151
|
Rehm K, Panzer L, van Vliet V, Genot E, Linder S. Drebrin preserves endothelial integrity by stabilizing nectin at adherens junctions. J Cell Sci 2013; 126:3756-69. [DOI: 10.1242/jcs.129437] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Regulation of cell-cell contacts is essential for integrity of the vascular endothelium. Here, a critical role of the F-actin binding protein drebrin in maintaining endothelial integrity is revealed under conditions mimicking vascular flow. Drebrin knockdown leads to weakening of cell-cell contacts, characterized by loss of nectin from adherens junctions and its subsequent lysosomal degradation. Immunoprecipitation, FRAP and mitochondrial retargeting experiments show that nectin stabilization occurs through a chain of interactions: drebrin binding to F-actin, interaction of drebrin and afadin through their polyproline and PR1-2 regions, and recruitment of nectin through afadin's PDZ region. Key elements are drebrin's modules that confer binding to afadin and F-actin. Evidence is provided by constructs containing afadin's PDZ region coupled to drebrin's F-actin binding region or to lifeact, which restore junctional nectin under knockdown of drebrin or of both drebrin and afadin. Drebrin, containing binding sites for both afadin and F-actin, is thus uniquely equipped to stabilize nectin at endothelial junctions and to preserve endothelial integrity under vascular flow.
Collapse
|
152
|
Pérez-Gómez R, Slováková J, Rives-Quinto N, Krejci A, Carmena A. A Serrate-Notch-Canoe complex mediates glial-neuroepithelial cell interactions essential during Drosophila optic lobe development. J Cell Sci 2013; 126:4873-84. [DOI: 10.1242/jcs.125617] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
It is firmly established that neuron-glia interactions are fundamental across species for the correct establishment of a functional brain. Here, we found that the glia of the Drosophila larval brain display an essential non-autonomous role during the development of the optic lobe. The optic lobe develops from neuroepithelial cells that proliferate by dividing symmetrically until they switch to asymmetric/differentiative divisions generating neuroblasts. The proneural gene lethal of scute (l'sc) is transiently activated by the Epidermal Growth Factor Receptor (EGFR)/Ras signal transduction pathway at the leading edge of a proneural wave that sweeps from medial to lateral neuroepithelium promoting this switch. This process is tightly regulated by the tissue-autonomous function within the neuroepithelium of multiple signaling pathways, including EGFR/Ras and Notch. This study shows that the Notch ligand Serrate (Ser) is expressed in the glia and it forms a complex in vivo with Notch and Canoe, which colocalize at the adherens junctions of neuroepithelial cells. This complex is crucial for glial-neuroepithelial cell interactions during optic lobe development. Ser is tissue-autonomously required in the glia where it activates Notch to regulate its proliferation, and non-autonomously in the neuroepithelium where Ser induces Notch signaling to avoid the premature activation of the EGFR/Ras pathway and hence of L'sc. Interestingly, different Notch activity reporters showed very different expression patterns in the glia and in the neuroepithelium, suggesting the existence of tissue-specific factors that promote the expression of particular Notch target genes or/and a reporter response dependent on different thresholds of Notch signaling.
Collapse
|
153
|
Mandai K, Rikitake Y, Shimono Y, Takai Y. Afadin/AF-6 and Canoe. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 116:433-54. [DOI: 10.1016/b978-0-12-394311-8.00019-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
154
|
Monteiro AC, Parkos CA. Intracellular mediators of JAM-A-dependent epithelial barrier function. Ann N Y Acad Sci 2012; 1257:115-24. [PMID: 22671597 DOI: 10.1111/j.1749-6632.2012.06521.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Junctional adhesion molecule-A (JAM-A) is a critical signaling component of the apical junctional complex, a structure composed of several transmembrane and scaffold molecules that controls the passage of nutrients and solutes across epithelial surfaces. Observations from JAM-A-deficient epithelial cells and JAM-A knockout animals indicate that JAM-A is an important regulator of epithelial paracellular permeability; however, the mechanism(s) linking JAM-A to barrier function are not understood. This review highlights recent findings relevant to JAM-A-mediated regulation of epithelial permeability, focusing on the role of upstream and downstream signaling candidates. We draw on what is known about proteins reported to associate with JAM-A in other pathways and on known modulators of barrier function to propose candidate effectors that may mediate JAM-A regulation of epithelial paracellular permeability. Further investigation of pathways highlighted in this review may provide ideas for novel therapeutics that target debilitating conditions associated with barrier dysfunction, such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Ana C Monteiro
- Department of Pathology and Laboratory Medicine, Epithelial Pathobiology Research Unit, Emory University, Atlanta, Georgia 30322, USA
| | | |
Collapse
|
155
|
Citi S, Pulimeno P, Paschoud S. Cingulin, paracingulin, and PLEKHA7: signaling and cytoskeletal adaptors at the apical junctional complex. Ann N Y Acad Sci 2012; 1257:125-32. [PMID: 22671598 DOI: 10.1111/j.1749-6632.2012.06506.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Cingulin, paracingulin, and PLEKHA7 are proteins localized in the cytoplasmic region of the apical junctional complex of vertebrate epithelial cells. Cingulin has been detected at tight junctions (TJs), whereas paracingulin has been detected at both TJs and adherens junctions (AJs) and PLEKHA7 has been detected at AJs. One function of cingulin and paracingulin is to regulate the activity of Rho family GTPases at junctions through their direct interaction with guanidine exchange factors of RhoA and Rac1. Cingulin also contributes to the regulation of transcription of several genes in different types of cultured cells, in part through its ability to modulate RhoA activity. PLEKHA7, together with paracingulin, is part of a protein complex that links E-cadherin to the microtubule cytoskeleton at AJs. In this paper, we review the current knowledge about these proteins, including their discovery, the characterization of their expression, localization, structure, molecular interactions, and their roles in different developmental and disease model systems.
Collapse
Affiliation(s)
- Sandra Citi
- Department of Molecular Biology, University of Geneva, Switzerland.
| | | | | |
Collapse
|
156
|
Salaheldeen E, Kurio H, Howida A, Iida H. Molecular cloning and localization of a CEACAM2 isoform, CEACAM2-L, expressed in spermatids in mouse testis. Mol Reprod Dev 2012; 79:843-52. [PMID: 23070997 DOI: 10.1002/mrd.22123] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Accepted: 10/06/2012] [Indexed: 11/09/2022]
Abstract
Carcinoembryonic antigen (CEA) family, a subgroup of the immunoglobulin (Ig) superfamily, is divided into two sub-families: the CEA-related cell adhesion molecules (CEACAM) and the pregnancy-specific glycoproteins. The isoform CEACAM2 is expressed in mouse testis; in this study, we identified a novel isoform of Ceacam2, Ceacam2-Long (Ceacam2-L). CEACAM2-L is different from CEACAM2 in that it has much longer cytoplasmic tail region. Ceacam2-L starts to appear faintly in mouse testis after 3 weeks of postnatal development, and its expression level increased after 5 weeks. Immunoblot analysis confirmed the expression of CEACAM2-L in the seminiferous epithelium of mouse testis. Immunohistochemical data showed that CEACAM2-L was not observed on spermatogonia, spermatocytes, round spermatids, or Sertoli cells, but was seen at the plasma membrane of elongating spermatids in contact with extended cytoplasmic processes of Sertoli cells. CEACAM2-L was not detected at the head region of elongating spermatids, where the apical ectoplasmic specialization is constructed. These data suggest that CEACAM2-L might be a novel adhesion molecule contributing to cell-to-cell adhesion between elongating spermatids and Sertoli cells within the seminiferous epithelium.
Collapse
Affiliation(s)
- Elsaid Salaheldeen
- Laboratory of Zoology, Graduate School of Agriculture, Kyushu University, Higashiku Hakozaki, Fukuoka, Japan
| | | | | | | |
Collapse
|
157
|
Irie K, Shimizu K, Sakisaka T, Ikeda W, Takai Y. Roles of nectins in cell adhesion, signaling and polarization. Handb Exp Pharmacol 2012:343-72. [PMID: 20455098 DOI: 10.1007/978-3-540-68170-0_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Nectins are Ca(2+)-independent immunoglobulin-like cell-cell adhesion molecules which constitute a family of four members. Nectins homophilically and heterophilically trans-interact and cause cell-cell adhesion. This nectin-based cell-cell adhesion plays roles in the organization of adherens junctions in epithelial cells and fibroblasts and synaptic junctions in neurons in cooperation with cadherins. The nectin-based cell-cell adhesion plays roles in the contacts between commissural axons and floor plate cells and in the organization of Sertoli cell-spermatid junctions in the testis, independently of cadherins. Nectins furthermore regulate intracellular signaling through Cdc42 and Rac small G proteins and cell polarization through cell polarity proteins. Pathologically, nectins serve as entry and cell-cell spread mediators of herpes simplex viruses.
Collapse
Affiliation(s)
- K Irie
- Department of Molecular Biology and Biochemistry, Osaka University Graduate School of Medicine/Faculty of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | | | | | | | | |
Collapse
|
158
|
Abstract
The cadherin-containing intercellular junctions, adherens junctions and desmosomes share an overall logical organization in which the extracellular regions of the cadherins on opposing cells interact, while their cytoplasmic domains are linked to the cytoskeleton through protein assemblies. In adherens junctions, beta-catenin binds to the cytoplasmic domain of cadherins and to alpha-catenin, which links the cadherin/beta-catenin complex to the actin cytoskeleton. In desmosomes, the beta-catenin homolog plakoglobin binds to desmosomal cadherins. The desmosomal cadherin/plakoglobin complex is linked to the intermediate filament system by the protein desmoplakin. In the past decade, components of these systems have been purified to homogeneity and studied biochemically and structurally, providing the beginnings of a mechanistic description of junction architecture and dynamics.
Collapse
Affiliation(s)
- H-J Choi
- Department of Structural Biology, Stanford University School of Medicine, 299 Campus Drive, West Stanford, CA, 94305-5126, USA
| | | |
Collapse
|
159
|
Slováková J, Speicher S, Sánchez-Soriano N, Prokop A, Carmena A. The actin-binding protein Canoe/AF-6 forms a complex with Robo and is required for Slit-Robo signaling during axon pathfinding at the CNS midline. J Neurosci 2012; 32:10035-44. [PMID: 22815517 PMCID: PMC6621277 DOI: 10.1523/jneurosci.6342-11.2012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 05/07/2012] [Accepted: 05/29/2012] [Indexed: 11/21/2022] Open
Abstract
Axon guidance is a key process during nervous system development and regeneration. One of the best established paradigms to study the mechanisms underlying this process is the axon decision of whether or not to cross the midline in the Drosophila CNS. An essential regulator of that decision is the well conserved Slit-Robo signaling pathway. Slit guidance cues act through Robo receptors to repel axons from the midline. Despite good progress in our knowledge about these proteins, the intracellular mechanisms associated with Robo function remain poorly defined. In this work, we found that the scaffolding protein Canoe (Cno), the Drosophila orthologue of AF-6/Afadin, is essential for Slit-Robo signaling. Cno is expressed along longitudinal axonal pioneer tracts, and longitudinal Robo/Fasciclin2-positive axons aberrantly cross the midline in cno mutant embryos. cno mutant primary neurons show a significant reduction of Robo localized in growth cone filopodia and Cno forms a complex with Robo in vivo. Moreover, the commissureless (comm) phenotype (i.e., lack of commissures due to constitutive surface presentation of Robo in all neurons) is suppressed in comm, cno double-mutant embryos. Specific genetic interactions between cno, slit, robo, and genes encoding other components of the Robo pathway, such as Neurexin-IV, Syndecan, and Rac GTPases, further confirm that Cno functionally interacts with the Slit-Robo pathway. Our data argue that Cno is a novel regulator of the Slit-Robo signaling pathway, crucial for regulating the subcellular localization of Robo and for transducing its signaling to the actin cytoskeleton during axon guidance at the midline.
Collapse
Affiliation(s)
- Jana Slováková
- Instituto de Neurociencias, CSIC/UMH, 03550 Sant Joan d'Alacant, Spain, and
| | - Stephan Speicher
- Instituto de Neurociencias, CSIC/UMH, 03550 Sant Joan d'Alacant, Spain, and
| | - Natalia Sánchez-Soriano
- Faculty of Life Sciences, Wellcome Trust Centre for Cell-Matrix Research, Manchester M13 9PT, United Kingdom
| | - Andreas Prokop
- Faculty of Life Sciences, Wellcome Trust Centre for Cell-Matrix Research, Manchester M13 9PT, United Kingdom
| | - Ana Carmena
- Instituto de Neurociencias, CSIC/UMH, 03550 Sant Joan d'Alacant, Spain, and
| |
Collapse
|
160
|
O'Keefe DD, Gonzalez-Niño E, Edgar BA, Curtiss J. Discontinuities in Rap1 activity determine epithelial cell morphology within the developing wing of Drosophila. Dev Biol 2012; 369:223-34. [PMID: 22776378 DOI: 10.1016/j.ydbio.2012.06.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Revised: 05/31/2012] [Accepted: 06/28/2012] [Indexed: 11/28/2022]
Abstract
Mechanisms that govern cell-fate specification within developing epithelia have been intensely investigated, with many of the critical intercellular signaling pathways identified, and well characterized. Much less is known, however, about downstream events that drive the morphological differentiation of these cells, once their fate has been determined. In the Drosophila wing-blade epithelium, two cell types predominate: vein and intervein. After cell proliferation is complete and adhesive cell-cell contacts have been refined, the vast majority of intervein cells adopt a hexagonal morphology. Within vein territories, however, cell-shape refinement results in trapezoids. Signaling events that differentiate between vein and intervein cell fates are well understood, but the genetic pathways underlying vein/intervein cyto-architectural differences remain largely undescribed. We show here that the Rap1 GTPase plays a critical role in determining cell-type-specific morphologies within the developing wing epithelium. Rap1, together with its effector Canoe, promotes symmetric distribution of the adhesion molecule DE-cadherin about the apicolateral circumference of epithelial cells. We provide evidence that in presumptive vein tissue Rap1/Canoe activity is down-regulated, resulting in adhesive asymmetries and non-hexagonal cell morphologies. In particular Canoe levels are reduced in vein cells as they morphologically differentiate. We also demonstrate that over-expression of Rap1 disrupts vein formation both in the developing epithelium and the adult wing blade. Therefore, vein/intervein morphological differences result, at least in part, from the patterned regulation of Rap1 activity.
Collapse
Affiliation(s)
- David D O'Keefe
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | | |
Collapse
|
161
|
Iwasawa N, Negishi M, Oinuma I. R-Ras controls axon branching through afadin in cortical neurons. Mol Biol Cell 2012; 23:2793-804. [PMID: 22593211 PMCID: PMC3395666 DOI: 10.1091/mbc.e12-02-0103] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 05/02/2012] [Accepted: 05/11/2012] [Indexed: 01/06/2023] Open
Abstract
Regulation of axon growth, guidance, and branching is essential for constructing a correct neuronal network. R-Ras, a Ras-family small GTPase, has essential roles in axon formation and guidance. During axon formation, R-Ras activates a series of phosphatidylinositol 3-kinase signaling, inducing activation of a microtubule-assembly promoter-collapsin response mediator protein-2. However, signaling molecules linking R-Ras to actin cytoskeleton-regulating axonal morphology remain obscure. Here we identify afadin, an actin-binding protein harboring Ras association (RA) domains, as an effector of R-Ras inducing axon branching through F-actin reorganization. We observe endogenous interaction of afadin with R-Ras in cortical neurons during the stage of axonal development. Ectopic expression of afadin increases axon branch number, and the RA domains and the carboxyl-terminal F-actin binding domain are required for this action. RNA interference knockdown experiments reveal that knockdown of endogenous afadin suppressed both basal and R-Ras-mediated axon branching in cultured cortical neurons. Subcellular localization analysis shows that active R-Ras-induced translocation of afadin and its RA domains is responsible for afadin localizing to the membrane and inducing neurite development in Neuro2a cells. Overall, our findings demonstrate a novel signaling pathway downstream of R-Ras that controls axon branching.
Collapse
Affiliation(s)
- Nariaki Iwasawa
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Manabu Negishi
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Izumi Oinuma
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, 4-1-8 Honcho Kawaguchi, Saitama 332-0012, Japan
| |
Collapse
|
162
|
Jannie KM, Stipp CS, Weiner JA. ALCAM regulates motility, invasiveness, and adherens junction formation in uveal melanoma cells. PLoS One 2012; 7:e39330. [PMID: 22745734 PMCID: PMC3383762 DOI: 10.1371/journal.pone.0039330] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 05/23/2012] [Indexed: 01/19/2023] Open
Abstract
ALCAM, a member of the immunoglobulin superfamily, has been implicated in numerous developmental events and has been repeatedly identified as a marker for cancer metastasis. Previous studies addressing ALCAM's role in cancer have, however, yielded conflicting results. Depending on the tumor cell type, ALCAM expression has been reported to be both positively and negatively correlated with cancer progression and metastasis in the literature. To better understand how ALCAM might regulate cancer cell behavior, we utilized a panel of defined uveal melanoma cell lines with high or low ALCAM levels, and directly tested the effects of manipulating these levels on cell motility, invasiveness, and adhesion using multiple assays. ALCAM expression was stably silenced by shRNA knockdown in a high-ALCAM cell line (MUM-2B); the resulting cells displayed reduced motility in gap-closure assays and a reduction in invasiveness as measured by a transwell migration assay. Immunostaining revealed that the silenced cells were defective in the formation of adherens junctions, at which ALCAM colocalizes with N-cadherin and ß-catenin in native cells. Additionally, we stably overexpressed ALCAM in a low-ALCAM cell line (MUM-2C); intriguingly, these cells did not exhibit any increase in motility or invasiveness, indicating that ALCAM is necessary but not sufficient to promote metastasis-associated cell behaviors. In these ALCAM-overexpressing cells, however, recruitment of ß-catenin and N-cadherin to adherens junctions was enhanced. These data confirm a previously suggested role for ALCAM in the regulation of adherens junctions, and also suggest a mechanism by which ALCAM might differentially enhance or decrease invasiveness, depending on the type of cadherin adhesion complexes present in tissues surrounding the primary tumor, and on the cadherin status of the tumor cells themselves.
Collapse
Affiliation(s)
- Karry M. Jannie
- Department of Biology, The University of Iowa, Iowa City, Iowa, United States of America
| | - Christopher S. Stipp
- Department of Biology, The University of Iowa, Iowa City, Iowa, United States of America
| | - Joshua A. Weiner
- Department of Biology, The University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
163
|
Chatterjee S, Seifried L, Feigin ME, Gibbons DL, Scuoppo C, Lin W, Rizvi ZH, Lind E, Dissanayake D, Kurie J, Ohashi P, Muthuswamy SK. Dysregulation of cell polarity proteins synergize with oncogenes or the microenvironment to induce invasive behavior in epithelial cells. PLoS One 2012; 7:e34343. [PMID: 22529912 PMCID: PMC3329530 DOI: 10.1371/journal.pone.0034343] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 02/26/2012] [Indexed: 11/18/2022] Open
Abstract
Changes in expression and localization of proteins that regulate cell and tissue polarity are frequently observed in carcinoma. However, the mechanisms by which changes in cell polarity proteins regulate carcinoma progression are not well understood. Here, we report that loss of polarity protein expression in epithelial cells primes them for cooperation with oncogenes or changes in tissue microenvironment to promote invasive behavior. Activation of ErbB2 in cells lacking the polarity regulators Scribble, Dlg1 or AF-6, induced invasive properties. This cooperation required the ability of ErbB2 to regulate the Par6/aPKC polarity complex. Inhibition of the ErbB2-Par6 pathway was sufficient to block ErbB2-induced invasion suggesting that two polarity hits may be needed for ErbB2 to promote invasion. Interestingly, in the absence of ErbB2 activation, either a combined loss of two polarity proteins, or exposure of cells lacking one polarity protein to cytokines IL-6 or TNFα induced invasive behavior in epithelial cells. We observed the invasive behavior only when cells were plated on a stiff matrix (Matrigel/Collagen-1) and not when plated on a soft matrix (Matrigel alone). Cells lacking two polarity proteins upregulated expression of EGFR and activated Akt. Inhibition of Akt activity blocked the invasive behavior identifying a mechanism by which loss of polarity promotes invasion of epithelial cells. Thus, we demonstrate that loss of polarity proteins confers phenotypic plasticity to epithelial cells such that they display normal behavior under normal culture conditions but display aggressive behavior in response to activation of oncogenes or exposure to cytokines.
Collapse
Affiliation(s)
- Samit Chatterjee
- Ontario Cancer Institute, Campbell Family Institute for Breast Cancer Research, University of Toronto, Toronto, Ontario, Canada
- Cold Spring Harbor Laboratory, Watson School of Biological Sciences, Cold Spring Harbor, New York, United States of America
| | - Laurie Seifried
- Ontario Cancer Institute, Campbell Family Institute for Breast Cancer Research, University of Toronto, Toronto, Ontario, Canada
| | - Michael E. Feigin
- Cold Spring Harbor Laboratory, Watson School of Biological Sciences, Cold Spring Harbor, New York, United States of America
| | - Don L. Gibbons
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Claudio Scuoppo
- Cold Spring Harbor Laboratory, Watson School of Biological Sciences, Cold Spring Harbor, New York, United States of America
| | - Wei Lin
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Zain H. Rizvi
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Evan Lind
- Ontario Cancer Institute, Campbell Family Institute for Breast Cancer Research, University of Toronto, Toronto, Ontario, Canada
| | - Dilan Dissanayake
- Ontario Cancer Institute, Campbell Family Institute for Breast Cancer Research, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan Kurie
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Pam Ohashi
- Ontario Cancer Institute, Campbell Family Institute for Breast Cancer Research, University of Toronto, Toronto, Ontario, Canada
| | - Senthil K. Muthuswamy
- Ontario Cancer Institute, Campbell Family Institute for Breast Cancer Research, University of Toronto, Toronto, Ontario, Canada
- Cold Spring Harbor Laboratory, Watson School of Biological Sciences, Cold Spring Harbor, New York, United States of America
- * E-mail:
| |
Collapse
|
164
|
Tang VW, Brieher WM. α-Actinin-4/FSGS1 is required for Arp2/3-dependent actin assembly at the adherens junction. ACTA ACUST UNITED AC 2012; 196:115-30. [PMID: 22232703 PMCID: PMC3255975 DOI: 10.1083/jcb.201103116] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We have developed an in vitro assay to study actin assembly at cadherin-enriched cell junctions. Using this assay, we demonstrate that cadherin-enriched junctions can polymerize new actin filaments but cannot capture preexisting filaments, suggesting a mechanism involving de novo synthesis. In agreement with this hypothesis, inhibition of Arp2/3-dependent nucleation abolished actin assembly at cell-cell junctions. Reconstitution biochemistry using the in vitro actin assembly assay identified α-actinin-4/focal segmental glomerulosclerosis 1 (FSGS1) as an essential factor. α-Actinin-4 specifically localized to sites of actin incorporation on purified membranes and at apical junctions in Madin-Darby canine kidney cells. Knockdown of α-actinin-4 decreased total junctional actin and inhibited actin assembly at the apical junction. Furthermore, a point mutation of α-actinin-4 (K255E) associated with FSGS failed to support actin assembly and acted as a dominant negative to disrupt actin dynamics at junctional complexes. These findings demonstrate that α-actinin-4 plays an important role in coupling actin nucleation to assembly at cadherin-based cell-cell adhesive contacts.
Collapse
Affiliation(s)
- Vivian W Tang
- Department of Cell and Developmental Biology, University of Illinois, Urbana, IL 61801, USA
| | | |
Collapse
|
165
|
Paschoud S, Guillemot L, Citi S. Distinct domains of paracingulin are involved in its targeting to the actin cytoskeleton and regulation of apical junction assembly. J Biol Chem 2012; 287:13159-69. [PMID: 22315225 DOI: 10.1074/jbc.m111.315622] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Paracingulin is an M(r) 150-160 kDa cytoplasmic protein of vertebrate epithelial tight and adherens junctions and comprises globular head, coiled-coil rod, and globular tail domains. Unlike its homologous tight junction protein cingulin, paracingulin has been implicated in the control of junction assembly and has been localized at extrajunctional sites in association with actin filaments. Here we analyze the role of paracingulin domains, and specific regions within the head and rod domains, in the function and localization of paracingulin by inducible overexpression of exogenous proteins in epithelial Madin Darby canine kidney (MDCK) cells and by expression of mutated and chimeric constructs in Rat1 fibroblasts and MDCK cells. The overexpression of the rod + tail domains of paracingulin perturbs the development of the tight junction barrier and Rac1 activation during junction assembly by the calcium switch, indicating that regulation of junction assembly by paracingulin is mediated by these domains. Conversely, only constructs containing the head domain target to junctions in MDCK cells and Rat1 fibroblasts. Furthermore, expression of chimeric cingulin and paracingulin constructs in Rat1 fibroblasts and MDCK cells identifies specific sequences within the head and rod domains of paracingulin as critical for targeting to actin filaments and regulation of junction assembly, respectively. In summary, we characterize the functionally important domains of paracingulin that distinguish it from cingulin.
Collapse
Affiliation(s)
- Serge Paschoud
- Department of Molecular Biology, University of Geneva, 4 Boulevard d'Yvoy, 1205 Geneva, Switzerland
| | | | | |
Collapse
|
166
|
Li X, Lynn BD, Nagy JI. The effector and scaffolding proteins AF6 and MUPP1 interact with connexin36 and localize at gap junctions that form electrical synapses in rodent brain. Eur J Neurosci 2012; 35:166-81. [PMID: 22211808 DOI: 10.1111/j.1460-9568.2011.07947.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Electrical synapses formed by neuronal gap junctions composed of connexin36 (Cx36) occur in most major structures in the mammalian central nervous system. These synapses link ensembles of neurons and influence their network properties. Little is known about the macromolecular constituents of neuronal gap junctions or how transmission through electrical synapses is regulated at the level of channel conductance or gap junction assembly/disassembly. Such knowledge is a prerequisite to understanding the roles of gap junctions in neuronal circuitry. Gap junctions share similarities with tight and adhesion junctions in that all three reside at close plasma membrane appositions, and therefore may associate with similar structural and regulatory proteins. Previously, we reported that the tight junction-associated protein zonula occludens-1 (ZO-1) interacts with Cx36 and is localized at gap junctions. Here, we demonstrate that two proteins known to be associated with tight and adherens junctions, namely AF6 and MUPP1, are components of neuronal gap junctions in rodent brain. By immunofluorescence, AF6 and MUPP1 were co-localized with Cx36 in many brain areas. Co-immunoprecipitation and pull-down approaches revealed an association of Cx36 with AF6 and MUPP1, which required the C-terminus PDZ domain interaction motif of Cx36 for interaction with the single PDZ domain of AF6 and with the 10th PDZ domain of MUPP1. As AF6 is a target of the cAMP/Epac/Rap1 signalling pathway and MUPP1 is a scaffolding protein that interacts with CaMKII, the present results suggest that AF6 may be a target for cAMP/Epac/Rap1 signalling at electrical synapses, and that MUPP1 may contribute to anchoring CaMKII at these synapses.
Collapse
Affiliation(s)
- X Li
- Department of Physiology, Faculty of Medicine, University of Manitoba, 745 Bannatyne Ave., Winnipeg, Manitoba, Canada
| | | | | |
Collapse
|
167
|
Shimono Y, Rikitake Y, Mandai K, Mori M, Takai Y. Immunoglobulin superfamily receptors and adherens junctions. Subcell Biochem 2012; 60:137-170. [PMID: 22674071 DOI: 10.1007/978-94-007-4186-7_7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The immunogroblin (Ig) superfamily proteins characterized by the presence of Ig-like domains are involved in various cellular functions. The properties of the Ig-like domains to form rod-like structures and to bind specifically to other proteins make them ideal for cell surface receptors and cell adhesion molecules (CAMs). Ig-CAMs, nectins in mammals and Echinoid in Drosophila, are crucial components of cadherin-based adherens junctions in the epithelium. Nectins form cell-cell adhesion by their trans-interactions and recruit cadherins to the nectin-initiated cell-cell adhesion site to establish adherens junctions. Thereafter junction adhesion molecules, occludin, and claudins, are recruited to the apical side of adherens junctions to establish tight junctions. The recruitment of these molecules by nectins is mediated both by the direct and indirect interactions of afadin with many proteins, such as catenins, and zonula occludens proteins, and by the nectin-induced reorganization of the actin cytoskeleton. Nectins contribute to the formation of both homotypic and heterotypic types of cell-cell junctions, such as synapses in the brain, contacts between pigment and non-pigment cell layers of the ciliary epithelium in the eye, Sertoli cell-spermatid junctions in the testis, and sensory cells and supporting cells in the sensory organs. In addition, cis- and trans-interactions of nectins with various cell surface proteins, such as integrins, growth factor receptors, and nectin-like molecules (Necls) play important roles in the regulation of many cellular functions, such as cell polarization, movement, proliferation, differentiation, survival, and cell sorting. Furthermore, the Ig-CAMs are implicated in many human diseases including viral infections, ectodermal dysplasia, cancers, and Alzheimer's disease.
Collapse
Affiliation(s)
- Yohei Shimono
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, 650-0017, Kobe, Japan
| | | | | | | | | |
Collapse
|
168
|
Assimakopoulos SF, Papageorgiou I, Charonis A. Enterocytes’ tight junctions: From molecules to diseases. World J Gastrointest Pathophysiol 2011; 2:123-37. [PMID: 22184542 PMCID: PMC3241743 DOI: 10.4291/wjgp.v2.i6.123] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 08/26/2011] [Accepted: 10/31/2011] [Indexed: 02/06/2023] Open
Abstract
Tight junctions (TJs) are structures between cells where cells appear in the closest possible contact. They are responsible for sealing compartments when epithelial sheets are generated. They regulate the permeability of ions, (macro) molecules and cells via the paracellular pathway. Their structure at the electron microscopic level has been well known since the 1970s; however, only recently has their macromolecular composition been revealed. This review first examines the major macromolecular components of the TJs (occludin, claudins, junctional adhesion molecule and tricellulin) and then the associated macromolecules at the intracellular plaque [zonula occludens (ZO)-1, ZO-2, ZO-3, AF-6, cingulin, 7H6]. Emphasis is given to their interactions in order to begin to understand the mode of assembly of TJs. The functional significance of TJs is detailed and several mechanisms and factors involved are discussed briefly. Emphasis is given to the role of intestinal TJs and the alterations observed or speculated in diverse disease states. Specifically, intestinal TJs may exert a pathogenetic role in intestinal (inflammatory bowel disease, celiac disease) and extraintestinal diseases (diabetes type 1, food allergies, autoimmune diseases). Additionally, intestinal TJs may be secondarily disrupted during the course of diverse diseases, subsequently allowing the bacterial translocation phenomenon and promoting the systemic inflammatory response, which is often associated with clinical deterioration. The major questions in the field are highlighted.
Collapse
|
169
|
Fukumoto Y, Kurita S, Takai Y, Ogita H. Role of scaffold protein afadin dilute domain-interacting protein (ADIP) in platelet-derived growth factor-induced cell movement by activating Rac protein through Vav2 protein. J Biol Chem 2011; 286:43537-48. [PMID: 22027834 DOI: 10.1074/jbc.m111.308858] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cell movement is an important cellular function not only in physiological but also in pathological conditions. Although numerous studies have been conducted to reveal the mechanism of cell movement, the full picture has yet to be depicted, likely due to the complex features of cell movement. We show here that the scaffold protein afadin dilute domain-interacting protein (ADIP), an afadin-binding protein, is involved in the regulation of cell movement. ADIP localized at the leading edge of moving cells in response to platelet-derived growth factor (PDGF) and was required for the formation of the leading edge and the promotion of cell movement. Impaired cell movement observed in ADIP knockdown cells was not rescued by expression of an ADIP mutant that is incapable of binding to afadin, leading to the notion that the function of ADIP in moving cells depends on its interaction with afadin. Knockdown of ADIP as well as knockdown of afadin inhibited the activation of the small G protein Rac, which is important for the formation of the leading edge and the promotion of cell movement. Furthermore, ADIP interacted with Vav2, a GDP/GTP exchange factor for Rac, in a Src phosphorylation-dependent manner, suggesting that ADIP mediates the activation of Rac through Vav2. These results indicate that ADIP plays an essential role in PDGF-induced cell movement by interacting with afadin and Vav2 and regulating the activation of Rac.
Collapse
Affiliation(s)
- Yuri Fukumoto
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo 650-0017, Japan
| | | | | | | |
Collapse
|
170
|
Wee B, Johnston CA, Prehoda KE, Doe CQ. Canoe binds RanGTP to promote Pins(TPR)/Mud-mediated spindle orientation. ACTA ACUST UNITED AC 2011; 195:369-76. [PMID: 22024168 PMCID: PMC3206335 DOI: 10.1083/jcb.201102130] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The scaffolding protein Canoe regulates spindle orientation by binding to RanGTP and recruiting RanGTP and Mud to the cell cortex. Regulated spindle orientation maintains epithelial tissue integrity and stem cell asymmetric cell division. In Drosophila melanogaster neural stem cells (neuroblasts), the scaffolding protein Canoe (Afadin/Af-6 in mammals) regulates spindle orientation, but its protein interaction partners and mechanism of action are unknown. In this paper, we use our recently developed induced cell polarity system to dissect the molecular mechanism of Canoe-mediated spindle orientation. We show that a previously uncharacterized portion of Canoe directly binds the Partner of Inscuteable (Pins) tetratricopeptide repeat (TPR) domain. The Canoe–PinsTPR interaction recruits Canoe to the cell cortex and is required for activation of the PinsTPR-Mud (nuclear mitotic apparatus in mammals) spindle orientation pathway. We show that the Canoe Ras-association (RA) domains directly bind RanGTP and that both the CanoeRA domains and RanGTP are required to recruit Mud to the cortex and activate the Pins/Mud/dynein spindle orientation pathway.
Collapse
Affiliation(s)
- Brett Wee
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA
| | | | | | | |
Collapse
|
171
|
Benson DL, Huntley GW. Synapse adhesion: a dynamic equilibrium conferring stability and flexibility. Curr Opin Neurobiol 2011; 22:397-404. [PMID: 22019151 DOI: 10.1016/j.conb.2011.09.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 09/23/2011] [Accepted: 09/28/2011] [Indexed: 11/29/2022]
Abstract
Cell adhesion molecules (CAMs) linked to cytoskeleton generate stable cell-cell junctions. Cadherins provide a canonical example, but paradoxically, they participate in a multitude of transient and regulatable interactions. Their extracellular binding generates weak adhesion that is modified by clustering; interactions with F-actin are regulated, can be transient, and can alter F-actin dynamics. Additionally, cadherin recycling from the cell surface can modify the size and location of junctions and strength of adhesion. In epithelial cells, this ongoing dynamic behavior is important for maintaining stable junctions. Recent work supports that cadherins act similarly at synapses where their actions are likely to be shared by integrins and other actin-linked CAMs. Together the collaborative activities of such CAMs provide a stable, but flexible structure that can promote and support changes in synapse shape and size while maintaining stable junctions to permit information flow.
Collapse
Affiliation(s)
- Deanna L Benson
- Department of Neuroscience and the Friedman Brain Institute, Mount Sinai School of Medicine, New York, NY 10029, United States.
| | | |
Collapse
|
172
|
Kim J, Chang A, Dudak A, Federoff HJ, Lim ST. Characterization of nectin processing mediated by presenilin-dependent γ-secretase. J Neurochem 2011; 119:945-56. [PMID: 21910732 DOI: 10.1111/j.1471-4159.2011.07479.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Nectins play an important role in forming various intercellular junctions including synapses. This role is regulated by several secretases present at intercellular junctions. We have investigated presenilin (PS)-dependent secretase-mediated processing of nectins in PS1 KO cells and primary hippocampal neurons. The loss of PS1/γ-secretase activity delayed the processing of nectin-1 and caused the accumulation of its full-length and C-terminal fragments. Over-expression of PS2 in PS1 KO cells compensated for the loss of PS1, suggesting that PS2 also has the ability to regulate nectin-1 processing. In mouse brain slices, a pronounced increase in levels of 30 and 24 kDa C-terminal fragments in response to chemical long-term potentiation was observed. The mouse brain synaptosomal fractionation study indicated that nectin-1 localized to post-synaptic and preferentially pre-synaptic membranes and that shedding occurs in both compartments. These data suggest that nectin-1 shedding and PS-dependent intramembrane cleavage occur at synapses, and is a regulated event during conditions of synaptic plasticity in the brain. Point mutation analysis identified several residues within the transmembrane domain that play a critical role in the positioning of cleavage sites by ectodomain sheddases. Nectin-3, which forms hetero-trans-dimers with nectin-1, also undergoes intramembrane cleavage mediated by PS1/γ-secretase, suggesting that PS1/γ-secreatse activity regulates synapse formation and remodeling by nectin processing.
Collapse
Affiliation(s)
- Jinsook Kim
- Department of Neuroscience, Georgetown University Medical Center, NW, Washington, District of Columbia, USA
| | | | | | | | | |
Collapse
|
173
|
González-Mariscal L, Quirós M, Díaz-Coránguez M. ZO proteins and redox-dependent processes. Antioxid Redox Signal 2011; 15:1235-53. [PMID: 21294657 DOI: 10.1089/ars.2011.3913] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
SIGNIFICANCE ZO-1, ZO-2, and ZO-3 are scaffold proteins of the tight junction (TJ) that belong to the MAGUK protein family characterized for exhibiting PDZ, SH3, and GuK domains. ZO proteins are present only in multicellular organisms, being the placozoa the first to have them. ZO proteins associate among themselves and with other integral and adaptor proteins of the TJ, of the ZA and of gap junctions, as with numerous signaling proteins and the actin cytoskeleton. ZO proteins are also present at the nucleus of proliferating cells. RECENT ADVANCES Oxidative stress disassembles the TJs of endothelial and epithelial cells. CRITICAL ISSUES Oxidative stress alters ZO proteins expression and localization, in conditions like hypoxia, bacterial and viral infections, vitamin deficiencies, age-related diseases, diabetes and inflammation, alcohol and tobacco consumption. FUTURE DIRECTIONS Molecules present in the signaling pathways triggered by oxidative stress can be targets for therapeutic intervention.
Collapse
Affiliation(s)
- Lorenza González-Mariscal
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies (Cinvestav), Mexico DF, México.
| | | | | |
Collapse
|
174
|
Citi S, Spadaro D, Schneider Y, Stutz J, Pulimeno P. Regulation of small GTPases at epithelial cell-cell junctions. Mol Membr Biol 2011; 28:427-44. [DOI: 10.3109/09687688.2011.603101] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
175
|
Zaichick SV, Bohannon KP, Smith GA. Alphaherpesviruses and the cytoskeleton in neuronal infections. Viruses 2011; 3:941-81. [PMID: 21994765 PMCID: PMC3185784 DOI: 10.3390/v3070941] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 06/03/2011] [Accepted: 06/17/2011] [Indexed: 12/13/2022] Open
Abstract
Following infection of exposed peripheral tissues, neurotropic alphaherpesviruses invade nerve endings and deposit their DNA genomes into the nuclei of neurons resident in ganglia of the peripheral nervous system. The end result of these events is the establishment of a life-long latent infection. Neuroinvasion typically requires efficient viral transmission through a polarized epithelium followed by long-distance transport through the viscous axoplasm. These events are mediated by the recruitment of the cellular microtubule motor proteins to the intracellular viral particle and by alterations to the cytoskeletal architecture. The focus of this review is the interplay between neurotropic herpesviruses and the cytoskeleton.
Collapse
Affiliation(s)
- Sofia V Zaichick
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | | | | |
Collapse
|
176
|
Tanaka-Okamoto M, Hori K, Ishizaki H, Itoh Y, Onishi S, Yonemura S, Takai Y, Miyoshi J. Involvement of afadin in barrier function and homeostasis of mouse intestinal epithelia. J Cell Sci 2011; 124:2231-40. [PMID: 21652626 PMCID: PMC3115770 DOI: 10.1242/jcs.081000] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Afadin interacts with the cytoplasmic region of nectins, which are immunoglobulin-like cell adhesion molecules at adherens junctions, and links them to the actin cytoskeleton. Afadin regulates activities of cells in culture such as directional motility, proliferation and survival. We used Cre-loxP technology to generate mice conditionally lacking afadin specifically in the intestinal epithelia after birth. The loss of afadin caused increased paracellular permeability in the intestinal mucosa and enhanced susceptibility to the tissue destruction induced by dextran sulfate sodium. The junctional architecture of the intestinal epithelia appeared to be preserved, whereas the deficiency of afadin caused the mislocalization of nectin-2 and nectin-3 from adherens junctions to basolateral membrane domains but not that of other components of apical junctions. By contrast, such phenotypic changes were undetected in mice lacking nectin-2, nectin-3 or both. These findings suggest that afadin plays crucial roles, independently of the role as the nectin-afadin module, in barrier function and homeostasis of the intestinal epithelia once the epithelial structure has been established.
Collapse
Affiliation(s)
- Miki Tanaka-Okamoto
- Department of Molecular Biology, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka 537-8511, Japan
| | | | | | | | | | | | | | | |
Collapse
|
177
|
Fournier G, Cabaud O, Josselin E, Chaix A, Adélaïde J, Isnardon D, Restouin A, Castellano R, Dubreuil P, Chaffanet M, Birnbaum D, Lopez M. Loss of AF6/afadin, a marker of poor outcome in breast cancer, induces cell migration, invasiveness and tumor growth. Oncogene 2011; 30:3862-74. [DOI: 10.1038/onc.2011.106] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
178
|
Slováková J, Carmena A. Canoe functions at the CNS midline glia in a complex with Shotgun and Wrapper-Nrx-IV during neuron-glia interactions. Development 2011; 138:1563-71. [PMID: 21389054 DOI: 10.1242/dev.056192] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Vertebrates and insects alike use glial cells as intermediate targets to guide growing axons. Similar to vertebrate oligodendrocytes, Drosophila midline glia ensheath and separate axonal commissures. Neuron-glia interactions are crucial during these events, although the proteins involved remain largely unknown. Here, we show that Canoe (Cno), the Drosophila ortholog of AF-6, and the DE-cadherin Shotgun (Shg) are highly restricted to the interface between midline glia and commissural axons. cno mutant analysis, genetic interactions and co-immunoprecipitation assays unveil Cno function as a novel regulator of neuron-glia interactions, forming a complex with Shg, Wrapper and Neurexin IV, the homolog of vertebrate Caspr/paranodin. Our results also support additional functions of Cno, independent of adherens junctions, as a regulator of adhesion and signaling events in non-epithelial tissues.
Collapse
Affiliation(s)
- Jana Slováková
- Instituto de Neurociencias de Alicante, CSIC/UMH, Sant Joan d'Alacant, Alicante, Spain
| | | |
Collapse
|
179
|
Zhang L, Jouret F, Rinehart J, Sfakianos J, Mellman I, Lifton RP, Young LH, Caplan MJ. AMP-activated protein kinase (AMPK) activation and glycogen synthase kinase-3β (GSK-3β) inhibition induce Ca2+-independent deposition of tight junction components at the plasma membrane. J Biol Chem 2011; 286:16879-90. [PMID: 21383016 DOI: 10.1074/jbc.m110.186932] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Extracellular Ca(2+) is essential for the development of stable epithelial tight junctions. We find that in the absence of extracellular Ca(2+), AMP-activated protein kinase (AMPK) activation and glycogen synthase kinase (GSK)-3β inhibition independently induce the localization of epithelial tight junction components to the plasma membrane. The Ca(2+)-independent deposition of junctional proteins induced by AMPK activation and GSK-3β inhibition is independent of E-cadherin. Furthermore, the nectin-afadin system is required for the deposition of tight junction components induced by AMPK activation, but it is not required for that induced by GSK-3β inhibition. Phosphorylation studies demonstrate that afadin is a substrate for AMPK. These data demonstrate that two kinases involved in regulating cell growth and metabolism act through distinct pathways to influence the deposition of the components of epithelial tight junctions.
Collapse
Affiliation(s)
- Li Zhang
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | | | | | | | | | | | | | |
Collapse
|
180
|
Dudak A, Kim J, Cheong B, Federoff HJ, Lim ST. Membrane palmitoylated proteins regulate trafficking and processing of nectins. Eur J Cell Biol 2011; 90:365-75. [PMID: 21371776 DOI: 10.1016/j.ejcb.2011.01.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 01/07/2011] [Accepted: 01/07/2011] [Indexed: 11/25/2022] Open
Abstract
Nectins are cell-cell adhesion molecules involved in the formation of various intercellular junctions and the establishment of apical-basal polarity at cell-cell adhesion sites. To have a better understanding of the roles of nectins in the formation of cell-cell junctions, we searched for new cytoplasmic binding partners for nectin. We report that nectin-1α associates with membrane palmitoylated protein 3 (MPP3), one of the human homologues of a Drosophila tumor suppressor gene, Disc large. Two major forms of MPP3 at 66 and 98 kDa were detected, in conjunction with nectin-1α, suggesting that an association between the two may occur in various cell types. Nectin-1α recruits MPP3 to cell-cell contact sites, mediated by a PDZ-binding motif at the carboxyl terminus of nectin-1α. Association with MPP3 increases cell surface expression of nectin-1α and enhances nectin-1α ectodomain shedding, indicating that MPP3 regulates trafficking and processing of nectin-1α. Further study showed that MPP3 interacts with nectin-3α, but not with nectin-2α, showing that the association of nectins with MPP3 is isoform-specific. MPP5, another MPP family member, interacts with nectins with varying affinity and facilitates surface expression of nectin-1α, nectin-2α, and nectin-3α. These data suggest that wide interactions between nectins and MPP family members may occur in various cell-cell junctions and that these associations may regulate trafficking and processing of nectins.
Collapse
Affiliation(s)
- Amanda Dudak
- Department of Neuroscience, Georgetown University Medical Center, 3970 Reservoir Rd. NW, Washington, DC 20057, USA
| | | | | | | | | |
Collapse
|
181
|
Narita H, Yamamoto Y, Suzuki M, Miyazaki N, Yoshida A, Kawai K, Iwasaki K, Nakagawa A, Takai Y, Sakisaka T. Crystal Structure of the cis-Dimer of Nectin-1: implications for the architecture of cell-cell junctions. J Biol Chem 2011; 286:12659-69. [PMID: 21325282 DOI: 10.1074/jbc.m110.197368] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In multicellular organisms, cells are interconnected by cell adhesion molecules. Nectins are immunoglobulin (Ig)-like cell adhesion molecules that mediate homotypic and heterotypic cell-cell adhesion, playing key roles in tissue organization. To mediate cell-cell adhesion, nectin molecules dimerize in cis on the surface of the same cell, followed by trans-dimerization of the cis-dimers between the neighboring cells. Previous cell biological studies deduced that the first Ig-like domain of nectin and the second Ig-like domain are involved in trans-dimerization and cis-dimerization, respectively. However, to understand better the steps involved in nectin adhesion, the structural basis for the dimerization of nectin must be determined. In this study, we determined the first crystal structure of the entire extracellular region of nectin-1. In the crystal, nectin-1 formed a V-shaped homophilic dimer through the first Ig-like domain. Structure-based site-directed mutagenesis of the first Ig-like domain identified four essential residues that are involved in the homophilic dimerization. Upon mutating the four residues, nectin-1 significantly decreased cis-dimerization on the surface of cultured cells and abolished the homophilic and heterophilic adhesion activities. These results indicate that, in contrast with the previous notion, our structure represents a cis-dimer. Thus, our findings clearly reveal the structural basis for the cis-dimerization of nectins through the first Ig-like domains.
Collapse
Affiliation(s)
- Hirotaka Narita
- Laboratory of Supramolecular Crystallography, Research Center for Structural and Functional Proteomics, Institute for Protein Research, Osaka University, Suita 565-0871, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Daane JM, Enders AC, Downs KM. Mesothelium of the murine allantois exhibits distinct regional properties. J Morphol 2011; 272:536-56. [PMID: 21284019 DOI: 10.1002/jmor.10928] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 10/18/2010] [Accepted: 10/25/2010] [Indexed: 11/06/2022]
Abstract
The rodent allantois is thought to be unique amongst mammals in not having an endodermal component. Here, we have investigated the mesothelium, or outer surface, of murine umbilical precursor tissue, the allantois (∼7.25-8.5 days postcoitum, dpc) to discover whether it exhibits the properties of an epithelium. A combination of morphology, challenge with biotinylated dextran amines (BDAs), and immunohistochemistry revealed that the mesothelium of the mouse allantois exhibits distinct regional properties. By headfold stages (∼7.75-8.0 dpc), distal mesothelium was generally squamous in shape, and highly permeable to BDA challenge, whereas ventral proximal mesothelium, referred to as "ventral cuboidal mesothelium" (VCM) for the characteristic cuboidal shape of its cells, was relatively impermeable. Although "dorsal cuboidal mesothelium" (DCM) resembled the VCM in cell shape, its permeability to BDA was intermediate between the other two regions. Results of immunostaining for Zonula Occludens-1 (ZO-1) and Epithelial-cadherin (E-cadherin), together with transmission electron microscopy (TEM), suggested that impermeability in the VCM may be due to greater cellular contact area between cells and close packing rather than to maturity of tight junctions, the latter of which, by comparison with the visceral yolk sac, appeared to be rare or absent from the allantoic surface. Both VCM and DCM exhibited an ultrastructure more favorable for protein synthesis than did the distal squamous mesothelium; however, at most stages, VCM exhibited robust afadin (AF-6), whereas the DCM uniquely contained alpha-4-integrin. These observations demonstrate that the allantoic mesothelium is not a conventional epithelium but possesses regional ultrastructural, functional and molecular differences that may play important roles in the correct deployment of the umbilical cord and its associated vascular, hematopoietic, and other cell types.
Collapse
Affiliation(s)
- Jacob M Daane
- Department of Anatomy, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin 53706, USA
| | | | | |
Collapse
|
183
|
Rikitake Y, Takai Y. Directional Cell Migration. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2011; 287:97-143. [DOI: 10.1016/b978-0-12-386043-9.00003-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
184
|
PLEKHA7 is an adherens junction protein with a tissue distribution and subcellular localization distinct from ZO-1 and E-cadherin. PLoS One 2010; 5:e12207. [PMID: 20808826 PMCID: PMC2924883 DOI: 10.1371/journal.pone.0012207] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Accepted: 07/22/2010] [Indexed: 01/13/2023] Open
Abstract
The pleckstrin-homology-domain-containing protein PLEKHA7 was recently identified as a protein linking the E-cadherin-p120 ctn complex to the microtubule cytoskeleton. Here we characterize the expression, tissue distribution and subcellular localization of PLEKHA7 by immunoblotting, immunofluorescence microscopy, immunoelectron microscopy, and northern blotting in mammalian tissues. Anti-PLEKHA7 antibodies label the junctional regions of cultured kidney epithelial cells by immunofluorescence microscopy, and major polypeptides of Mr ∼135 kDa and ∼145 kDa by immunoblotting of lysates of cells and tissues. Two PLEKHA7 transcripts (∼5.5 kb and ∼6.5 kb) are detected in epithelial tissues. PLEKHA7 is detected at epithelial junctions in sections of kidney, liver, pancreas, intestine, retina, and cornea, and its tissue distribution and subcellular localization are distinct from ZO-1. For example, PLEKHA7 is not detected within kidney glomeruli. Similarly to E-cadherin, p120 ctn, β-catenin and α-catenin, PLEKHA7 is concentrated in the apical junctional belt, but unlike these adherens junction markers, and similarly to afadin, PLEKHA7 is not localized along the lateral region of polarized epithelial cells. Immunoelectron microscopy definitively establishes that PLEKHA7 is localized at the adherens junctions in colonic epithelial cells, at a mean distance of 28 nm from the plasma membrane. In summary, we show that PLEKHA7 is a cytoplasmic component of the epithelial adherens junction belt, with a subcellular localization and tissue distribution that is distinct from that of ZO-1 and most AJ proteins, and we provide the first description of its distribution and localization in several tissues.
Collapse
|
185
|
Matsuda M, Yamashita JK, Tsukita S, Furuse M. abLIM3 is a novel component of adherens junctions with actin-binding activity. Eur J Cell Biol 2010; 89:807-16. [PMID: 20709423 DOI: 10.1016/j.ejcb.2010.07.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2010] [Revised: 07/09/2010] [Accepted: 07/14/2010] [Indexed: 01/26/2023] Open
Abstract
The interactions of adhesion molecules with dense actin filaments via cytoplasmic plaque proteins are crucial for the adhesive function of adherens junctions (AJs) in epithelial and endothelial cells. Using localization-based expression cloning, we identified abLIM3, a member of the actin-binding LIM (abLIM) protein family, as a component of the junctional complex. Immunolocalization studies revealed that abLIM3 was localized at AJs in limited cell types, including hepatocytes, bronchial epithelial cells, mesothelial cells and endothelial cells lining muscular tissues. Deletion mutant analyses in cultured cells showed that the C-terminal dematin-like domain of abLIM3, which bound to actin filaments in vitro, was colocalized with phalloidin-stained filamentous actin, whereas the N-terminal LIM domains of abLIM3 were sufficient for recruitment to cell-cell contacts. These results suggest that abLIM3 is involved in anchoring LIM domain-binding components of AJs to circumferential actin bundles in specific cell types.
Collapse
Affiliation(s)
- Miho Matsuda
- Department of Cell Biology, Faculty of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | | | | | | |
Collapse
|
186
|
Tallafuss A, Constable JRL, Washbourne P. Organization of central synapses by adhesion molecules. Eur J Neurosci 2010; 32:198-206. [PMID: 20646051 DOI: 10.1111/j.1460-9568.2010.07340.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Synapses are the primary means for transmitting information from one neuron to the next. They are formed during the development of the nervous system, and the formation of appropriate synapses is crucial for the establishment of neuronal circuits that underlie behavior and cognition. Understanding how synapses form and are maintained will allow us to address developmental disorders such as autism, mental retardation and possibly also psychological disorders. A number of biochemical and proteomic studies have revealed a diverse and vast assortment of molecules that are present at the synapse. It is now important to untangle this large array of proteins and determine how it assembles into a functioning unit. Here we focus on recent reports describing how synaptic cell adhesion molecules interact with and organize the presynaptic and postsynaptic specializations of both excitatory and inhibitory central synapses.
Collapse
|
187
|
Liedtke M, Ayton PM, Somervaille TCP, Smith KS, Cleary ML. Self-association mediated by the Ras association 1 domain of AF6 activates the oncogenic potential of MLL-AF6. Blood 2010; 116:63-70. [PMID: 20395419 PMCID: PMC2904581 DOI: 10.1182/blood-2009-09-243386] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2009] [Accepted: 04/04/2010] [Indexed: 11/20/2022] Open
Abstract
MLL is a common target for chromosomal translocations associated with acute leukemia resulting in its fusion with a large variety of nuclear or cytoplasmic proteins that may activate its oncogenic properties by distinct but poorly understood mechanisms. The MLL-AF6 fusion gene represents the most common leukemogenic fusion of mixed lineage leukemia (MLL) to a cytoplasmic partner protein. Here, we identified a highly conserved Ras association (RA1) domain at the amino-terminus of AF6 as the minimal region sufficient for MLL-AF6 mediated myeloid progenitor immortalization in vitro and short latency leukemogenesis in vivo. Moreover, the ability of RA1 to activate MLL oncogenesis is conserved with its Drosophila ortholog, Canoe. Although the AF6 RA1 domain has previously been defined as an interaction surface for guanosine triphosphate-bound Ras, single amino acid substitutions known to abolish the AF6-Ras interaction did not abrogate MLL-AF6-mediated oncogenesis. Furthermore, fusion of MLL to heterologous RA domains of c-Raf1 or RalGDS, or direct fusion of MLL to constitutively active K-RAS, H-RAS, or RAP1 was not sufficient for oncogenic activation of MLL. Rather, the AF6 RA1 domain efficiently mediated self-association, suggesting that constitutive MLL self-association is a more common pathogenic mechanism for MLL oncogenesis than indicated by previous studies of rare MLL fusion partners.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Binding Sites/genetics
- Blotting, Western
- Bone Marrow Cells/cytology
- Bone Marrow Cells/metabolism
- Cell Line
- Cell Transformation, Neoplastic/genetics
- Cells, Cultured
- Genetic Vectors/genetics
- Humans
- Kinesins/genetics
- Kinesins/metabolism
- Leukemia, Experimental/genetics
- Leukemia, Experimental/metabolism
- Leukemia, Experimental/pathology
- Leukemia, Myeloid/genetics
- Leukemia, Myeloid/metabolism
- Leukemia, Myeloid/pathology
- Mice
- Mice, Inbred C57BL
- Molecular Sequence Data
- Mutation
- Myeloid-Lymphoid Leukemia Protein/genetics
- Myeloid-Lymphoid Leukemia Protein/metabolism
- Myosins/genetics
- Myosins/metabolism
- Neoplasm Transplantation
- Retroviridae/genetics
- Sequence Homology, Amino Acid
- Transduction, Genetic
- ras Proteins/genetics
- ras Proteins/metabolism
Collapse
Affiliation(s)
- Michaela Liedtke
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|
188
|
Tawa H, Rikitake Y, Takahashi M, Amano H, Miyata M, Satomi-Kobayashi S, Kinugasa M, Nagamatsu Y, Majima T, Ogita H, Miyoshi J, Hirata KI, Takai Y. Role of Afadin in Vascular Endothelial Growth Factor– and Sphingosine 1-Phosphate–Induced Angiogenesis. Circ Res 2010; 106:1731-42. [DOI: 10.1161/circresaha.110.216747] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale
:
Angiogenesis contributes to physiological and pathological conditions, including atherosclerosis. The Rap1 small G protein regulates vascular integrity and angiogenesis. However, little is known about the effectors of Rap1 involved in angiogenesis. It is not known whether afadin, an adherens junction protein that connects immunoglobulin-like adhesion molecule nectins to the actin cytoskeleton and binds activated Rap1, plays a role in angiogenesis.
Objective
:
We investigated the role of endothelial afadin in angiogenesis and attempted to clarify the underlying molecular mechanism.
Methods and Results
:
Treatment of human umbilical vein endothelial cells (HUVECs) with vascular endothelial growth factor (VEGF) and sphingosine 1-phosphate (S1P) induced the activation of Rap1. Activated Rap1 regulated intracellular localization of afadin. Knockdown of Rap1 or afadin by small interfering RNA inhibited the VEGF- and S1P-induced capillary-like network formation, migration, and proliferation, and increased the serum deprivation-induced apoptosis of HUVECs. Knockdown of Rap1 or afadin decreased the accumulation of adherens and tight junction proteins to the cell–cell contact sites. Rap1 regulated the interaction between afadin and phosphatidylinositol 3-kinase (PI3K), recruitment of the afadin–PI3K complex to the leading edge, and the activation of Akt, indicating the involvement of Rap1 and afadin in the PI3K–Akt signaling pathway. Binding of afadin to Rap1 regulated the activity of Rap1 in a positive-feedback manner. In vivo, conditional deletion of afadin in mouse vascular endothelium using a Cre-loxP system impaired the VEGF- and S1P-induced angiogenesis.
Conclusions
:
These results demonstrate a novel molecular mechanism by which Rap1 and afadin regulate the VEGF- and S1P-induced angiogenesis.
Collapse
Affiliation(s)
- Hideto Tawa
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Yoshiyuki Rikitake
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Motonori Takahashi
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Hisayuki Amano
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Muneaki Miyata
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Seimi Satomi-Kobayashi
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Mitsuo Kinugasa
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Yuichi Nagamatsu
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Takashi Majima
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Hisakazu Ogita
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Jun Miyoshi
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Ken-ichi Hirata
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Yoshimi Takai
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| |
Collapse
|
189
|
Kim J, Lilliehook C, Dudak A, Prox J, Saftig P, Federoff HJ, Lim ST. Activity-dependent alpha-cleavage of nectin-1 is mediated by a disintegrin and metalloprotease 10 (ADAM10). J Biol Chem 2010; 285:22919-26. [PMID: 20501653 DOI: 10.1074/jbc.m110.126649] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Nectin-1 is known to undergo ectodomain shedding by alpha-secretase and subsequent proteolytic processing by gamma-secretase. How secretase-mediated cleavage of nectin-1 is regulated in neuronal cells and how nectin-1 cleavage affects synaptic adhesion is poorly understood. We have investigated alpha-and gamma-secretase-mediated processing of nectin-1 in primary cortical neurons and identified which protease acts as a alpha-secretase. We report here that NMDA receptor activation, but not stimulation of AMPA or metabotropic glutamate receptors, resulted in robust alpha- and gamma-secretase cleavage of nectin-1 in mature cortical neurons. Cleavage of nectin-1 required influx of Ca(2+) through the NMDA receptor, and activation of calmodulin, but was not dependent on calcium/calmodulin-dependent protein kinase II (CaMKII) activation. We found that ADAM10 is the major secretase responsible for nectin-1 ectodomain cleavage in neurons and the brain. These observations suggest that alpha- and gamma-secretase processing of nectin-1 is a Ca(2+)/calmodulin-regulated event that occurs under conditions of activity-dependent synaptic plasticity and ADAM10 and gamma-secretase are responsible for these cleavage events.
Collapse
Affiliation(s)
- Jinsook Kim
- Neuroscience Department, Georgetown University Medical Center, Washington, D. C. 20057, USA
| | | | | | | | | | | | | |
Collapse
|
190
|
Guerin CM, Kramer SG. Cytoskeletal remodeling during myotube assembly and guidance: coordinating the actin and microtubule networks. Commun Integr Biol 2010; 2:452-7. [PMID: 19907716 DOI: 10.4161/cib.2.5.9158] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2009] [Accepted: 06/02/2009] [Indexed: 11/19/2022] Open
Abstract
The formation of a multinucleated muscle fiber from individual myoblasts is a complex morphological event that requires dramatic cytoskeletal rearrangements. This multistep process includes myoblast fusion, myotube migration and elongation, myotube target recognition, and finally attachment to form a stable adhesion complex. Many of the studies directed towards understanding the developmental process of muscle morphogenesis at the cellular level have relied on forward genetic screens in model systems such as Drosophila melanogaster for mutations affecting individual stages in myogenesis. Through the analyses of these gene products, proteins that regulate the actin or microtubule cytoskeleton have emerged as important players in each of these steps. We recently demonstrated that RacGAP50C, an essential protein that functions as a cytoskeletal regulator during cell division, also plays an important role in organizing the polarized microtubule network in the elongating myotube. Here we review the current literature regarding Drosophila myogenesis and illustrate several steps of muscle development with respect to the diverse roles that the cytoskeleton plays during this process. Furthermore, we discuss the significance of cytoskeletal coordination during these multiple steps.
Collapse
Affiliation(s)
- Colleen M Guerin
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, Piscataway, NJ 08854, USA
| | | |
Collapse
|
191
|
Stiles KM, Krummenacher C. Glycoprotein D actively induces rapid internalization of two nectin-1 isoforms during herpes simplex virus entry. Virology 2010; 399:109-119. [PMID: 20089288 DOI: 10.1016/j.virol.2009.12.034] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Revised: 11/16/2009] [Accepted: 12/23/2009] [Indexed: 01/22/2023]
Abstract
Entry of herpes simplex virus (HSV) occurs either by fusion at the plasma membrane or by endocytosis and fusion with an endosome. Binding of glycoprotein D (gD) to a receptor such as nectin-1 is essential in both cases. We show that virion gD triggered the rapid down-regulation of nectin-1 with kinetics similar to those of virus entry. In contrast, nectin-1 was not constitutively recycled from the surface of uninfected cells. Both the nectin-1alpha and beta isoforms were internalized in response to gD despite having different cytoplasmic tails. However, deletion of the nectin-1 cytoplasmic tail slowed down-regulation of nectin-1 and internalization of virions. These data suggest that nectin-1 interaction with a cytoplasmic protein is not required for its down-regulation. Overall, this study shows that gD binding actively induces the rapid internalization of various forms of nectin-1. We suggest that HSV activates a nectin-1 internalization pathway to use for endocytic entry.
Collapse
Affiliation(s)
- Katie M Stiles
- Department of Microbiology, School of Dental Medicine University of Pennsylvania, 240 S. 40th St., Philadelphia, PA 19104, USA.
| | - Claude Krummenacher
- Department of Biochemistry, School of Dental Medicine University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
192
|
Banga SS, Peng L, Dasgupta T, Palejwala V, Ozer HL. PHF10 is required for cell proliferation in normal and SV40-immortalized human fibroblast cells. Cytogenet Genome Res 2010; 126:227-42. [PMID: 20068294 PMCID: PMC3711003 DOI: 10.1159/000251960] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2009] [Indexed: 01/28/2023] Open
Abstract
Normal human diploid fibroblasts have limited life span in culture and undergo replicative senescence after 50-60 population doublings. On the contrary, cancer cells typically divide indefinitely and are immortal. Expression of SV40 large T and small t antigens in human fibroblasts transiently extends their life span by 20-30 population doublings and facilitates immortalization. We have identified a rearrangement in chromosome 6 shared by SV40-transformed human fibroblasts. Rearrangements involving chromosome 6 are among the most frequent in human carcinogenesis. In this paper, we extend analysis of the 6q26-q27 region, a putative site for a growth suppressor gene designated SEN6 involved in immortalization of SV40-transformed cells. Detailed molecular characterization of the rearranged chromosomes (6q*, normal appearing; and 6q(t), translocated) in the SV40-immortalized cell line HALneo by isolating each of these 2 chromosomes in mouse/HAL somatic cell hybrids is presented. Analysis of these mouse/HAL somatic cell hybrids with polymorphic and nonpolymorphic markers revealed that the 6q* has undergone a chromosomal break in the MLLT4 gene (alias AF6). This result in conjunction with previous published observations leads us to conclude that SEN6 lies between MLLT4 and TBP at chromosomal region 6q27. Examination of different genes (MLLT4, DLL1, FAM120B, PHF10) located within this interval that are expressed in HS74 normal fibroblast cells reveals that overexpression of epitope-tagged truncated PHF10 cDNAs resulted in reduced cell proliferation in multiple cell lines. Paradoxically, down-regulation of PHF10 by RNAi also resulted in loss of cell proliferation in normal fibroblast cells, indicating PHF10 function is required for cell growth. Taken together, these observations suggest that decreased cell proliferation with epitope-tagged truncated PHF10 proteins may be due to dominant negative effects or due to unregulated expression of these mutant proteins. Hence we conclude that PHF10 is not SEN6 but is required for cell growth.
Collapse
Affiliation(s)
- S S Banga
- Department of Microbiology and Molecular Genetics, New Jersey Medical School-University Hospital Cancer Center, University of Medicine and Dentistry of New Jersey, Newark, NJ 07103, USA. banga @ umdnj.edu
| | | | | | | | | |
Collapse
|
193
|
Ooshio T, Kobayashi R, Ikeda W, Miyata M, Fukumoto Y, Matsuzawa N, Ogita H, Takai Y. Involvement of the interaction of afadin with ZO-1 in the formation of tight junctions in Madin-Darby canine kidney cells. J Biol Chem 2009; 285:5003-12. [PMID: 20008323 DOI: 10.1074/jbc.m109.043760] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Tight junctions (TJs) and adherens junctions (AJs) are major junctional apparatuses in epithelial cells. Claudins and junctional adhesion molecules (JAMs) are major cell adhesion molecules (CAMs) at TJs, whereas cadherins and nectins are major CAMs at AJs. Claudins and JAMs are associated with ZO proteins, whereas cadherins are associated with beta- and alpha-catenins, and nectins are associated with afadin. We previously showed that nectins first form cell-cell adhesions where the cadherin-catenin complex is recruited to form AJs, followed by the recruitment of the JAM-ZO and claudin-ZO complexes to the apical side of AJs to form TJs. It is not fully understood how TJ components are recruited to the apical side of AJs. We studied the roles of afadin and ZO-1 in the formation of TJs in Madin-Darby canine kidney (MDCK) cells. Before the formation of TJs, ZO-1 interacted with afadin through the two proline-rich regions of afadin and the SH3 domain of ZO-1. During and after the formation of TJs, ZO-1 dissociated from afadin and associated with JAM-A. Knockdown of afadin impaired the formation of both AJs and TJs in MDCK cells, whereas knockdown of ZO-1 impaired the formation of TJs, but not AJs. Re-expression of full-length afadin restored the formation of both AJs and TJs in afadin-knockdown MDCK cells, whereas re-expression of afadin-DeltaPR1-2, which is incapable of binding to ZO-1, restored the formation of AJs, but not TJs. These results indicate that the transient interaction of afadin with ZO-1 is necessary for the formation of TJs in MDCK cells.
Collapse
Affiliation(s)
- Takako Ooshio
- Department of Molecular Biology and Biochemistry, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | |
Collapse
|
194
|
Miyata M, Ogita H, Komura H, Nakata S, Okamoto R, Ozaki M, Majima T, Matsuzawa N, Kawano S, Minami A, Waseda M, Fujita N, Mizutani K, Rikitake Y, Takai Y. Localization of nectin-free afadin at the leading edge and its involvement in directional cell movement induced by platelet-derived growth factor. J Cell Sci 2009; 122:4319-29. [DOI: 10.1242/jcs.048439] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Afadin is an actin-filament-binding protein that binds to nectin, an immunoglobulin-like cell-cell adhesion molecule, and plays an important role in the formation of adherens junctions. Here, we show that afadin, which did not bind to nectin and was localized at the leading edge of moving cells, has another role: enhancement of the directional, but not random, cell movement. When NIH3T3 cells were stimulated with platelet-derived growth factor (PDGF), afadin colocalized with PDGF receptor, αvβ3 integrin and nectin-like molecule-5 at the leading edge and facilitated the formation of leading-edge structures and directional cell movement in the direction of PDGF stimulation. However, these phenotypes were markedly perturbed by knockdown of afadin, and were dependent on the binding of afadin to active Rap1. Binding of Rap1 to afadin was necessary for the recruitment of afadin and the tyrosine phosphatase SHP-2 to the leading edge. SHP-2 was previously reported to tightly regulate the activation of PDGF receptor and its downstream signaling pathway for the formation of the leading edge. These results indicate that afadin has a novel role in PDGF-induced directional cell movement, presumably in cooperation with active Rap1 and SHP-2.
Collapse
Affiliation(s)
- Muneaki Miyata
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Hisakazu Ogita
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Hitomi Komura
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Shinsuke Nakata
- Department of Molecular Biology and Biochemistry, Osaka University Graduate School of Medicine, Osaka 565-0871 Japan
| | - Ryoko Okamoto
- Department of Molecular Biology and Biochemistry, Osaka University Graduate School of Medicine, Osaka 565-0871 Japan
| | - Misa Ozaki
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Takashi Majima
- Department of Molecular Biology and Biochemistry, Osaka University Graduate School of Medicine, Osaka 565-0871 Japan
| | - Naomi Matsuzawa
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Satoshi Kawano
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Akihiro Minami
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Masumi Waseda
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Naoyuki Fujita
- Department of Molecular Biology and Biochemistry, Osaka University Graduate School of Medicine, Osaka 565-0871 Japan
| | - Kiyohito Mizutani
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Yoshiyuki Rikitake
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Yoshimi Takai
- Division of Molecular and Cellular Biology, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| |
Collapse
|
195
|
Fetting JL, Spencer SA, Wolff T. The cell adhesion molecules Echinoid and Friend of Echinoid coordinate cell adhesion and cell signaling to regulate the fidelity of ommatidial rotation in the Drosophila eye. Development 2009; 136:3323-33. [PMID: 19736327 DOI: 10.1242/dev.038422] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Directed cellular movements are a universal feature of morphogenesis in multicellular organisms. Differential adhesion between the stationary and motile cells promotes these cellular movements to effect spatial patterning of cells. A prominent feature of Drosophila eye development is the 90 degrees rotational movement of the multicellular ommatidial precursors within a matrix of stationary cells. We demonstrate that the cell adhesion molecules Echinoid (Ed) and Friend of Echinoid (Fred) act throughout ommatidial rotation to modulate the degree of ommatidial precursor movement. We propose that differential levels of Ed and Fred between stationary and rotating cells at the initiation of rotation create a permissive environment for cell movement, and that uniform levels in these two populations later contribute to stopping the movement. Based on genetic data, we propose that ed and fred impart a second, independent, ;brake-like' contribution to this process via Egfr signaling. Ed and Fred are localized in largely distinct and dynamic patterns throughout rotation. However, ed and fred are required in only a subset of cells - photoreceptors R1, R7 and R6 - for normal rotation, cells that have only recently been linked to a role in planar cell polarity (PCP). This work also provides the first demonstration of a requirement for cone cells in the ommatidial rotation aspect of PCP. ed and fred also genetically interact with the PCP genes, but affect only the degree-of-rotation aspect of the PCP phenotype. Significantly, we demonstrate that at least one PCP protein, Stbm, is required in R7 to control the degree of ommatidial rotation.
Collapse
Affiliation(s)
- Jennifer L Fetting
- Department of Genetics, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | |
Collapse
|
196
|
Boettner B, Van Aelst L. Control of cell adhesion dynamics by Rap1 signaling. Curr Opin Cell Biol 2009; 21:684-93. [PMID: 19615876 PMCID: PMC2841981 DOI: 10.1016/j.ceb.2009.06.004] [Citation(s) in RCA: 200] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2009] [Revised: 05/16/2009] [Accepted: 06/17/2009] [Indexed: 10/20/2022]
Abstract
Individual cells in their particular environments adhere to the extracellular matrix (ECM) and their neighbours via integrin-containing and cadherin-containing complexes, respectively. The dynamics of these interactions regulate the formation and maintenance of complex tissues. An expanding body of evidence accentuates the role of the small Rap1 GTPase and its associated signaling network in many of these processes. In this review we will discuss more recently revealed roles of Rap1 signaling by primarily focusing on functions of the Rap1 effectors RIAM, KRIT-1/CCM1 and AF-6/Afadin in junctional regulation of the vascular system and in epithelial cells. Furthermore, we will describe novel findings on the Rap activator PDZ-GEF in the regulation of cell-cell adhesion between epithelial cells and within a stem cell niche.
Collapse
Affiliation(s)
- Benjamin Boettner
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA.
| | | |
Collapse
|
197
|
Franke WW. Discovering the molecular components of intercellular junctions--a historical view. Cold Spring Harb Perspect Biol 2009; 1:a003061. [PMID: 20066111 PMCID: PMC2773636 DOI: 10.1101/cshperspect.a003061] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The organization of metazoa is based on the formation of tissues and on tissue-typical functions and these in turn are based on cell-cell connecting structures. In vertebrates, four major forms of cell junctions have been classified and the molecular composition of which has been elucidated in the past three decades: Desmosomes, which connect epithelial and some other cell types, and the almost ubiquitous adherens junctions are based on closely cis-packed glycoproteins, cadherins, which are associated head-to-head with those of the hemi-junction domain of an adjacent cell, whereas their cytoplasmic regions assemble sizable plaques of special proteins anchoring cytoskeletal filaments. In contrast, the tight junctions (TJs) and gap junctions (GJs) are formed by tetraspan proteins (claudins and occludins, or connexins) arranged head-to-head as TJ seal bands or as paracrystalline connexin channels, allowing intercellular exchange of small molecules. The by and large parallel discoveries of the junction protein families are reported.
Collapse
Affiliation(s)
- Werner W Franke
- Helmholtz Group for Cell Biology, German Cancer Research Center, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany.
| |
Collapse
|
198
|
Meng W, Takeichi M. Adherens junction: molecular architecture and regulation. Cold Spring Harb Perspect Biol 2009; 1:a002899. [PMID: 20457565 DOI: 10.1101/cshperspect.a002899] [Citation(s) in RCA: 406] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The adherens junction (AJ) is an element of the cell-cell junction in which cadherin receptors bridge the neighboring plasma membranes via their homophilic interactions. Cadherins associate with cytoplasmic proteins, called catenins, which in turn bind to cytoskeletal components, such as actin filaments and microtubules. These molecular complexes further interact with other proteins, including signaling molecules, rendering the AJs into highly dynamic and regulatable structures. The AJs of such nature contribute to the physical linking of cells, as well as to the regulation of cell-cell contacts, which is essential for morphogenesis and remodeling of tissues and organs. Thus, elucidating the molecular architecture of the AJs and their regulatory mechanisms are crucial for understanding how the multicellular system is organized.
Collapse
Affiliation(s)
- Wenxiang Meng
- RIKEN Center for Developmental Biology, Chuo-ku, Kobe 650-0047, Japan
| | | |
Collapse
|
199
|
Fanning AS, Anderson JM. Zonula occludens-1 and -2 are cytosolic scaffolds that regulate the assembly of cellular junctions. Ann N Y Acad Sci 2009; 1165:113-20. [PMID: 19538295 DOI: 10.1111/j.1749-6632.2009.04440.x] [Citation(s) in RCA: 296] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The integrity of the tight junction barrier in epithelial and endothelial cells is critical to human health, but we still lack a detailed mechanistic knowledge of how the barrier is formed during development or responds to pathological and pharmacological insults. This limits our understanding of barrier dysfunction in disease and slows the development of therapeutic strategies. Recent studies confirm the long-maintained but previously unsupported view that the zonula occludens (ZO) proteins ZO-1 and ZO-2 are critical determinants of barrier formation. However, ZO proteins can also be components of adherens junctions, and recent studies suggest that ZO proteins may also promote the assembly and function of these junctions during epithelial morphogenesis. We review these studies and outline several recent observations that suggest that one role of ZO proteins is to regulate cytoskeletal dynamics at cell junctions. Finally, we propose a model by which the functional activities of ZO proteins in the adherens junction and tight junction are differentiated by a novel regulatory motif known as the U6 or acidic motif.
Collapse
Affiliation(s)
- Alan S Fanning
- The Department of Cell and Molecular Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7545, USA.
| | | |
Collapse
|
200
|
Sawyer JK, Harris NJ, Slep KC, Gaul U, Peifer M. The Drosophila afadin homologue Canoe regulates linkage of the actin cytoskeleton to adherens junctions during apical constriction. ACTA ACUST UNITED AC 2009; 186:57-73. [PMID: 19596848 PMCID: PMC2712996 DOI: 10.1083/jcb.200904001] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cadherin-based adherens junctions (AJs) mediate cell adhesion and regulate cell shape change. The nectin–afadin complex also localizes to AJs and links to the cytoskeleton. Mammalian afadin has been suggested to be essential for adhesion and polarity establishment, but its mechanism of action is unclear. In contrast, Drosophila melanogaster’s afadin homologue Canoe (Cno) has suggested roles in signal transduction during morphogenesis. We completely removed Cno from embryos, testing these hypotheses. Surprisingly, Cno is not essential for AJ assembly or for AJ maintenance in many tissues. However, morphogenesis is impaired from the start. Apical constriction of mesodermal cells initiates but is not completed. The actomyosin cytoskeleton disconnects from AJs, uncoupling actomyosin constriction and cell shape change. Cno has multiple direct interactions with AJ proteins, but is not a core part of the cadherin–catenin complex. Instead, Cno localizes to AJs by a Rap1- and actin-dependent mechanism. These data suggest that Cno regulates linkage between AJs and the actin cytoskeleton during morphogenesis.
Collapse
Affiliation(s)
- Jessica K Sawyer
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|