1
|
de Caestecker C, Macara IG. A size filter at the Golgi regulates apical membrane protein sorting. Nat Cell Biol 2024; 26:1678-1690. [PMID: 39237743 DOI: 10.1038/s41556-024-01500-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 08/12/2024] [Indexed: 09/07/2024]
Abstract
Despite decades of research, apical sorting of epithelial membrane proteins remains incompletely understood. We noted that apical cytoplasmic domains are smaller than those of basolateral proteins; however, the reason for this discrepancy is unknown. Here we used a synthetic biology approach to investigate whether a size barrier at the Golgi apparatus might hinder apical sorting of proteins with large cytoplasmic tails. We focused on Crb3, Ace2 and Muc1 as representative apical proteins with short cytoplasmic tails. By incorporating a streptavidin-binding peptide, these proteins can be trapped in the endoplasmic reticulum until addition of biotin, which triggers synchronous release to the Golgi and subsequent transport to the apical cortex. Strikingly, increasing the size of their cytoplasmic domains caused partial mislocalization to the basolateral cortex and significantly delayed Golgi departure. Moreover, N-glycosylation of 'large' Crb3 was delayed, and 'small' Crb3 segregated into spatially distinct Golgi regions. Biologically, Crb3 forms a complex through its cytoplasmic tail with the Pals1 protein, which could also delay departure, but although associated at the endoplasmic reticulum and Golgi, Pals1 disassociated before Crb3 departure. Notably, a non-dissociable mutant Pals1 hampered the exit of Crb3. We conclude that, unexpectedly, a size filter at the Golgi facilitates apical sorting of proteins with small cytoplasmic domains and that timely release of Pals1, to reduce cytoplasmic domain size, is essential for normal Crb3 sorting.
Collapse
Affiliation(s)
- Christian de Caestecker
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ian G Macara
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
2
|
Nzigou Mombo B, Bijonowski BM, Raab CA, Niland S, Brockhaus K, Müller M, Eble JA, Wegner SV. Reversible photoregulation of cell-cell adhesions with opto-E-cadherin. Nat Commun 2023; 14:6292. [PMID: 37813868 PMCID: PMC10562482 DOI: 10.1038/s41467-023-41932-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 09/22/2023] [Indexed: 10/11/2023] Open
Abstract
E-cadherin-based cell-cell adhesions are dynamically and locally regulated in many essential processes, including embryogenesis, wound healing and tissue organization, with dysregulation manifesting as tumorigenesis and metastasis. However, the lack of tools that would provide control of the high spatiotemporal precision observed with E-cadherin adhesions hampers investigation of the underlying mechanisms. Here, we present an optogenetic tool, opto-E-cadherin, that allows reversible control of E-cadherin-mediated cell-cell adhesions with blue light. With opto-E-cadherin, functionally essential calcium binding is photoregulated such that cells expressing opto-E-cadherin at their surface adhere to each other in the dark but not upon illumination. Consequently, opto-E-cadherin provides remote control over multicellular aggregation, E-cadherin-associated intracellular signalling and F-actin organization in 2D and 3D cell cultures. Opto-E-cadherin also allows switching of multicellular behaviour between single and collective cell migration, as well as of cell invasiveness in vitro and in vivo. Overall, opto-E-cadherin is a powerful optogenetic tool capable of controlling cell-cell adhesions at the molecular, cellular and behavioural level that opens up perspectives for the study of dynamics and spatiotemporal control of E-cadherin in biological processes.
Collapse
Affiliation(s)
- Brice Nzigou Mombo
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstraße 15, 48149, Münster, Germany
| | - Brent M Bijonowski
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstraße 15, 48149, Münster, Germany
| | - Christopher A Raab
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstraße 15, 48149, Münster, Germany
| | - Stephan Niland
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstraße 15, 48149, Münster, Germany
| | - Katrin Brockhaus
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstraße 15, 48149, Münster, Germany
| | - Marc Müller
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstraße 15, 48149, Münster, Germany
| | - Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstraße 15, 48149, Münster, Germany
| | - Seraphine V Wegner
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstraße 15, 48149, Münster, Germany.
| |
Collapse
|
3
|
Levic DS, Bagnat M. Polarized transport of membrane and secreted proteins during lumen morphogenesis. Semin Cell Dev Biol 2023; 133:65-73. [PMID: 35307284 PMCID: PMC9481742 DOI: 10.1016/j.semcdb.2022.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/09/2022] [Accepted: 03/12/2022] [Indexed: 10/18/2022]
Abstract
A ubiquitous feature of animal development is the formation of fluid-filled cavities or lumina, which transport gases and fluids across tissues and organs. Among different species, lumina vary drastically in size, scale, and complexity. However, all lumen formation processes share key morphogenetic principles that underly their development. Fundamentally, a lumen simply consists of epithelial cells that encapsulate a continuous internal space, and a common way of building a lumen is via opening and enlarging by filling it with fluid and/or macromolecules. Here, we discuss how polarized targeting of membrane and secreted proteins regulates lumen formation, mainly focusing on ion transporters in vertebrate model systems. We also discuss mechanistic differences observed among invertebrates and vertebrates and describe how the unique properties of the Na+/K+-ATPase and junctional proteins can promote polarization of immature epithelia to build lumina de novo in developing organs.
Collapse
Affiliation(s)
- Daniel S Levic
- Department of Cell Biology, Duke University, Durham, NC 27710, USA.
| | - Michel Bagnat
- Department of Cell Biology, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
4
|
E-Cadherin Regulates Mitochondrial Membrane Potential in Cancer Cells. Cancers (Basel) 2021; 13:cancers13205054. [PMID: 34680202 PMCID: PMC8534231 DOI: 10.3390/cancers13205054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/19/2022] Open
Abstract
Epithelial cancer cells often have unusually higher mitochondrial membrane potential (ΔΨm) than their normal counterparts, which has been associated with increased invasiveness in vitro and higher metastatic potential in vivo. However, the mechanisms by which ΔΨm in cancer cells is regulated in tumor microenvironment (TME) remain unclear. In this study, we used an in vitro micropatterning platform to recapitulate biophysical confinement cues in the TME and investigated the mechanisms by which these regulate cancer cell ΔΨm. We found that micropatterning resulted in a spatial distribution of ΔΨm, which correlated with the level of E-cadherin mediated intercellular adhesion. There was a stark contrast in the spatial distribution of ΔΨm in the micropattern of E-cadherin-negative breast cancer cells (MDA-MB-231) compared to that of the high E-cadherin expressing (MCF-7) cancer cells. Disruption and knockout of E-cadherin adhesions rescued the low ΔΨm found at the center of MCF-7 micropatterns with high E-cadherin expression, while E-cadherin overexpression in MDA-MB-231 and MCF-7 cells lowered their ΔΨm at the micropattern center. These results show that E-cadherin plays an important role in regulating the ΔΨm of cancer cells in the context of biophysical cues in TME.
Collapse
|
5
|
Hara T, Saeki K, Jinnouchi H, Kazuno S, Miura Y, Yokomizo T. The c-terminal region of BLT2 restricts its localization to the lateral membrane in a LIN7C-dependent manner. FASEB J 2021; 35:e21364. [PMID: 33481310 DOI: 10.1096/fj.202002640r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/26/2020] [Accepted: 12/28/2020] [Indexed: 11/11/2022]
Abstract
Leukotriene B4 receptor type 2 (BLT2) is a G protein-coupled receptor (GPCR) mainly expressed in epithelial cells, where it enhances barrier function. A unique characteristic of BLT2 is its restricted localization to the lateral membrane. However, the molecular mechanism underlying the localization of BLT2 to the lateral membrane and the physiological roles of laterally localized BLT2 are unknown. BLT1 is the most homologous GPCR to BLT2 and localizes to both the apical and lateral membranes. In this study, we generated chimeric receptors of BLT2 and BLT1 as well as deletion mutants of BLT2 to determine the region(s) of BLT2 responsible for its localization. Chimeric receptors containing the C-terminal domain of BLT2 localized only to the lateral membrane, and the C-terminal deletion mutant of BLT2 accumulated at the Golgi apparatus. Furthermore, the middle and C-terminal regions of BLT2 were important for maintaining epithelial barrier function. Proteomics analysis using the chimeric BLT-ascorbate peroxidase 2 biotinylation method showed that some proteins involved in intracellular protein transport, cell-cell junctions, and actin filament binding were located very close to the C-terminal domain of BLT2. Knockdown of lin-7 homolog C (LIN7C), a membrane trafficking protein, led to accumulation of BLT2 in the Golgi apparatus, resulting in diminished epithelial barrier function. These results suggest that the C-terminal region of BLT2 plays an important role in the transport of BLT2 from the Golgi apparatus to the plasma membrane in a LIN7C-dependent manner.
Collapse
Affiliation(s)
- Takuya Hara
- Department of Biochemistry, Juntendo University School of Medicine, Tokyo, Japan.,Fuji Research Laboratories, Kowa Co., Ltd, Shizuoka, Japan
| | - Kazuko Saeki
- Department of Biochemistry, Juntendo University School of Medicine, Tokyo, Japan
| | - Hiromi Jinnouchi
- Department of Biochemistry, Juntendo University School of Medicine, Tokyo, Japan
| | - Saiko Kazuno
- Laboratory of Proteomics and Biomolecular Science, Research Support Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshiki Miura
- Laboratory of Proteomics and Biomolecular Science, Research Support Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takehiko Yokomizo
- Department of Biochemistry, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
6
|
Kunii M, Noguchi Y, Yoshimura SI, Kanda S, Iwano T, Avriyanti E, Atik N, Sato T, Sato K, Ogawa M, Harada A. SNAP23 deficiency causes severe brain dysplasia through the loss of radial glial cell polarity. J Cell Biol 2021; 220:e201910080. [PMID: 33332551 PMCID: PMC7754684 DOI: 10.1083/jcb.201910080] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 08/23/2020] [Accepted: 10/23/2020] [Indexed: 02/06/2023] Open
Abstract
In the developing brain, the polarity of neural progenitor cells, termed radial glial cells (RGCs), is important for neurogenesis. Intercellular adhesions, termed apical junctional complexes (AJCs), at the apical surface between RGCs are necessary for cell polarization. However, the mechanism by which AJCs are established remains unclear. Here, we show that a SNARE complex composed of SNAP23, VAMP8, and Syntaxin1B has crucial roles in AJC formation and RGC polarization. Central nervous system (CNS)-specific ablation of SNAP23 (NcKO) results in mice with severe hypoplasia of the neocortex and no hippocampus or cerebellum. In the developing NcKO brain, RGCs lose their polarity following the disruption of AJCs and exhibit reduced proliferation, increased differentiation, and increased apoptosis. SNAP23 and its partner SNAREs, VAMP8 and Syntaxin1B, are important for the localization of an AJC protein, N-cadherin, to the apical plasma membrane of RGCs. Altogether, SNARE-mediated localization of N-cadherin is essential for AJC formation and RGC polarization during brain development.
Collapse
Affiliation(s)
- Masataka Kunii
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Laboratory of Molecular Traffic, Department of Molecular and Cellular Biology, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | - Yuria Noguchi
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shin-ichiro Yoshimura
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Satoshi Kanda
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tomohiko Iwano
- Department of Anatomy and Cell Biology, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Erda Avriyanti
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Dermatology and Venereology, Faculty of Medicine, Padjadjaran University, Bandung, Indonesia
| | - Nur Atik
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Biomedical Sciences, Faculty of Medicine, Padjadjaran University, Bandung, Indonesia
| | - Takashi Sato
- Laboratory of Developmental Biology and Metabolism, Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | - Ken Sato
- Laboratory of Molecular Traffic, Department of Molecular and Cellular Biology, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | | | - Akihiro Harada
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Laboratory of Molecular Traffic, Department of Molecular and Cellular Biology, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| |
Collapse
|
7
|
Karwelat D, Schmeck B, Ringel M, Benedikter BJ, Hübner K, Beinborn I, Maisner A, Schulte LN, Vollmeister E. Influenza virus-mediated suppression of bronchial Chitinase-3-like 1 secretion promotes secondary pneumococcal infection. FASEB J 2020; 34:16432-16448. [PMID: 33095949 DOI: 10.1096/fj.201902988rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 09/29/2020] [Accepted: 10/02/2020] [Indexed: 12/19/2022]
Abstract
Infections of the lung are among the leading causes of death worldwide. Despite the preactivation of innate defense programs during viral infection, secondary bacterial infection substantially elevates morbidity and mortality rates. Particularly problematic are co-infections with influenza A virus (IAV) and the major bacterial pathogen Streptococcus pneumoniae. However, the molecular processes underlying the severe course of such co-infections are not fully understood. Previously, the absence of secreted glycoprotein Chitinase-3-like 1 (CHI3L1) was shown to increase pneumococcal replication in mice. We therefore hypothesized that an IAV preinfection decreases CHI3L1 levels to promote pneumococcal infection. Indeed, in an air-liquid interface model of primary human bronchial epithelial cells (hBECs), IAV preinfection interfered with apical but not basolateral CHI3L1 release. Confocal time-lapse microscopy revealed that the gradual loss of apical CHI3L1 localization during co-infection with influenza and S. pneumoniae coincided with the disappearance of goblet as well as ciliated cells and increased S. pneumoniae replication. Importantly, extracellular restoration of CHI3L1 levels using recombinant protein significantly reduced bacterial load in influenza preinfected bronchial models. Thus, recombinant CHI3L1 may provide a novel therapeutic means to lower morbidity and mortality associated with post-influenza pneumococcal infections.
Collapse
Affiliation(s)
- Diana Karwelat
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Marburg, Philipps University Marburg, Hesse, Germany
| | - Bernd Schmeck
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Marburg, Philipps University Marburg, Hesse, Germany.,Department of Pulmonary and Critical Care Medicine, University Medical Center Marburg, Universities of Giessen and Marburg Lung Center, Philipps University Marburg, Hesse, Germany.,German Center for Lung Research (DZL), Marburg, Hesse, Germany.,Center for Synthetic Microbiology (SYNMIKRO), Philipps University Marburg, Marburg, Hesse, Germany
| | - Marc Ringel
- Institute of Virology, Philipps University Marburg, Marburg, Hesse, Germany
| | - Birke J Benedikter
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Marburg, Philipps University Marburg, Hesse, Germany
| | - Kathleen Hübner
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Marburg, Philipps University Marburg, Hesse, Germany
| | - Isabell Beinborn
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Marburg, Philipps University Marburg, Hesse, Germany
| | - Andrea Maisner
- Institute of Virology, Philipps University Marburg, Marburg, Hesse, Germany
| | - Leon N Schulte
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Marburg, Philipps University Marburg, Hesse, Germany.,German Center for Lung Research (DZL), Marburg, Hesse, Germany
| | - Evelyn Vollmeister
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Marburg, Philipps University Marburg, Hesse, Germany
| |
Collapse
|
8
|
Kell MJ, Ang SF, Pigati L, Halpern A, Fölsch H. Novel function for AP-1B during cell migration. Mol Biol Cell 2020; 31:2475-2493. [PMID: 32816642 PMCID: PMC7851849 DOI: 10.1091/mbc.e20-04-0256] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The epithelial cell-specific clathrin adaptor protein (AP)-1B has a well-established role in polarized sorting of cargos to the basolateral membrane. Here we show that β1 integrin was dependent on AP-1B and its coadaptor, autosomal recessive hypercholesterolemia protein (ARH), for sorting to the basolateral membrane. We further demonstrate an unprecedented role for AP-1B at the basal plasma membrane during collective cell migration of epithelial sheets. During wound healing, expression of AP-1B (and ARH in AP–1B-positive cells) slowed epithelial-cell migration. We show that AP-1B colocalized with β1 integrin in focal adhesions during cell migration using confocal microscopy and total internal reflection fluorescence microscopy on fixed specimens. Further, AP-1B labeling in cell protrusions was distinct from labeling for the endocytic adaptor complex AP-2. Using stochastic optical reconstruction microscopy we identified numerous AP–1B-coated structures at or close to the basal plasma membrane in cell protrusions. In addition, immunoelectron microscopy showed AP-1B in coated pits and vesicles at the plasma membrane during cell migration. Lastly, quantitative real-time reverse transcription PCR analysis of human epithelial-derived cell lines revealed a loss of AP-1B expression in highly migratory metastatic cancer cells suggesting that AP-1B’s novel role at the basal plasma membrane during cell migration might be an anticancer mechanism.
Collapse
Affiliation(s)
- Margaret Johnson Kell
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Su Fen Ang
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Lucy Pigati
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Abby Halpern
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Heike Fölsch
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|
9
|
Munthe E, Raiborg C, Stenmark H, Wenzel EM. Clathrin regulates Wnt/β-catenin signaling by affecting Golgi to plasma membrane transport of transmembrane proteins. J Cell Sci 2020; 133:jcs244467. [PMID: 32546530 DOI: 10.1242/jcs.244467] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 06/04/2020] [Indexed: 12/19/2022] Open
Abstract
The canonical Wnt/β-catenin signaling pathway regulates cell proliferation in development and adult tissue homeostasis. Dysregulated signaling contributes to human diseases, in particular cancer. Growing evidence suggests a role for clathrin and/or endocytosis in the regulation of this pathway, but conflicting results exist and demand a deeper mechanistic understanding. We investigated the consequences of clathrin depletion on Wnt/β-catenin signaling in cell lines and found a pronounced reduction in β-catenin protein levels, which affects the amount of nuclear β-catenin and β-catenin target gene expression. Although we found no evidence that clathrin affects β-catenin levels via endocytosis or multivesicular endosome formation, an inhibition of protein transport through the biosynthetic pathway led to reduced levels of a Wnt co-receptor, low-density lipoprotein receptor-related protein 6 (LRP6), and cell adhesion molecules of the cadherin family, thereby affecting steady-state levels of β-catenin. We conclude that clathrin impacts on Wnt/β-catenin signaling by controlling exocytosis of transmembrane proteins, including cadherins and Wnt co-receptors that together control the membrane-bound and soluble pools of β-catenin.
Collapse
Affiliation(s)
- Else Munthe
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, N-0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316 Oslo, Norway
| | - Camilla Raiborg
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, N-0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316 Oslo, Norway
| | - Harald Stenmark
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, N-0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316 Oslo, Norway
| | - Eva Maria Wenzel
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, N-0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, N-0316 Oslo, Norway
| |
Collapse
|
10
|
Damiano V, Spessotto P, Vanin G, Perin T, Maestro R, Santarosa M. The Autophagy Machinery Contributes to E-cadherin Turnover in Breast Cancer. Front Cell Dev Biol 2020; 8:545. [PMID: 32714931 PMCID: PMC7344152 DOI: 10.3389/fcell.2020.00545] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/09/2020] [Indexed: 12/19/2022] Open
Abstract
Autophagy is an intracellular catabolic process that is increasingly being recognized as a crucial factor in several human diseases including cancers. Mounting evidence suggests that autophagy allows tumor cells to overcome otherwise fatal stresses and to increase dissemination. Nevertheless, how autophagy controls these processes and in particular how it impinges on cell-cell adhesion is still poorly understood. Here, we investigate the role of autophagy in the turnover of the epithelial adhesion molecule E-cadherin in the context of breast cancer. We demonstrated in breast cancer cell lines that autophagy impinges on E-cadherin expression and in the configuration of adherens junctions. Besides, we showed that E-cadherin colocalizes with LC3B and SQSTM1/p62, two components of the autophagosome machinery. Pull down and immunoprecipitation analyses provided evidence that E-cadherin and SQSTM1/p62 physically interact. Moreover, the physical closeness of E-cadherin and SQSTM1/p62 was demonstrated by proximity ligation assays in breast cancer cell lines and primary tumors. Finally, we proved that the silencing of SQSTM1/p62 diminished the E-cadherin/LC3B colocalization, further supporting the role of SQSTM1/p62 in E-cadherin delivery to autophagosomes. These findings suggest that the activation of autophagy, reported in breast cancers with poor prognosis and in dormant breast cancer cells, may contribute to the control of tumor progression via downmodulation of E-cadherin protein levels.
Collapse
Affiliation(s)
- Valentina Damiano
- Unit of Oncogenetics and Functional Oncogenomics, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Paola Spessotto
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Giulia Vanin
- Unit of Oncogenetics and Functional Oncogenomics, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Tiziana Perin
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Roberta Maestro
- Unit of Oncogenetics and Functional Oncogenomics, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Manuela Santarosa
- Unit of Oncogenetics and Functional Oncogenomics, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| |
Collapse
|
11
|
Bartle EI, Rao TC, Beggs RR, Dean WF, Urner TM, Kowalczyk AP, Mattheyses AL. Protein exchange is reduced in calcium-independent epithelial junctions. J Cell Biol 2020; 219:151763. [PMID: 32399559 PMCID: PMC7265307 DOI: 10.1083/jcb.201906153] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 01/29/2020] [Accepted: 03/11/2020] [Indexed: 12/29/2022] Open
Abstract
Desmosomes are cell–cell junctions that provide mechanical integrity to epithelial and cardiac tissues. Desmosomes have two distinct adhesive states, calcium-dependent and hyperadhesive, which balance tissue plasticity and strength. A highly ordered array of cadherins in the adhesive interface is hypothesized to drive hyperadhesion, but how desmosome structure confers adhesive state is still elusive. We employed fluorescence polarization microscopy to show that cadherin order is not required for hyperadhesion induced by pharmacologic and genetic approaches. FRAP experiments in cells treated with the PKCα inhibitor Gö6976 revealed that cadherins, plakoglobin, and desmoplakin have significantly reduced exchange in and out of hyperadhesive desmosomes. To test whether this was a result of enhanced keratin association, we used the desmoplakin mutant S2849G, which conferred reduced protein exchange. We propose that inside-out regulation of protein exchange modulates adhesive function, whereby proteins are “locked in” to hyperadhesive desmosomes while protein exchange confers plasticity on calcium-dependent desmosomes, thereby providing rapid control of adhesion.
Collapse
Affiliation(s)
- Emily I Bartle
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Tejeshwar C Rao
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Reena R Beggs
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - William F Dean
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Tara M Urner
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Andrew P Kowalczyk
- Departments of Cell Biology and Dermatology, Emory University, Atlanta, GA
| | - Alexa L Mattheyses
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
12
|
Lakshmi Narayan PK, Kajon AE. Human mastadenovirus-B (HAdV-B)-specific E3-CR1β and E3-CR1γ glycoproteins interact with each other and localize at the plasma membrane of non-polarized airway epithelial cells. Virology 2020; 546:67-78. [PMID: 32452418 PMCID: PMC7158847 DOI: 10.1016/j.virol.2020.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/20/2020] [Accepted: 04/06/2020] [Indexed: 11/23/2022]
Abstract
The E3 region of all simian and human types classified within species Human mastadenovirus B (HAdV-B) encodes two unique highly conserved ORFs of unknown function designated E3-CR1β and E3-CR1γ. We generated a HAdV-3 mutant encoding small epitope tags at the N-termini of both E3-CR1β and E3-CR1γ (HAdV-3 N-tag wt) and a double knock out (HAdV-3 N-tag DKO) mutant virus that does not express either protein. Our studies show that HAdV-3 E3-CR1β and E3-CR1γ are type I transmembrane proteins that are produced predominantly at late times post infection, are glycosylated, co-localize at the plasma membrane of non-polarized epithelial cells, and interact with each other. At their extreme C-termini HAdV-B E3-CR1β and E3-CR1γ possess a conserved di-leucine motif followed by a class II PDZ domain binding motif (PBM). HAdV-3 E3-CR1β and E3-CR1γ are dispensable for virus growth, progeny release, spread, and plaque formation in A549 cells. HAdV-B E3-CR1β and E3-CR1γ are type I transmembrane proteins. HAdV-B E3-CR1β and E3-CR1γ possess a C-terminal class II PDZ binding motif. HAdV-3 E3-CR1β and E3-CR1γ interact and co-localize at the plasma membrane. HAdV-3 E3-CR1β and E3-CR1γ are dispensable for virus progeny release and spread.
Collapse
Affiliation(s)
| | - Adriana E Kajon
- Infectious Disease Program, Lovelace Respiratory Research Institute, Albuquerque, NM, USA.
| |
Collapse
|
13
|
Ollech D, Pflästerer T, Shellard A, Zambarda C, Spatz JP, Marcq P, Mayor R, Wombacher R, Cavalcanti-Adam EA. An optochemical tool for light-induced dissociation of adherens junctions to control mechanical coupling between cells. Nat Commun 2020; 11:472. [PMID: 31980653 PMCID: PMC6981158 DOI: 10.1038/s41467-020-14390-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 01/02/2020] [Indexed: 01/19/2023] Open
Abstract
The cadherin-catenin complex at adherens junctions (AJs) is essential for the formation of cell-cell adhesion and epithelium integrity; however, studying the dynamic regulation of AJs at high spatio-temporal resolution remains challenging. Here we present an optochemical tool which allows reconstitution of AJs by chemical dimerization of the force bearing structures and their precise light-induced dissociation. For the dimerization, we reconstitute acto-myosin connection of a tailless E-cadherin by two ways: direct recruitment of α-catenin, and linking its cytosolic tail to the transmembrane domain. Our approach enables a specific ON-OFF switch for mechanical coupling between cells that can be controlled spatially on subcellular or tissue scale via photocleavage. The combination with cell migration analysis and traction force microscopy shows a wide-range of applicability and confirms the mechanical contribution of the reconstituted AJs. Remarkably, in vivo our tool is able to control structural and functional integrity of the epidermal layer in developing Xenopus embryos.
Collapse
Affiliation(s)
- Dirk Ollech
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, D-69120, Heidelberg, Germany
- Department of Biophysical Chemistry, Institute of Physical Chemistry, Heidelberg University, INF 253, D-69120, Heidelberg, Germany
- Applied Physics Department, Science for Life Laboratory and KTH Royal Technical University, Tomtebodavägen 23A, S-17165, Stockholm, Sweden
| | - Tim Pflästerer
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, D-69120, Heidelberg, Germany
- Department of Biophysical Chemistry, Institute of Physical Chemistry, Heidelberg University, INF 253, D-69120, Heidelberg, Germany
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, INF 364, D-69120, Heidelberg, Germany
| | - Adam Shellard
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK
| | - Chiara Zambarda
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, D-69120, Heidelberg, Germany
- Department of Biophysical Chemistry, Institute of Physical Chemistry, Heidelberg University, INF 253, D-69120, Heidelberg, Germany
| | - Joachim Pius Spatz
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, D-69120, Heidelberg, Germany
- Department of Biophysical Chemistry, Institute of Physical Chemistry, Heidelberg University, INF 253, D-69120, Heidelberg, Germany
| | - Philippe Marcq
- PMMH, CNRS, ESPCI Paris, PSL University, Sorbonne Université, Université de Paris, F-75005, Paris, France
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK
| | - Richard Wombacher
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, INF 364, D-69120, Heidelberg, Germany.
| | - Elisabetta Ada Cavalcanti-Adam
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, D-69120, Heidelberg, Germany.
- Department of Biophysical Chemistry, Institute of Physical Chemistry, Heidelberg University, INF 253, D-69120, Heidelberg, Germany.
| |
Collapse
|
14
|
Kassouf T, Larive RM, Morel A, Urbach S, Bettache N, Marcial Medina MC, Mèrezègue F, Freiss G, Peter M, Boissière-Michot F, Solassol J, Montcourrier P, Coopman P. The Syk Kinase Promotes Mammary Epithelial Integrity and Inhibits Breast Cancer Invasion by Stabilizing the E-Cadherin/Catenin Complex. Cancers (Basel) 2019; 11:cancers11121974. [PMID: 31817924 PMCID: PMC6966528 DOI: 10.3390/cancers11121974] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 12/20/2022] Open
Abstract
While first discovered in immunoreceptor signaling, the Syk protein kinase behaves as a tumor and metastasis suppressor in epithelial cells. Its reduced expression in breast and other carcinomas is correlated with decreased survival and increased metastasis risk, but its action mechanism remains largely unknown. Using phosphoproteomics we found that Syk phosphorylated E-cadherin and α-, β-, and p120-catenins on multiple tyrosine residues that concentrate at intercellular junctions. Increased Syk expression and activation enhanced E-cadherin/catenin phosphorylation, promoting their association and complex stability. In human breast cancer cells, Syk stimulated intercellular aggregation, E-cadherin recruitment and retention at adherens junctions, and promoted epithelial integrity, whereas it inhibited cell migration and invasion. Opposite effects were obtained with Syk knockdown or non-phosphorylatable mutant E-cadherin expression. Mechanistically, Syk stimulated the interaction of the E-cadherin/catenin complex with zonula occludens proteins and the actin cytoskeleton. Conditional Syk knockout in the lactating mouse mammary gland perturbed alveologenesis and disrupted E-cadherin localization at adherens junctions, corroborating the observations in cells. Hence, Syk is involved in the maintenance of the epithelial integrity of the mammary gland via the phosphorylation and stabilization of the E-cadherin/catenin adherens junction complex, thereby inhibiting cell migration and malignant tumor invasion.
Collapse
Affiliation(s)
- Toufic Kassouf
- IRCM, Inserm, CNRS, Universit@#xE9; de Montpellier, ICM, 208 Rue des Apothicaires, 34298 Montpellier, France; (T.K.); (R.M.L.); (G.F.); (M.P.); (J.S.)
- CRBM, CNRS, Université de Montpellier, 1919 Route de Mende, 34293 Montpellier, France;
| | - Romain Maxime Larive
- IRCM, Inserm, CNRS, Universit@#xE9; de Montpellier, ICM, 208 Rue des Apothicaires, 34298 Montpellier, France; (T.K.); (R.M.L.); (G.F.); (M.P.); (J.S.)
- IBMM, Université de Montpellier, CNRS, ENSCM, 15 avenue Charles Flahault - BP 14491, 34093 Montpellier, France;
| | - Anne Morel
- CRBM, CNRS, Université de Montpellier, 1919 Route de Mende, 34293 Montpellier, France;
| | - Serge Urbach
- Functional Proteomics Platform, IGF, Université de Montpellier, CNRS, INSERM, 141 rue de la Cardonille, 34094 Montpellier, France;
| | - Nadir Bettache
- IBMM, Université de Montpellier, CNRS, ENSCM, 15 avenue Charles Flahault - BP 14491, 34093 Montpellier, France;
| | | | - Fabrice Mèrezègue
- BioMV Department, Université de Montpellier CC25000, Place Eugène Bataillon, 34095 Montpellier, France;
| | - Gilles Freiss
- IRCM, Inserm, CNRS, Universit@#xE9; de Montpellier, ICM, 208 Rue des Apothicaires, 34298 Montpellier, France; (T.K.); (R.M.L.); (G.F.); (M.P.); (J.S.)
| | - Marion Peter
- IRCM, Inserm, CNRS, Universit@#xE9; de Montpellier, ICM, 208 Rue des Apothicaires, 34298 Montpellier, France; (T.K.); (R.M.L.); (G.F.); (M.P.); (J.S.)
| | | | - Jérôme Solassol
- IRCM, Inserm, CNRS, Universit@#xE9; de Montpellier, ICM, 208 Rue des Apothicaires, 34298 Montpellier, France; (T.K.); (R.M.L.); (G.F.); (M.P.); (J.S.)
| | - Philippe Montcourrier
- IRCM, Inserm, CNRS, Universit@#xE9; de Montpellier, ICM, 208 Rue des Apothicaires, 34298 Montpellier, France; (T.K.); (R.M.L.); (G.F.); (M.P.); (J.S.)
| | - Peter Coopman
- IRCM, Inserm, CNRS, Universit@#xE9; de Montpellier, ICM, 208 Rue des Apothicaires, 34298 Montpellier, France; (T.K.); (R.M.L.); (G.F.); (M.P.); (J.S.)
- Correspondence: ; Tel.: +33-467-61-3191
| |
Collapse
|
15
|
Oraiopoulou ME, Tampakaki M, Tzamali E, Tamiolakis T, Makatounakis V, Vakis AF, Zacharakis G, Sakkalis V, Papamatheakis J. A 3D tumor spheroid model for the T98G Glioblastoma cell line phenotypic characterization. Tissue Cell 2019; 59:39-43. [PMID: 31383287 DOI: 10.1016/j.tice.2019.05.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 05/23/2019] [Accepted: 05/23/2019] [Indexed: 01/25/2023]
Abstract
Major Glioblastoma's hallmarks include proliferation, invasion and heterogeneity. Biological 3D tumor spheroid models can serve as intermediate systems between traditional 2D cell culture and complex in vivo models. Tumor spheroids have been shown to more accurately reproduce the spatial organization and microenvironmental factors of in vivo micro-tumors, such as relevant gradients of nutrients and other molecular agents, while they maintain cell-to-cell and cell-to-matrix interactions. In vitro 3D assays are useful to monitor these properties. Here, we test the suitability of the well-known T98 G Glioblastoma cell line in such a 3D assay. The doubling time and death rate parameters of T98 G are estimated, as well as their spheroidal growth-expansion curves with and without the presence of basement membrane substrate. The T98 G invasive profile is characterized by collective morphology and proliferation-associated invasion. We show that the T98 G secondary GB cell line exhibits both invasive and proliferative capabilities in 3D and thus, can serve as control cell line for the 3D in vitro study of primary GB cell cultures.
Collapse
Affiliation(s)
- Mariam-Eleni Oraiopoulou
- School of Medicine, University of Crete, Heraklion, Crete, Greece; Institute of Computer Science, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
| | - Maria Tampakaki
- School of Medicine, University of Crete, Heraklion, Crete, Greece; Institute of Computer Science, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
| | - Eleftheria Tzamali
- Institute of Computer Science, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
| | - Theodoros Tamiolakis
- School of Medicine, University of Crete, Heraklion, Crete, Greece; Institute of Computer Science, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
| | - Venediktos Makatounakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
| | - Antonios F Vakis
- School of Medicine, University of Crete, Heraklion, Crete, Greece; Neurosurgery Clinic, University General Hospital of Heraklion, Heraklion, Greece
| | - Giannis Zacharakis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
| | - Vangelis Sakkalis
- Institute of Computer Science, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
| | - Joseph Papamatheakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece; Department of Biology, University of Crete, Heraklion, Crete, Greece.
| |
Collapse
|
16
|
Destination and consequences of Panx1 and mutant expression in polarized MDCK cells. Exp Cell Res 2019; 381:235-247. [PMID: 31102595 DOI: 10.1016/j.yexcr.2019.05.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/09/2019] [Accepted: 05/11/2019] [Indexed: 12/12/2022]
Abstract
The channel-forming membrane glycoprotein pannexin 1 (Panx1) is best characterized as an ATP release channel. To investigate the trafficking and sorting of Panx1, we used polarized MDCK cells and non-polarized BICR-M1Rk cells to track the fate of GFP-tagged Panx1. In non-polarized cells, Panx1 was found throughout the plasma membrane, including the lamellipodia of the tumor cells and the cell surface-targeting domain was mapped to residues 307-379. Panx1 was preferentially enriched at the apical membrane domain of polarized MDCK cells grown as monolayer sheets or as spheroids. Residual Panx1 localized within basolateral membranes of polarized MDCK cells was independent of a putative dileucine sorting motif LL365/6 found within the C-terminal of Panx1. Unexpectedly, stable expression of a Panx1 mutant, where a putative tyrosine-based basolateral sorting motif (YxxØ) was mutated (Y308F), or a truncated Δ379 Panx1 mutant, caused MDCK cells to lose cell-cell contacts and their ability to polarize as they underwent a switch to a more fibroblast-like phenotype. We conclude that Panx1 is preferentially delivered to the apical domain of polarized epithelial cells, and Panx1 mutants drive phenotypic changes to MDCK cells preventing their polarization.
Collapse
|
17
|
Jacquemet G, Stubb A, Saup R, Miihkinen M, Kremneva E, Hamidi H, Ivaska J. Filopodome Mapping Identifies p130Cas as a Mechanosensitive Regulator of Filopodia Stability. Curr Biol 2019; 29:202-216.e7. [PMID: 30639111 PMCID: PMC6345628 DOI: 10.1016/j.cub.2018.11.053] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/25/2018] [Accepted: 11/20/2018] [Indexed: 01/09/2023]
Abstract
Filopodia are adhesive cellular protrusions specialized in the detection of extracellular matrix (ECM)-derived cues. Although ECM engagement at focal adhesions is known to trigger the recruitment of hundreds of proteins ("adhesome") to fine-tune cellular behavior, the components of the filopodia adhesions remain undefined. Here, we performed a structured-illumination-microscopy-based screen to map the localization of 80 target proteins, linked to cell adhesion and migration, within myosin-X-induced filopodia. We demonstrate preferential enrichment of several adhesion proteins to either filopodia tips, filopodia shafts, or shaft subdomains, suggesting divergent, spatially restricted functions for these proteins. Moreover, proteins with phosphoinositide (PI) binding sites are particularly enriched in filopodia. This, together with the strong localization of PI(3,4)P2 in filopodia tips, predicts critical roles for PIs in regulating filopodia ultra-structure and function. Our mapping further reveals that filopodia adhesions consist of a unique set of proteins, the filopodome, that are distinct from classical nascent adhesions, focal adhesions, and fibrillar adhesions. Using live imaging, we observe that filopodia adhesions can give rise to nascent adhesions, which, in turn, form focal adhesions. We demonstrate that p130Cas (BCAR1) is recruited to filopodia tips via its C-terminal Cas family homology domain (CCHD) and acts as a mechanosensitive regulator of filopodia stability. Finally, we demonstrate that our map based on myosin-X-induced filopodia can be translated to endogenous filopodia and fascin- and IRSp53-mediated filopodia.
Collapse
Affiliation(s)
- Guillaume Jacquemet
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.
| | - Aki Stubb
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Rafael Saup
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Mitro Miihkinen
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Elena Kremneva
- Institute of Biotechnology, University of Helsinki, PO Box 56, 00014 Helsinki, Finland
| | - Hellyeh Hamidi
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland; Department of Biochemistry, University of Turku, Turku, Finland.
| |
Collapse
|
18
|
Wieduwild R, Howarth M. Assembling and decorating hyaluronan hydrogels with twin protein superglues to mimic cell-cell interactions. Biomaterials 2018; 180:253-264. [DOI: 10.1016/j.biomaterials.2018.07.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/03/2018] [Accepted: 07/11/2018] [Indexed: 12/30/2022]
|
19
|
Sehgal P, Kong X, Wu J, Sunyer R, Trepat X, Leckband D. Epidermal growth factor receptor and integrins control force-dependent vinculin recruitment to E-cadherin junctions. J Cell Sci 2018; 131:jcs206656. [PMID: 29487179 PMCID: PMC5897709 DOI: 10.1242/jcs.206656] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 02/07/2018] [Indexed: 12/30/2022] Open
Abstract
This study reports novel findings that link E-cadherin (also known as CDH1)-mediated force-transduction signaling to vinculin targeting to intercellular junctions via epidermal growth factor receptor (EGFR) and integrins. These results build on previous findings that demonstrated that mechanically perturbed E-cadherin receptors activate phosphoinositide 3-kinase and downstream integrins in an EGFR-dependent manner. Results of this study show that this EGFR-mediated kinase cascade controls the force-dependent recruitment of vinculin to stressed E-cadherin complexes - a key early signature of cadherin-based mechanotransduction. Vinculin targeting requires its phosphorylation at tyrosine 822 by Abl family kinases (hereafter Abl), but the origin of force-dependent Abl activation had not been identified. We now present evidence that integrin activation, which is downstream of EGFR signaling, controls Abl activation, thus linking E-cadherin to Abl through a mechanosensitive signaling network. These findings place EGFR and integrins at the center of a positive-feedback loop, through which force-activated E-cadherin signals regulate vinculin recruitment to cadherin complexes in response to increased intercellular tension.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Poonam Sehgal
- Department of Biochemistry, University of Illinois, Urbana-Champaign, IL 61802, USA
| | - Xinyu Kong
- Department of Biochemistry, University of Illinois, Urbana-Champaign, IL 61802, USA
| | - Jun Wu
- Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana-Champaign, IL 61802, USA
| | - Raimon Sunyer
- Institute for Bioengineering of Catalonia, Barcelona, Spain 08028
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Barcelona, Spain 08028
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia, Barcelona, Spain 08028
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Barcelona, Spain 08028
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain 08028
| | - Deborah Leckband
- Department of Biochemistry, University of Illinois, Urbana-Champaign, IL 61802, USA
- Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana-Champaign, IL 61802, USA
- Department of Chemistry, University of Illinois, Urbana-Champaign, IL 61802, USA
| |
Collapse
|
20
|
Bridges RJ, Bradbury NA. Cystic Fibrosis, Cystic Fibrosis Transmembrane Conductance Regulator and Drugs: Insights from Cellular Trafficking. Handb Exp Pharmacol 2018; 245:385-425. [PMID: 29460152 DOI: 10.1007/164_2018_103] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The eukaryotic cell is organized into membrane-delineated compartments that are characterized by specific cadres of proteins sustaining biochemically distinct cellular processes. The appropriate subcellular localization of proteins is key to proper organelle function and provides a physiological context for cellular processes. Disruption of normal trafficking pathways for proteins is seen in several genetic diseases, where a protein's absence for a specific subcellular compartment leads to organelle disruption, and in the context of an individual, a disruption of normal physiology. Importantly, several drug therapies can also alter protein trafficking, causing unwanted side effects. Thus, a deeper understanding of trafficking pathways needs to be appreciated as novel therapeutic modalities are proposed. Despite the promising efficacy of novel therapeutic agents, the intracellular bioavailability of these compounds has proved to be a potential barrier, leading to failures in treatments for various diseases and disorders. While endocytosis of drug moieties provides an efficient means of getting material into cells, the subsequent release and endosomal escape of materials into the cytosol where they need to act has been a barrier. An understanding of cellular protein/lipid trafficking pathways has opened up strategies for increasing drug bioavailability. Approaches to enhance endosomal exit have greatly increased the cytosolic bioavailability of drugs and will provide a means of investigating previous drugs that may have been shelved due to their low cytosolic concentration.
Collapse
Affiliation(s)
- Robert J Bridges
- Department of Physiology and Biophysics, Chicago Medical School, North Chicago, IL, USA
| | - Neil A Bradbury
- Department of Physiology and Biophysics, Chicago Medical School, North Chicago, IL, USA.
| |
Collapse
|
21
|
Terciolo C, Dobric A, Ouaissi M, Siret C, Breuzard G, Silvy F, Marchiori B, Germain S, Bonier R, Hama A, Owens R, Lombardo D, Rigot V, André F. Saccharomyces boulardii CNCM I-745 Restores intestinal Barrier Integrity by Regulation of E-cadherin Recycling. J Crohns Colitis 2017; 11:999-1010. [PMID: 28333335 DOI: 10.1093/ecco-jcc/jjx030] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 02/21/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Alteration in intestinal permeability is the main factor underlying the pathogenesis of many diseases affecting the gut, such as inflammatory bowel disease [IBD]. Characterization of molecules targeting the restoration of intestinal barrier integrity is therefore vital for the development of alternative therapies. The yeast Saccharomyces boulardii CNCM I-745 [Sb], used to prevent and treat antibiotic-associated infectious and functional diarrhea, may have a beneficial effect in the treatment of IBD. METHODS We analyzed the impact of Sb supernatant on tissue integrity and components of adherens junctions using cultured explants of colon from both IBD and healthy patients. To evaluate the pathways by which Sb regulates the expression of E-cadherin at the cell surface, we developed in vitro assays using human colonic cell lines, including cell aggregation, a calcium switch assay, real-time measurement of transepithelial electrical resistance [TEER] and pulse-chase experiments. RESULTS We showed that Sb supernatant treatment of colonic explants protects the epithelial morphology and maintains E-cadherin expression at the cell surface. In vitro experiments revealed that Sb supernatant enhances E-cadherin delivery to the cell surface by re-routing endocytosed E-cadherin back to the plasma membrane. This process, involving Rab11A-dependent recycling endosome, leads to restoration of enterocyte adherens junctions, in addition to the overall restoration and strengthening of intestinal barrier function. CONCLUSION These findings open new possibilities of discovering novel options for prevention and therapy of diseases that affect intestinal permeability.
Collapse
Affiliation(s)
- Chloé Terciolo
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | - Aurélie Dobric
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | - Mehdi Ouaissi
- Aix-Marseille Université, Inserm, AP-HM, UMR 911, CRO2, Marseille, France
| | - Carole Siret
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | - Gilles Breuzard
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | - Françoise Silvy
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | | | | | - Renaté Bonier
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | - Adel Hama
- Ecole des Mines de Saint Etienne, BEL, Gardanne, France
| | - Roisin Owens
- Ecole des Mines de Saint Etienne, BEL, Gardanne, France
| | | | - Véronique Rigot
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| | - Frédéric André
- Aix-Marseille Université, Inserm, UMR 911, CRO2, Marseille, France
| |
Collapse
|
22
|
Abstract
A crucial step in the life cycle of Plasmodium parasites is the transition from the liver stage to the blood stage. Hepatocyte-derived merozoites reach the blood vessels of the liver inside host cell-derived vesicles called merosomes. The molecular basis of merosome formation is only partially understood. Here we show that Plasmodium berghei liver stage merozoites, upon rupture of the parasitophorous vacuole membrane, destabilize the host cell membrane (HCM) and induce separation of the host cell actin cytoskeleton from the HCM. At the same time, the phospholipid and protein composition of the HCM appears to be substantially altered. This includes the loss of a phosphatidylinositol 4,5-bisphosphate (PIP2) reporter and the PIP2-dependent actin-plasma membrane linker ezrin from the HCM. Furthermore, transmembrane domain-containing proteins and palmitoylated and myristoylated proteins, as well as glycosylphosphatidylinositol-anchored proteins, lose their HCM localization. Collectively, these findings provide an explanation of HCM destabilization during Plasmodium liver stage egress and thereby contribute to our understanding of the molecular mechanisms that lead to merosome formation. Egress from host cells is an essential process for intracellular pathogens, allowing successful infection of other cells and thereby spreading the infection. Here we describe the molecular details of a novel egress strategy of Plasmodium parasites infecting hepatocytes. We show that toward the end of the liver stage, parasites induce a breakdown of the host cell actin cytoskeleton, leading to destabilization of the host cell plasma membrane. This, in turn, results in the formation of membrane vesicles (merosomes), in which parasites can safely migrate from liver tissue to the bloodstream to infect red blood cells and start the pathogenic phase of malaria.
Collapse
|
23
|
Blepharocheilodontic syndrome is a CDH1 pathway-related disorder due to mutations in CDH1 and CTNND1. Genet Med 2017; 19:1013-1021. [PMID: 28301459 DOI: 10.1038/gim.2017.11] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 01/16/2017] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Blepharocheilodontic (BCD) syndrome is a rare autosomal dominant condition characterized by eyelid malformations, cleft lip/palate, and ectodermal dysplasia. The molecular basis of BCD syndrome remains unknown. METHODS We recruited 11 patients from 8 families and performed exome sequencing for 5 families with de novo BCD syndrome cases and targeted Sanger sequencing in the 3 remaining families. RESULTS We identified five CDH1 heterozygous missense mutations and three CTNND1 heterozygous truncating mutations leading to loss-of-function or haploinsufficiency. Establishment of detailed genotype-phenotype correlations was not possible because of the size of the cohort; however, the phenotype seems to appear more severe in case of CDH1 mutations. Functional analysis of CDH1 mutations confirmed their deleterious impact and suggested accelerated E-cadherin degradation. CONCLUSION Mutations in CDH1 encoding the E-cadherin were previously reported in hereditary diffuse gastric cancer as well as in nonsyndromic cleft lip/palate. Mutations in CTNND1 have never been reported before. The encoded protein, p120ctn, prevents E-cadherin endocytosis and stabilizes its localization at the cell surface. Conditional deletion of Cdh1 and Ctnnd1 in various animal models induces features reminiscent of BCD syndrome and underlines critical role of the E-cadherin-p120ctn interaction in eyelid, craniofacial, and tooth development. Our data assert BCD syndrome as a CDH1 pathway-related disorder due to mutations in CDH1 and CTNND1 and widen the phenotypic spectrum of E-cadherin anomalies.Genet Med advance online publication 09 March 2017.
Collapse
|
24
|
Brüser L, Bogdan S. Adherens Junctions on the Move-Membrane Trafficking of E-Cadherin. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a029140. [PMID: 28096264 DOI: 10.1101/cshperspect.a029140] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cadherin-based adherens junctions are conserved structures that mediate epithelial cell-cell adhesion in invertebrates and vertebrates. Despite their pivotal function in epithelial integrity, adherens junctions show a remarkable plasticity that is a prerequisite for tissue architecture and morphogenesis. Epithelial cadherin (E-cadherin) is continuously turned over and undergoes cycles of endocytosis, sorting and recycling back to the plasma membrane. Mammalian cell culture and genetically tractable model systems such as Drosophila have revealed conserved, but also distinct, mechanisms in the regulation of E-cadherin membrane trafficking. Here, we discuss our current knowledge about molecules and mechanisms controlling endocytosis, sorting and recycling of E-cadherin during junctional remodeling.
Collapse
Affiliation(s)
- Lena Brüser
- Institut für Neurobiologie, Universität Münster, Badestraße 9, 48149 Münster, Germany
| | - Sven Bogdan
- Institut für Neurobiologie, Universität Münster, Badestraße 9, 48149 Münster, Germany.,Institut für Physiologie und Pathophysiologie, Abteilung Molekulare Zellphysiologie, Phillips-Universität Marburg, Emil-Mannkopff-Straße 2, 35037 Marburg, Germany
| |
Collapse
|
25
|
Bryant DM, Yap AS. Editorial overview: Membrane traffic and cell polarity. Traffic 2016; 17:1231-1232. [DOI: 10.1111/tra.12433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 09/05/2016] [Indexed: 01/03/2023]
Affiliation(s)
- David M. Bryant
- Cancer Research UK; Beatson Institute; Glasgow UK
- Institute of Cancer Sciences; University of Glasgow; Glasgow UK
| | - Alpha S. Yap
- Division of Cell Biology and Molecular Medicine, Institute for Molecular Bioscience; The University of Queensland; Brisbane Australia
| |
Collapse
|
26
|
Cadwell CM, Su W, Kowalczyk AP. Cadherin tales: Regulation of cadherin function by endocytic membrane trafficking. Traffic 2016; 17:1262-1271. [PMID: 27624909 DOI: 10.1111/tra.12448] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/09/2016] [Accepted: 09/09/2016] [Indexed: 12/17/2022]
Abstract
Cadherins are the primary adhesion molecules in adherens junctions and desmosomes and play essential roles in embryonic development. Although significant progress has been made in understanding cadherin structure and function, we lack a clear vision of how cells confer plasticity upon adhesive junctions to allow for cellular rearrangements during development, wound healing and metastasis. Endocytic membrane trafficking has emerged as a fundamental mechanism by which cells confer a dynamic state to adhesive junctions. Recent studies indicate that the juxtamembrane domain of classical cadherins contains multiple endocytic motifs, or "switches," that can be used by cellular membrane trafficking machinery to regulate adhesion. The cadherin-binding protein p120-catenin (p120) appears to be the master regulator of access to these switches, thereby controlling cadherin endocytosis and turnover. This review focuses on p120 and other cadherin-binding proteins, ubiquitin ligases, and growth factors as key modulators of cadherin membrane trafficking.
Collapse
Affiliation(s)
- Chantel M Cadwell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Wenji Su
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia.,Biochemistry, Cell, and Developmental Biology Graduate Training Program, Emory University, Atlanta, Georgia
| | - Andrew P Kowalczyk
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia.,Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
27
|
Fiorino C, Harrison RE. E-cadherin is important for cell differentiation during osteoclastogenesis. Bone 2016; 86:106-18. [PMID: 26959175 DOI: 10.1016/j.bone.2016.03.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 01/29/2016] [Accepted: 03/04/2016] [Indexed: 01/05/2023]
Abstract
E-cadherin, a protein responsible for intercellular adhesion between epithelial cells, is also expressed in the monocyte/macrophage lineage. In this study we have explored the involvement of E-cadherin during receptor activator of nuclear factor-κB ligand (RANKL)-stimulated osteoclast differentiation. Osteoclastogenesis involves a period of precursor expansion followed by multiple fusion events to generate a multinuclear osteoclast that is capable of bone resorption. We asked whether E-cadherin participated in early precursor interactions and recognition or was a component of the osteoclast fusion machinery. Here, we show that endogenous E-cadherin expression is the highest during early stages of osteoclast differentiation, with surface expression visible on small precursor cells (fewer than four nuclei per cell) in both RAW 264.7 cells and primary macrophages. Blocking E-cadherin function with neutralizing antibodies prior to the onset of fusion delayed the expression of TRAP, Cathepsin K, DC-STAMP and NFATc1 and significantly diminished multinucleated osteoclast formation. Conversely, E-cadherin-GFP overexpressing macrophages displayed earlier NFATc1 nuclear translocation along with faster formation of multinucleated osteoclasts compared to control macrophages. Through live imaging we identified that disrupting E-cadherin function prolonged the proliferative phase of the precursor population while concomitantly decreasing the proportion of migrating precursors. The lamellipodium and polarized membrane extensions appeared to be the principal sites of fusion, indicating precursor migration was a critical factor contributing to osteoclast fusion. These findings demonstrate that E-cadherin-mediated cell-cell contacts can modulate osteoclast-specific gene expression and prompt differentiating osteoclast precursors toward migratory and fusion activities.
Collapse
Affiliation(s)
- Cara Fiorino
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario M1C 1A4, Canada; Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario M1C 1A4, Canada
| | - Rene E Harrison
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario M1C 1A4, Canada; Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario M1C 1A4, Canada.
| |
Collapse
|
28
|
Shashikanth N, Kisting MA, Leckband DE. Kinetic Measurements Reveal Enhanced Protein-Protein Interactions at Intercellular Junctions. Sci Rep 2016; 6:23623. [PMID: 27009566 PMCID: PMC4806357 DOI: 10.1038/srep23623] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/07/2016] [Indexed: 12/26/2022] Open
Abstract
The binding properties of adhesion proteins are typically quantified from measurements with soluble fragments, under conditions that differ radically from the confined microenvironment of membrane bound proteins in adhesion zones. Using classical cadherin as a model adhesion protein, we tested the postulate that confinement within quasi two-dimensional intercellular gaps exposes weak protein interactions that are not detected in solution binding assays. Micropipette-based measurements of cadherin-mediated, cell-cell binding kinetics identified a unique kinetic signature that reflects both adhesive (trans) bonds between cadherins on opposing cells and lateral (cis) interactions between cadherins on the same cell. In solution, proposed lateral interactions were not detected, even at high cadherin concentrations. Mutations postulated to disrupt lateral cadherin association altered the kinetic signatures, but did not affect the adhesive (trans) binding affinity. Perturbed kinetics further coincided with altered cadherin distributions at junctions, wound healing dynamics, and paracellular permeability. Intercellular binding kinetics thus revealed cadherin interactions that occur within confined, intermembrane gaps but not in solution. Findings further demonstrate the impact of these revealed interactions on the organization and function of intercellular junctions.
Collapse
Affiliation(s)
- Nitesh Shashikanth
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, Illinois- 61801, USA
| | - Meridith A Kisting
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, Illinois- 61801, USA
| | - Deborah E Leckband
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, Illinois- 61801, USA.,Department of Chemical and Biomolecular Engineering, and University of Illinois Urbana-Champaign, Urbana, Illinois- 61801, USA.,Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, Illinois- 61801, USA
| |
Collapse
|
29
|
Identification of E-cadherin signature motifs functioning as cleavage sites for Helicobacter pylori HtrA. Sci Rep 2016; 6:23264. [PMID: 26983597 PMCID: PMC4794652 DOI: 10.1038/srep23264] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 03/02/2016] [Indexed: 12/19/2022] Open
Abstract
The cell adhesion protein and tumour suppressor E-cadherin exhibits important functions in the prevention of gastric cancer. As a class-I carcinogen, Helicobacter pylori (H. pylori) has developed a unique strategy to interfere with E-cadherin functions. In previous studies, we have demonstrated that H. pylori secretes the protease high temperature requirement A (HtrA) which cleaves off the E-cadherin ectodomain (NTF) on epithelial cells. This opens cell-to-cell junctions, allowing bacterial transmigration across the polarised epithelium. Here, we investigated the molecular mechanism of the HtrA-E-cadherin interaction and identified E-cadherin cleavage sites for HtrA. Mass-spectrometry-based proteomics and Edman degradation revealed three signature motifs containing the [VITA]-[VITA]-x-x-D-[DN] sequence pattern, which were preferentially cleaved by HtrA. Based on these sites, we developed a substrate-derived peptide inhibitor that selectively bound and inhibited HtrA, thereby blocking transmigration of H. pylori. The discovery of HtrA-targeted signature sites might further explain why we detected a stable 90 kDa NTF fragment during H. pylori infection, but also additional E-cadherin fragments ranging from 105 kDa to 48 kDa in in vitro cleavage experiments. In conclusion, HtrA targets E-cadherin signature sites that are accessible in in vitro reactions, but might be partially masked on epithelial cells through functional homophilic E-cadherin interactions.
Collapse
|
30
|
Woichansky I, Beretta CA, Berns N, Riechmann V. Three mechanisms control E-cadherin localization to the zonula adherens. Nat Commun 2016; 7:10834. [PMID: 26960923 PMCID: PMC4792928 DOI: 10.1038/ncomms10834] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/25/2016] [Indexed: 12/13/2022] Open
Abstract
E-cadherin localization to the zonula adherens is fundamental for epithelial differentiation but the mechanisms controlling localization are unclear. Using the Drosophila follicular epithelium we genetically dissect E-cadherin transport in an in vivo model. We distinguish three mechanisms mediating E-cadherin accumulation at the zonula adherens. Two membrane trafficking pathways deliver newly synthesized E-cadherin to the plasma membrane. One is Rab11 dependent and targets E-cadherin directly to the zonula adherens, while the other transports E-cadherin to the lateral membrane. Lateral E-cadherin reaches the zonula adherens by endocytosis and targeted recycling. We show that this pathway is dependent on RabX1, which provides a functional link between early and recycling endosomes. Moreover, we show that lateral E-cadherin is transported to the zonula adherens by an apically directed flow within the plasma membrane. Differential activation of these pathways could facilitate cell shape changes during morphogenesis, while their misregulation compromises cell adhesion and tissue architecture in differentiated epithelia.
Collapse
Affiliation(s)
- Innokenty Woichansky
- Department of Cell and Molecular Biology and Division of Signaling and Functional Genomics at the German Cancer Research Center (DKFZ), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Strasse 13-17, D-68167 Mannheim, Germany
| | - Carlo Antonio Beretta
- Heidelberg University, COS and Nikon Imaging Center at the University of Heidelberg, Bioquant, D-69120 Heidelberg, Germany
- Excellenzcluster CellNetworks, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Nicola Berns
- Department of Cell and Molecular Biology and Division of Signaling and Functional Genomics at the German Cancer Research Center (DKFZ), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Strasse 13-17, D-68167 Mannheim, Germany
| | - Veit Riechmann
- Department of Cell and Molecular Biology and Division of Signaling and Functional Genomics at the German Cancer Research Center (DKFZ), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Strasse 13-17, D-68167 Mannheim, Germany
| |
Collapse
|
31
|
Ramena G, Yin Y, Yu Y, Walia V, Elble RC. CLCA2 Interactor EVA1 Is Required for Mammary Epithelial Cell Differentiation. PLoS One 2016; 11:e0147489. [PMID: 26930581 PMCID: PMC4773014 DOI: 10.1371/journal.pone.0147489] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 01/05/2016] [Indexed: 12/12/2022] Open
Abstract
CLCA2 is a p53-, p63-inducible transmembrane protein that is frequently downregulated in breast cancer. It is induced during differentiation of human mammary epithelial cells, and its knockdown causes epithelial-to-mesenchymal transition (EMT). To determine how CLCA2 promotes epithelial differentiation, we searched for interactors using membrane dihybrid screening. We discovered a strong interaction with the cell junctional protein EVA1 (Epithelial V-like Antigen 1) and confirmed it by co-immunoprecipitation. Like CLCA2, EVA1 is a type I transmembrane protein that is regulated by p53 and p63. It is thought to mediate homophilic cell-cell adhesion in diverse epithelial tissues. We found that EVA1 is frequently downregulated in breast tumors and breast cancer cell lines, especially those of mesenchymal phenotype. Moreover, knockdown of EVA1 in immortalized human mammary epithelial cells (HMEC) caused EMT, implying that EVA1 is essential for epithelial differentiation. Both EVA1 and CLCA2 co-localized with E-cadherin at cell-cell junctions. The interacting domains were delimited by deletion analysis, revealing the site of interaction to be the transmembrane segment (TMS). The primary sequence of the CLCA2 TMS was found to be conserved in CLCA2 orthologs throughout mammals, suggesting that its interaction with EVA1 co-evolved with the mammary gland. A screen for other junctional interactors revealed that CLCA2 was involved in two different complexes, one with EVA1 and ZO-1, the other with beta catenin. Overexpression of CLCA2 caused downregulation of beta catenin and beta catenin-activated genes. Thus, CLCA2 links a junctional adhesion molecule to cytosolic signaling proteins that modulate proliferation and differentiation. These results may explain how attenuation of CLCA2 causes EMT and why CLCA2 and EVA1 are frequently downregulated in metastatic breast cancer cell lines.
Collapse
Affiliation(s)
- Grace Ramena
- Dept of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois, 62794, United States of America
| | - Yufang Yin
- Dept of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, 62794, United States of America
| | - Yang Yu
- Dept of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois, 62794, United States of America
| | - Vijay Walia
- Laboratory of Cell and Developmental Signaling, National Cancer Institute-Frederick, Frederick, Maryland, 21702, United States of America
| | - Randolph C. Elble
- Dept of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, 62794, United States of America
- Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, Illinois, 62794, United States of America
- * E-mail:
| |
Collapse
|
32
|
Rizwan A, Cheng M, Bhujwalla ZM, Krishnamachary B, Jiang L, Glunde K. Breast cancer cell adhesome and degradome interact to drive metastasis. NPJ Breast Cancer 2015; 1:15017. [PMID: 28721370 PMCID: PMC5515192 DOI: 10.1038/npjbcancer.2015.17] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 09/02/2015] [Accepted: 09/17/2015] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Although primary breast tumors are detected early in most cases, it is inevitable that many patients remain at risk for future recurrence and death due to micrometastases. We investigated interactions between the degradome and the adhesome that drive metastasis, and have focused on matrix metalloproteases (MMPs) within the degradome and integrins and E-cadherin within the adhesome. AIMS The aim of this study is to identify interaction networks between adhesion molecules and degradative enzymes in breast cancer metastasis. METHODS We compared non-metastatic (BT-474, T47D, MCF7) and metastatic (MDA-MB-231, SUM149, SUM159) human breast cancer cell lines and xenografts, in which we measured growth rate, migration, invasion, colony formation, protein expression, and enzyme activity in vitro and in vivo. RESULTS The metastatic breast cancer lines and xenografts displayed higher expression and activity levels of MMPs, which was also confirmed by noninvasive imaging in vivo. These metastatic breast cancer models also displayed elevated heterophilic cell-extracellular matrix (ECM) and lower homophilic cell-cell adhesion compared with those of non-metastatic models. This was conferred by an increased expression of the heterophilic cell adhesion molecule integrin β1 (ITGB1) and a decreased expression of the homophilic cell adhesion molecule E-cadherin. Inhibition of MMPs in metastatic cells led to a reduced expression of ITGB1, and stimulation of ITGB1 resulted in higher MMP activities in metastatic cancer cells, demonstrating reciprocal dependencies between degradome and adhesome. Re-expression of E-cadherin (CDH1) led to an increased expression of the precursor form of ITGB1. CONCLUSIONS Our results point toward a concerted interdependence of MMPs, ITGB1, and CDH1 that is critical for breast cancer metastasis.
Collapse
Affiliation(s)
- Asif Rizwan
- The Johns Hopkins University In Vivo Cellular and Molecular Imaging Center, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Menglin Cheng
- The Johns Hopkins University In Vivo Cellular and Molecular Imaging Center, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zaver M Bhujwalla
- The Johns Hopkins University In Vivo Cellular and Molecular Imaging Center, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,The Johns Hopkins University School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Balaji Krishnamachary
- The Johns Hopkins University In Vivo Cellular and Molecular Imaging Center, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lu Jiang
- The Johns Hopkins University In Vivo Cellular and Molecular Imaging Center, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kristine Glunde
- The Johns Hopkins University In Vivo Cellular and Molecular Imaging Center, Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,The Johns Hopkins University School of Medicine, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| |
Collapse
|
33
|
Farr GA, Hull M, Stoops EH, Bateson R, Caplan MJ. Dual pulse-chase microscopy reveals early divergence in the biosynthetic trafficking of the Na,K-ATPase and E-cadherin. Mol Biol Cell 2015; 26:4401-11. [PMID: 26424804 PMCID: PMC4666135 DOI: 10.1091/mbc.e14-09-1385] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 09/24/2015] [Indexed: 11/14/2022] Open
Abstract
The trafficking of newly synthesized Na,K-ATPase and E-cadherin is observed in polarized epithelial cells. E-cadherin’s exit from the Golgi complex is not susceptible to 19°C temperature block. Furthermore, these proteins exit the Golgi and are delivered to the basolateral cell surface in separate vascular carriers. Recent evidence indicates that newly synthesized membrane proteins that share the same distributions in the plasma membranes of polarized epithelial cells can pursue a variety of distinct trafficking routes as they travel from the Golgi complex to their common destination at the cell surface. In most polarized epithelial cells, both the Na,K-ATPase and E-cadherin are localized to the basolateral domains of the plasma membrane. To examine the itineraries pursued by newly synthesized Na,K-ATPase and E-cadherin in polarized MDCK epithelial cells, we used the SNAP and CLIP labeling systems to fluorescently tag temporally defined cohorts of these proteins and observe their behaviors simultaneously as they traverse the secretory pathway. These experiments reveal that E-cadherin is delivered to the cell surface substantially faster than is the Na,K-ATPase. Furthermore, the surface delivery of newly synthesized E-cadherin to the plasma membrane was not prevented by the 19°C temperature block that inhibits the trafficking of most proteins, including the Na,K-ATPase, out of the trans-Golgi network. Consistent with these distinct behaviors, populations of newly synthesized E-cadherin and Na,K-ATPase become separated from one another within the trans-Golgi network, suggesting that they are sorted into different carrier vesicles that mediate their post-Golgi trafficking.
Collapse
Affiliation(s)
- Glen A Farr
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520-8026
| | - Michael Hull
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520-8026
| | - Emily H Stoops
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520-8026
| | - Rosalie Bateson
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520-8026
| | - Michael J Caplan
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520-8026 )
| |
Collapse
|
34
|
de la Fuente-Ortega E, Gravotta D, Perez Bay A, Benedicto I, Carvajal-Gonzalez JM, Lehmann GL, Lagos CF, Rodríguez-Boulan E. Basolateral sorting of chloride channel 2 is mediated by interactions between a dileucine motif and the clathrin adaptor AP-1. Mol Biol Cell 2015; 26:1728-42. [PMID: 25739457 PMCID: PMC4436783 DOI: 10.1091/mbc.e15-01-0047] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 02/25/2015] [Indexed: 01/03/2023] Open
Abstract
ClC-2 is a ubiquitous chloride channel that regulates cell volume, ion transport, and acid-base balance. Mice knocked out for ClC-2 are blind and sterile. Basolateral localization of ClC-2 in epithelia is mediated by the interaction of a dileucine motif with a highly conserved pocket in the γ1-σ1A hemicomplex of AP-1. In spite of the many key cellular functions of chloride channels, the mechanisms that mediate their subcellular localization are largely unknown. ClC-2 is a ubiquitous chloride channel usually localized to the basolateral domain of epithelia that regulates cell volume, ion transport, and acid–base balance; mice knocked out for ClC-2 are blind and sterile. Previous work suggested that CLC-2 is sorted basolaterally by TIFS812LL, a dileucine motif in CLC-2's C-terminal domain. However, our in silico modeling of ClC-2 suggested that this motif was buried within the channel's dimerization interface and identified two cytoplasmically exposed dileucine motifs, ESMI623LL and QVVA635LL, as candidate sorting signals. Alanine mutagenesis and trafficking assays support a scenario in which ESMI623LL acts as the authentic basolateral signal of ClC-2. Silencing experiments and yeast three-hybrid assays demonstrated that both ubiquitous (AP-1A) and epithelium-specific (AP-1B) forms of the tetrameric clathrin adaptor AP-1 are capable of carrying out basolateral sorting of ClC-2 through interactions of ESMI623LL with a highly conserved pocket in their γ1-σ1A hemicomplex.
Collapse
Affiliation(s)
- Erwin de la Fuente-Ortega
- Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, NY 10065
| | - Diego Gravotta
- Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, NY 10065
| | - Andres Perez Bay
- Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, NY 10065
| | - Ignacio Benedicto
- Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, NY 10065
| | | | - Guillermo L Lehmann
- Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, NY 10065
| | - Carlos F Lagos
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago Centro 8330074, Santiago, Chile Facultad de Ciencia, Universidad San Sebastián, Providencia 7510157, Santiago, Chile
| | - Enrique Rodríguez-Boulan
- Dyson Vision Research Institute, Department of Ophthalmology, Weill Cornell Medical College, New York, NY 10065
| |
Collapse
|
35
|
Pelissier-Rota MA, Chartier NT, Jacquier-Sarlin MR. Dynamic Regulation of Adherens Junctions: Implication in Cell Differentiation and Tumor Development. INTERCELLULAR COMMUNICATION IN CANCER 2015:53-149. [DOI: 10.1007/978-94-017-7380-5_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
36
|
Stoops EH, Caplan MJ. Trafficking to the apical and basolateral membranes in polarized epithelial cells. J Am Soc Nephrol 2014; 25:1375-86. [PMID: 24652803 DOI: 10.1681/asn.2013080883] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Renal epithelial cells must maintain distinct protein compositions in their apical and basolateral membranes in order to perform their transport functions. The creation of these polarized protein distributions depends on sorting signals that designate the trafficking route and site of ultimate functional residence for each protein. Segregation of newly synthesized apical and basolateral proteins into distinct carrier vesicles can occur at the trans-Golgi network, recycling endosomes, or a growing assortment of stations along the cellular trafficking pathway. The nature of the specific sorting signal and the mechanism through which it is interpreted can influence the route a protein takes through the cell. Cell type-specific variations in the targeting motifs of a protein, as are evident for Na,K-ATPase, demonstrate a remarkable capacity to adapt sorting pathways to different developmental states or physiologic requirements. This review summarizes our current understanding of apical and basolateral trafficking routes in polarized epithelial cells.
Collapse
Affiliation(s)
- Emily H Stoops
- Departments of Cellular & Molecular Physiology and Cell Biology, Yale University School of Medicine, New Haven, Connecticut
| | - Michael J Caplan
- Departments of Cellular & Molecular Physiology and Cell Biology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
37
|
Kowalczyk AP, Nanes BA. Adherens junction turnover: regulating adhesion through cadherin endocytosis, degradation, and recycling. Subcell Biochem 2014; 60:197-222. [PMID: 22674073 DOI: 10.1007/978-94-007-4186-7_9] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Adherens junctions are important mediators of intercellular adhesion, but they are not static structures. They are regularly formed, broken, and rearranged in a variety of situations, requiring changes in the amount of cadherins, the main adhesion molecule in adherens junctions, present at the cell surface. Thus, endocytosis, degradation, and recycling of cadherins are crucial for dynamic regulation of adherens junctions and control of intercellular adhesion. In this chapter, we review the involvement of cadherin endocytosis in development and disease. We discuss the various endocytic pathways available to cadherins, the adaptors involved, and the sorting of internalized cadherin for recycling or lysosomal degradation. In addition, we review the regulatory pathways controlling cadherin endocytosis and degradation, including regulation of cadherin endocytosis by catenins, cadherin ubiquitination, and growth factor receptor signaling pathways. Lastly, we discuss the proteolytic cleavage of cadherins at the plasma membrane.
Collapse
Affiliation(s)
- Andrew P Kowalczyk
- Department of Cell Biology, Emory University School of Medicine, 30332, Atlanta, GA, USA,
| | | |
Collapse
|
38
|
|
39
|
Van Itallie CM, Tietgens AJ, Aponte A, Fredriksson K, Fanning AS, Gucek M, Anderson JM. Biotin ligase tagging identifies proteins proximal to E-cadherin, including lipoma preferred partner, a regulator of epithelial cell-cell and cell-substrate adhesion. J Cell Sci 2013; 127:885-95. [PMID: 24338363 DOI: 10.1242/jcs.140475] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Known proteins associated with the cell-adhesion protein E-cadherin include catenins and proteins involved in signaling, trafficking and actin organization. However, the list of identified adherens junction proteins is likely to be incomplete, limiting investigation into this essential cell structure. To expand the inventory of potentially relevant proteins, we expressed E-cadherin fused to biotin ligase in MDCK epithelial cells, and identified by mass spectrometry neighboring proteins that were biotinylated. The most abundant of the 303 proteins identified were catenins and nearly 40 others that had been previously reported to influence cadherin function. Many others could be rationalized as novel candidates for regulating the adherens junction, cytoskeleton, trafficking or signaling. We further characterized lipoma preferred partner (LPP), which is present at both cell contacts and focal adhesions. Knockdown of LPP demonstrated its requirement for E-cadherin-dependent adhesion and suggested that it plays a role in coordination of the cell-cell and cell-substrate cytoskeletal interactions. The analysis of LPP function demonstrates proof of principle that the proteomic analysis of E-cadherin proximal proteins expands the inventory of components and tools for understanding the function of E-cadherin.
Collapse
Affiliation(s)
- Christina M Van Itallie
- Laboratory of Tight Junction Structure and Function, NHLBI, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Ivanov AI, Naydenov NG. Dynamics and regulation of epithelial adherens junctions: recent discoveries and controversies. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 303:27-99. [PMID: 23445808 DOI: 10.1016/b978-0-12-407697-6.00002-7] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Adherens junctions (AJs) are evolutionarily conserved plasma-membrane structures that mediate cell-cell adhesions in multicellular organisms. They are organized by several types of adhesive integral membrane proteins, most notably cadherins and nectins that are clustered and stabilized by a number of cytoplasmic scaffolds. AJs are key regulators of tissue architecture and dynamics via control of cell proliferation, polarity, shape, motility, and survival. They are absolutely critical for normal tissue morphogenesis and their disruption results in pathological abnormalities in different tissues. Although the field of adherens-junction research dramatically progressed in recent years, a number of important questions remain controversial and poorly understood. This review outlines basic principles that regulate organization of AJs in mammalian epithelia and discusses recent advances and standing controversies in the field. A special attention is paid to the regulation of AJs by vesicle trafficking and the intracellular cytoskeleton as well as roles and mechanisms of adherens-junction disruption during tumor progression and tissue inflammation.
Collapse
Affiliation(s)
- Andrei I Ivanov
- Department of Human and Molecular Genetics, Virginia Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA.
| | | |
Collapse
|
41
|
Regulation of the Epithelial-Mesenchymal Transition by Claudin-3 and Claudin-4. PLoS One 2013; 8:e67496. [PMID: 23805314 PMCID: PMC3689737 DOI: 10.1371/journal.pone.0067496] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 05/20/2013] [Indexed: 01/06/2023] Open
Abstract
The mechanisms that control intracellular adhesion are central to the process of invasion and metastasis. Claudin-3 (CLDN3) and claudin-4 (CLDN4) are major structural molecules of the tight junctions that link epithelial cells. Our prior work has demonstrated that knockdown of the expression of either CLDN3 or CLDN4 produces marked changes in the phenotype of ovarian carcinoma cells including increases in growth rate in vivo, migration, invasion, metastasis, and drug resistance, similar to those produced by the epithelial-to-mesenchymal transition (EMT). We postulated that these changes may result from the ability of CLDN3 or CLDN4 to suppress EMT. In this study we found that knockdown of either CLDN3 or CLDN4 increased cell size and resulted in flattened morphology. While knockdown of CLDN3 or CLDN4 did not alter the expression of vimentin, it significantly down-regulated the level of E-cadherin and up-regulated N-cadherin expression. Conversely, over-expression of CLDN3 or CLDN4 in a cell line that does not express endogenous CLDN3 or CLDN4 decreased N-cadherin expression. Re-expression of E-cadherin in the CLDN3 or CLDN4 knockdown cells reduced migration, invasion and tumor growth in vivo. Loss of either CLDN3 or CLDN4 resulted in activation of the PI3K pathway as evidenced by increased Akt phosphorylation, elevated cellular PIP3 content and PI3K activity as well as up-regulation of the mRNA and protein levels of the transcription factor Twist. Taken together, these findings suggest that CLDN3 and CLDN4 function to sustain an epithelial phenotype and that their loss promotes EMT.
Collapse
|
42
|
The polarization of the G-protein activated potassium channel GIRK5 to the vegetal pole of Xenopus laevis oocytes is driven by a di-leucine motif. PLoS One 2013; 8:e64096. [PMID: 23717539 PMCID: PMC3661522 DOI: 10.1371/journal.pone.0064096] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 04/07/2013] [Indexed: 11/19/2022] Open
Abstract
The G protein-coupled inwardly-rectifying potassium channels (known as GIRK or Kir3) form functional heterotetramers gated by G-βγ subunits. GIRK channels participate in heart rate modulation and neuronal postsynaptic inhibition in mammals. In Xenopus laevis oocytes, GIRK5 is a functional homomultimer. Previously, we found that phosphorylation of a tyrosine (Y16) at its N-terminus downregulates the surface expression of GIRK5. In this work, we elucidated the subcellular localization and trafficking of GIRK5 in oocytes. Several EGFP-GIRK5 chimeras were produced and an ECFP construct was used to identify the endoplasmic reticulum (ER). Whereas GIRK5-WT was retained in the ER at the animal pole, the phospho-null GIRK5-Y16A was localized to the vegetal pole. Interestingly, a construct with an N-terminal Δ25 deletion produced an even distribution of the channel in the whole oocyte. Through an alanine-scan, we identified an acidic cluster/di-leucine sorting-signal recognition motif between E17 and I22. We quantified the effect of each amino acid residue within this di-leucine motif in determining the distribution of GIRK5 to the animal and vegetal poles. We found that Y16 and I22 contributed to functional expression and were dominant in the polarization of GIRK5. We thus conclude that the N-terminal acidic di-leucine motif of GIRK5 determines its retention and polarized trafficking within Xl oocytes.
Collapse
|
43
|
|
44
|
Jenkins PM, Vasavda C, Hostettler J, Davis JQ, Abdi K, Bennett V. E-cadherin polarity is determined by a multifunction motif mediating lateral membrane retention through ankyrin-G and apical-lateral transcytosis through clathrin. J Biol Chem 2013; 288:14018-14031. [PMID: 23530049 PMCID: PMC3656260 DOI: 10.1074/jbc.m113.454439] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
We report a highly conserved motif in the E-cadherin juxtamembrane domain that determines apical-lateral polarity by conferring both restricted mobility at the lateral membrane and transcytosis of apically mis-sorted protein to the lateral membrane. Mutations causing either increased lateral membrane mobility or loss of apical-lateral transcytosis result in partial mis-sorting of E-cadherin in Madin-Darby canine kidney cells. However, loss of both activities results in complete loss of polarity. We present evidence that residues required for restricted mobility mediate retention at the lateral membrane through interaction with ankyrin-G, whereas dileucine residues conferring apical-lateral transcytosis act through a clathrin-dependent process and function in an editing pathway. Ankyrin-G interaction with E-cadherin is abolished by the same mutations resulting in increased E-cadherin mobility. Clathrin heavy chain knockdown and dileucine mutation of E-cadherin both cause the same partial loss of polarity of E-cadherin. Moreover, clathrin knockdown causes no further change in polarity of E-cadherin with dileucine mutation but does completely randomize E-cadherin mutants lacking ankyrin-binding. Dileucine mutation, but not loss of ankyrin binding, prevented transcytosis of apically mis-sorted E-cadherin to the lateral membrane. Finally, neurofascin, which binds ankyrin but lacks dileucine residues, exhibited partial apical-lateral polarity that was abolished by mutation of its ankyrin-binding site but was not affected by clathrin knockdown. The polarity motif thus integrates complementary activities of lateral membrane retention through ankyrin-G and apical-lateral transcytosis of mis-localized protein through clathrin. Together, the combination of retention and editing function to ensure a high fidelity steady state localization of E-cadherin at the lateral membrane.
Collapse
Affiliation(s)
- Paul M Jenkins
- Howard Hughes Medical Institute, Duke University Medical Center, Durham, North Carolina 27710; Department of Biochemistry, Duke University Medical Center, Durham, North Carolina 27710
| | - Chirag Vasavda
- Department of Biochemistry, Duke University Medical Center, Durham, North Carolina 27710
| | - Janell Hostettler
- Howard Hughes Medical Institute, Duke University Medical Center, Durham, North Carolina 27710
| | - Jonathan Q Davis
- Howard Hughes Medical Institute, Duke University Medical Center, Durham, North Carolina 27710
| | - Khadar Abdi
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27710
| | - Vann Bennett
- Howard Hughes Medical Institute, Duke University Medical Center, Durham, North Carolina 27710; Department of Biochemistry, Duke University Medical Center, Durham, North Carolina 27710; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27710; Department of Neurobiology, Duke University Medical Center, Durham, North Carolina 27710.
| |
Collapse
|
45
|
Fibroblast growth factor 2 induces E-cadherin down-regulation via PI3K/Akt/mTOR and MAPK/ERK signaling in ovarian cancer cells. PLoS One 2013; 8:e59083. [PMID: 23554977 PMCID: PMC3598697 DOI: 10.1371/journal.pone.0059083] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 02/11/2013] [Indexed: 11/19/2022] Open
Abstract
Fibroblast growth factor 2 (FGF2) is produced by ovarian cancer cells and it has been suggested to play an important role in tumor progression. In this study, we report that FGF2 treatment down-regulated E-cadherin by up-regulating its transcriptional repressors, Slug and ZEB1, in human ovarian cancer cells. The pharmacological inhibition of phosphatidylinositol-3-kinase (PI3K), mammalian target of rapamycin (mTOR), and MEK suggests that both PI3K/Akt/mTOR and MAPK/ERK signaling are required for FGF2-induced E-cadherin down-regulation. Moreover, FGF2 up-regulated Slug and ZEB1 expression via the PI3K/Akt/mTOR and MAPK/ERK signaling pathways, respectively. Finally, FGF2-induced cell invasion was abolished by the inhibition of the PI3K/Akt/mTOR and MAPK/ERK pathways, and the forced expression of E-cadherin diminished the intrinsic invasiveness of ovarian cancer cells as well as the FGF2-induced cell invasion. This study demonstrates a novel mechanism in which FGF2 down-regulates E-cadherin expression through the activation of PI3K/Akt/mTOR and MAPK/ERK signaling, and the up-regulation of Slug and ZEB1 in human ovarian cancer cells.
Collapse
|
46
|
Bibee KP, Augustin R, Gazit V, Moley KH. The apical sorting signal for human GLUT9b resides in the N-terminus. Mol Cell Biochem 2013; 376:163-73. [PMID: 23361362 DOI: 10.1007/s11010-013-1564-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Accepted: 01/18/2013] [Indexed: 12/11/2022]
Abstract
The two splice variants of human glucose transporter 9 (hGLUT9) are targeted to different polarized membranes. hGLUT9a traffics to the basolateral membrane, whereas hGLUT9b traffics to the apical region. This study examines the sorting mechanism of these variants, which differ only in their N-terminal domain. Mutating a di-leucine motif unique to GLUT9a did not affect targeting. Chimeric proteins were made using GLUT1, a basolaterally targeted transporter, and GLUT3, an apically targeted protein whose signal lies in the C-terminus. Overexpression of the chimeric proteins in polarized cells demonstrates that the N-terminus of hGLUT9b contains a signal capable of redirecting GLUT1 to the apical membrane. The N-terminus of hGLUT9a, however, does not contain a basolateral signal sufficient enough to redirect GLUT3. Portions of the GLUT9a N-terminus were substituted with corresponding portions of the GLUT9b N-terminus to determine the motif responsible for apical targeting. The first 16 amino acids were not found to be a sufficient apical signal. The last ten amino acids of the N-termini differ only in amino-acid class at one location. In the B-form, leucine, a hydrophobic residue, is substituted for lysine, a basic residue, found in the A-form. However, mutation of the leucine in hGLUT9b to a lysine resulted in retention of the apical signal. We therefore believe the apical signal exists as an interplay between the final ten amino acids of the N-terminus and another motif within the protein such as the intracellular loop or other motifs within the N-terminus.
Collapse
Affiliation(s)
- Kristin P Bibee
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
47
|
Abstract
Epithelial cells have an apical-basolateral axis of polarity, which is required for epithelial functions including barrier formation, vectorial ion transport and sensory perception. Here we review what is known about the sorting signals, machineries and pathways that maintain this asymmetry, and how polarity proteins interface with membrane-trafficking pathways to generate membrane domains de novo. It is becoming apparent that membrane traffic does not simply reinforce polarity, but is critical for the generation of cortical epithelial cell asymmetry.
Collapse
|
48
|
Saito M, Tucker DK, Kohlhorst D, Niessen CM, Kowalczyk AP. Classical and desmosomal cadherins at a glance. J Cell Sci 2012; 125:2547-52. [PMID: 22833291 DOI: 10.1242/jcs.066654] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Masataka Saito
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA
| | | | | | | | | |
Collapse
|
49
|
Xia H, Ooi LLPJ, Hui KM. MiR-214 targets β-catenin pathway to suppress invasion, stem-like traits and recurrence of human hepatocellular carcinoma. PLoS One 2012; 7:e44206. [PMID: 22962603 PMCID: PMC3433464 DOI: 10.1371/journal.pone.0044206] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 07/30/2012] [Indexed: 12/18/2022] Open
Abstract
The down-regulation of miR-214 has previously been observed in human hepatocellular carcinoma (HCC). Here, we demonstrated the down-regulation of miR-214 is associated with cell invasion, stem-like traits and early recurrence of HCC. Firstly, we validated the suppression of miR-214 in human HCC by real-time quantitative RT-PCR (qRT-PCR) in 20 paired tumor and non-tumor liver tissues of HCC patients and 10 histologically normal liver tissues from colorectal cancer patients with liver metastases. Further qRT-PCR analysis of 50 HCC tissues from an independent cohort of HCC patients of whom 29 with early recurrent disease (<2 years) and 21 with late recurrent disease demonstrated that the suppression of miR-214 was significantly more suppressed in samples from HCC patients with early recurrent disease compared those from patients with no recurrence. Re-expression of miR-214 significantly suppressed the growth of HCC cells in vitro and reduced their tumorigenicity in vivo. The enhancer of zeste homologue 2 (EZH2) and β-catenin (CTNNB1) was identified as two potential direct downstream targets of miR-214 through bioinformatics analysis and experimentally validated the miRNA-target interactions with a dual-firefly luciferase reporter assay. In corroborate with this, both EZH2 and CTNNB1 are found to be significantly overexpressed in human HCC biopsies. Since EZH2 can regulate CTNNB1, CTNNB1 can also be an indirect target of miR-214 through EZH2. Silencing EZH2 or CTNNB1 expression suppressed the growth and invasion of HCC cells and induced E-cadherin (CDH1), known to inhibit cell invasion and metastasis. Furthermore, the silencing of miR-214 or overexpression of EZH2 increased EpCAM+ stem-like cells through the activation of CTNNB1. Interestingly, the up-regulation of EZH2, CTNNB1 and the down-regulation of CDH1 in HCC patients correlated with early recurrent disease and can be an independent predictor of poor survival. Therefore, miR-214 can directly or indirectly target CTNNB1 to modulate the β-catenin signaling pathway in HCC.
Collapse
MESH Headings
- Animals
- Antigens, CD
- Cadherins/genetics
- Cadherins/metabolism
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/mortality
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- Computational Biology
- Enhancer of Zeste Homolog 2 Protein
- Gene Expression Regulation, Neoplastic
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/mortality
- Liver Neoplasms/pathology
- Male
- Mice
- Mice, Nude
- MicroRNAs/genetics
- MicroRNAs/metabolism
- MicroRNAs/pharmacology
- Neoplasm Invasiveness
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/metabolism
- Neoplasm Recurrence, Local/mortality
- Neoplasm Recurrence, Local/pathology
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Polycomb Repressive Complex 2/genetics
- Polycomb Repressive Complex 2/metabolism
- Protein Binding
- Signal Transduction
- Survival Rate
- Tumor Cells, Cultured
- beta Catenin/genetics
- beta Catenin/metabolism
Collapse
Affiliation(s)
- Hongping Xia
- Bek Chai Heah Laboratory of Cancer Genomics, Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, Singapore, Singapore
| | | | - Kam M. Hui
- Bek Chai Heah Laboratory of Cancer Genomics, Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, Singapore, Singapore
- Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, A*STAR, Biopolis Drive Proteos, Singapore, Singapore
- * E-mail:
| |
Collapse
|
50
|
Li HC, Kucher V, Li EY, Conforti L, Zahedi KA, Soleimani M. The role of aspartic acid residues 405 and 416 of the kidney isotype of sodium-bicarbonate cotransporter 1 in its targeting to the plasma membrane. Am J Physiol Cell Physiol 2012; 302:C1713-30. [PMID: 22442137 DOI: 10.1152/ajpcell.00147.2011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The NH(2) terminus of the sodium-bicarbonate cotransporter 1 (NBCe1) plays an important role in its targeting to the plasma membrane. To identify the amino acid residues that contribute to the targeting of NBCe1 to the plasma membrane, polarized MDCK cells were transfected with expression constructs coding for green fluorescent protein (GFP)-tagged NBCe1 NH(2)-terminal deletion mutants, and the localization of GFP-tagged proteins was analyzed by confocal microscopy. Our results indicate that the amino acids between residues 399 and 424 of NBCe1A contain important sequences that contribute to its localization to the plasma membrane. Site-directed mutagenesis studies showed that GFP-NBCe1A mutants D405A and D416A are retained in the cytoplasm of the polarized MDCK epithelial cells. Examination of functional activities of D405A and D416A reveals that their activities are reduced compared with the wild-type NBCe1A. Similarly, aspartic acid residues 449 and 460 of pancreatic NBCe1 (NBCe1B), which correspond to residues 405 and 416 of NBCe1A, are also required for its full functional activity and accurate targeting to the plasma membrane. In addition, while replacement of D416 with glutamic acid did not affect the targeting or functional activity of NBCe1A, substitution of D405 with glutamic acid led to the retention of the mutated protein in the intracellular compartment and impaired functional activity. These studies demonstrate that aspartic acid residues 405 and 416 in the NH(2) terminus of NBCe1A are important in its accurate targeting to the plasma membrane.
Collapse
Affiliation(s)
- Hong C Li
- Department of Medicine, University of Cincinnati, Ohio 45267-0585, USA.
| | | | | | | | | | | |
Collapse
|