation in culture. We observed that progressively decreasing the seeding density of normal rat kidney-52E (NRK-52E) or MCF-10A epithelial cells from confluence, indeed, released cells from growth arrest. Unexpectedly, a further decrease in seeding density so that cells were isolated from neighboring cells decreased proliferation. Experiments using microengineered substrates showed that E-cadherin engagement stimulated the peak in proliferation at intermediate seeding densities, and that the proliferation arrest at high densities did not involve E-cadherin, but rather resulted from a crowding-dependent decrease in cell spreading against the underlying substrate. Rac1 activity, which was induced by E-cadherin engagement specifically at intermediate seeding densities, was required for the cadherin-stimulated proliferation, and the control of Rac1 activation by E-cadherin was mediated by p120-catenin. Together, these findings demonstrate a stimulatory role for E-cadherin in proliferative regulation, and identify a simple mechanism by which cell–cell contact may trigger or inhibit epithelial cell proliferation in different settings.
Collapse
Affiliation(s)
- Wendy F Liu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
246
|
Elia LP, Yamamoto M, Zang K, Reichardt LF. p120 catenin regulates dendritic spine and synapse development through Rho-family GTPases and cadherins. Neuron 2006; 51:43-56. [PMID: 16815331 PMCID: PMC2587166 DOI: 10.1016/j.neuron.2006.05.018] [Citation(s) in RCA: 165] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2005] [Revised: 02/17/2006] [Accepted: 05/23/2006] [Indexed: 11/20/2022]
Abstract
Both the cadherin-catenin complex and Rho-family GTPases have been shown to regulate dendrite development. We show here a role for p120 catenin (p120ctn) in regulating spine and synapse formation in the developing mouse brain. p120catenin gene deletion in hippocampal pyramidal neurons in vivo resulted in reduced spine and synapse densities along dendrites. In addition, p120 catenin loss resulted in reduced cadherin levels and misregulation of Rho-family GTPases, with decreased Rac1 and increased RhoA activity. Analyses in vitro indicate that the reduced spine density reflects aberrant Rho-family GTPase signaling, whereas the effects on spine maturation appear to result from reduced cadherin levels and possibly aberrant Rho-family GTPase signaling. Thus, p120ctn acts as a signal coordinator between cadherins and Rho-family GTPases to regulate cytoskeletal changes required during spine and synapse development.
Collapse
Affiliation(s)
- Lisa P Elia
- Howard Hughes Medical Institute and Department of Physiology, 1550 Fourth Street, University of California, San Francisco, San Francisco, California 94143, USA
| | | | | | | |
Collapse
|
247
|
McCrea PD, Park JI. Developmental functions of the P120-catenin sub-family. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2006; 1773:17-33. [PMID: 16942809 DOI: 10.1016/j.bbamcr.2006.06.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2006] [Revised: 06/21/2006] [Accepted: 06/26/2006] [Indexed: 01/11/2023]
Abstract
For more than a decade, cell, developmental and cancer investigators have brought about a wide interest in the biology of catenin proteins, an attraction being their varied functions within differing cellular compartments. While the diversity of catenin localizations and roles has been intriguing, it has also posed a challenge to the clear interpretation of loss- or gain-of-function developmental phenotypes. The most deeply studied member of the larger catenin family is beta-catenin, whose contributions span areas including cell adhesion and intracellular signaling/ transcriptional control. More recently, attention has been directed towards p120-catenin, which in conjunction with the p120-catenin sub-family members ARVCF- and delta-catenins, are the subjects of this review. Although the requirement for vertebrate versus invertebrate p120-catenin are at variance, vertebrate p120-catenin sub-family members may each inter-link cadherin, cytoskeletal and gene regulatory functions in embryogenesis and disease.
Collapse
Affiliation(s)
- Pierre D McCrea
- Department of Biochemistry and Molecular Biology, Program in Genes and Development, University of Texas MD Anderson Cancer Center, University of Texas Graduate School of Biomedical Science, Houston TX 77030, USA.
| | | |
Collapse
|
248
|
Deramaudt TB, Takaoka M, Upadhyay R, Bowser MJ, Porter J, Lee A, Rhoades B, Johnstone CN, Weissleder R, Hingorani SR, Mahmood U, Rustgi AK. N-cadherin and keratinocyte growth factor receptor mediate the functional interplay between Ki-RASG12V and p53V143A in promoting pancreatic cell migration, invasion, and tissue architecture disruption. Mol Cell Biol 2006; 26:4185-200. [PMID: 16705170 PMCID: PMC1489079 DOI: 10.1128/mcb.01055-05] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2005] [Revised: 07/10/2005] [Accepted: 03/17/2006] [Indexed: 01/24/2023] Open
Abstract
The genetic basis of pancreatic ductal adenocarcinoma, which constitutes the most common type of pancreatic malignancy, involves the sequential activation of oncogenes and inactivation of tumor suppressor genes. Among the pivotal genetic alterations are Ki-RAS oncogene activation and p53 tumor suppressor gene inactivation. We explain that the combination of these genetic events facilitates pancreatic carcinogenesis as revealed in novel three-dimensional cell (spheroid cyst) culture and in vivo subcutaneous and orthotopic xenotransplantation models. N-cadherin, a member of the classic cadherins important in the regulation of cell-cell adhesion, is induced in the presence of Ki-RAS mutation but subsequently downregulated with the acquisition of p53 mutation as revealed by gene microarrays and corroborated by reverse transcription-PCR and Western blotting. N-cadherin modulates the capacity of pancreatic ductal cells to migrate and invade, in part via complex formation with keratinocyte growth factor receptor and neural cell adhesion molecule and in part via interaction with p120-catenin. However, modulation of these complexes by Ki-RAS and p53 leads to enhanced cell migration and invasion. This preferentially induces the downstream effector AKT over mitogen-activated protein kinase to execute changes in cellular behavior. Thus, we are able to define molecules that in part are directly affected by Ki-RAS and p53 during pancreatic ductal carcinogenesis, and this provides a platform for potential new molecularly based therapeutic interventions.
Collapse
Affiliation(s)
- Therese B Deramaudt
- Gastroenterology Division, University of Pennsylvania, 415 Curie Boulevard, Philadelphia, PA 19104-2144, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
249
|
Guillemot L, Citi S. Cingulin regulates claudin-2 expression and cell proliferation through the small GTPase RhoA. Mol Biol Cell 2006; 17:3569-77. [PMID: 16723500 PMCID: PMC1525245 DOI: 10.1091/mbc.e06-02-0122] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
In mouse embryoid bodies, mutation of the tight junction protein cingulin results in changes in gene expression. Here, we studied the function of cingulin using a gene silencing approach in Madin-Darby canine kidney (MDCK) cells. Cingulin-depleted cells show higher protein and mRNA levels of claudin-2 and ZO-3, increased RhoA activity, activation of G1/S phase transition, and increased cell density. The effects of cingulin depletion on claudin-2 expression, cell proliferation, and density are reversed by coexpression of either a dominant-negative form of RhoA (RhoAN19) or the Rho-inhibiting enzyme C3 transferase. However, the increase in ZO-3 protein and mRNA levels is not reversed by inhibition of either RhoA, p38, extracellular signal-regulated kinase (ERK), or c-Jun NH2-terminal kinase (JNK), suggesting that cingulin modulates ZO-3 expression by a different mechanism. JNK is implicated in the regulation of claudin-2 levels independently of cingulin depletion and RhoA activity, indicating distinct roles of RhoA- and JNK-dependent pathways in the control of claudin-2 expression. Finally, cingulin depletion does not significantly alter the barrier function of monolayers and the overall molecular organization of tight junctions. These results provide novel insights about the mechanisms of cingulin function and the signaling pathways controlling claudin-2 expression in MDCK cells.
Collapse
Affiliation(s)
- Laurent Guillemot
- *Department of Molecular Biology, University of Geneva, CH-1211 Geneva, Switzerland; and
| | - Sandra Citi
- *Department of Molecular Biology, University of Geneva, CH-1211 Geneva, Switzerland; and
| |
Collapse
|
250
|
Abstract
Cadherins are expressed on the cell surface as a dimer in the membrane of one cell (cis dimer) that interacts with a cis dimer on an adjacent cell to form an adhesive trans dimer. It is well established that both cis and trans dimers must form for the cadherin to be an effective adhesion protein. In addition to their adhesive activity cadherins also play an important role in modulating cell behavior by regulating cell motility and signal transduction. Whether or not cis or trans dimers are necessary for the nonadhesive functions of cadherins has not been addressed. Here we show that N-cadherin cis dimers are necessary to induce cell motility in epithelial cells and that N-cadherin's ability to modulate the steady state levels of activated small GTPases requires both cis and trans dimers.
Collapse
Affiliation(s)
- Young J Kim
- Department of Oral Biology, College of Dentistry, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | | | |
Collapse
|