1
|
Naturale VF, Pickett MA, Feldman JL. Persistent cell contacts enable E-cadherin/HMR-1- and PAR-3-based symmetry breaking within a developing C. elegans epithelium. Dev Cell 2023; 58:1830-1846.e12. [PMID: 37552986 PMCID: PMC10592304 DOI: 10.1016/j.devcel.2023.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 05/10/2023] [Accepted: 07/17/2023] [Indexed: 08/10/2023]
Abstract
Tissue-wide patterning is essential to multicellular development, requiring cells to individually generate polarity axes and coordinate them in space and time with neighbors. Using the C. elegans intestinal epithelium, we identified a patterning mechanism that is informed by cell contact lifetime asymmetry and executed via the scaffolding protein PAR-3 and the transmembrane protein E-cadherin/HMR-1. Intestinal cells break symmetry as PAR-3 and HMR-1 recruit apical determinants into punctate "local polarity complexes" (LPCs) at homotypic contacts. LPCs undergo an HMR-1-based migration to a common midline, thereby establishing tissue-wide polarity. Thus, symmetry breaking results from PAR-3-dependent intracellular polarization coupled to HMR-1-based tissue-level communication, which occurs through a non-adhesive signaling role for HMR-1. Differential lifetimes between homotypic and heterotypic cell contacts are created by neighbor exchanges and oriented divisions, patterning where LPCs perdure and thereby breaking symmetry. These cues offer a logical and likely conserved framework for how epithelia without obvious molecular asymmetries can polarize.
Collapse
Affiliation(s)
| | - Melissa A Pickett
- Department of Biology, Stanford University, Stanford, CA 94305, USA; Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Jessica L Feldman
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
2
|
Wu Y, Sun SX. Mechanics of cell-cell junctions. Biophys J 2023; 122:3354-3368. [PMID: 37475215 PMCID: PMC10465726 DOI: 10.1016/j.bpj.2023.07.011] [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: 02/02/2023] [Revised: 06/01/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023] Open
Abstract
Tissue cells in epithelial or endothelial monolayers are connected through cell-cell junctions, which are stabilized by transmembrane E-cadherin bonds and intracellular actin filaments. These bonds and junctions play a crucial role in maintaining the barrier function of epithelia and endothelia and are believed to transmit forces between cells. Additionally, E-cadherin bonds can impact the shape of cell-cell junctions. In this study, we develop a continuum mechanical model of the cell-cell junction by explicitly incorporating the cell membrane, distributions of E-cadherin bonds, cytoplasmic fluid pressure, and F-actin dynamics. The static force-balanced version of the model is able to analyze the influences of cell cortical tension, actin dynamics, and cytoplasmic pressure on the junction shape and E-cadherin bonds. Furthermore, an extended model that incorporates fluid flow, across the cell boundary as well as around the cell, is also examined. This model can couple cell-shape changes with cell cortical tension and fluid flow, and predicts the additional effect of fluid motion on cell-cell junction mechanics. Taken together, our models serve as an intermediate link between molecular-scale models of cell-junction molecules and cell-scale models of tissue and epithelia.
Collapse
Affiliation(s)
- Yufei Wu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland; Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Sean X Sun
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland; Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland; Center for Cell Dynamics, Johns Hopkins School of Medicine, Baltimore, Maryland.
| |
Collapse
|
3
|
Nishiguchi S, Kasai RS, Uchihashi T. Antiparallel dimer structure of CELSR cadherin in solution revealed by high-speed atomic force microscopy. Proc Natl Acad Sci U S A 2023; 120:e2302047120. [PMID: 37094146 PMCID: PMC10160967 DOI: 10.1073/pnas.2302047120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/03/2023] [Indexed: 04/26/2023] Open
Abstract
Cadherin EGF LAG seven-pass G-type receptors (CELSR) cadherins, members of the cadherin superfamily, and adhesion G-protein-coupled receptors, play a vital role in cell-cell adhesion. The mutual binding of the extracellular domains (ectodomains) of CELSR cadherins between cells is crucial for tissue formation, including the establishment of planar cell polarity, which directs the proper patterning of cells. CELSR cadherins possess nine cadherin ectodomains (EC1-EC9) and noncadherin ectodomains. However, the structural and functional mechanisms of the binding mode of CELSR cadherins have not been determined. In this study, we investigated the binding mode of CELSR cadherins using single-molecule fluorescence microscopy, high-speed atomic force microscopy (HS-AFM), and bead aggregation assay. The fluorescence microscopy analysis results indicated that the trans-dimer of the CELSR cadherin constitutes the essential adhesive unit between cells. HS-AFM analysis and bead aggregation assay results demonstrated that EC1-EC8 entirely overlap and twist to form antiparallel dimer conformations and that the binding of EC1-EC4 is sufficient to sustain bead aggregation. The interaction mechanism of CELSR cadherin may elucidate the variation of the binding mechanism within the cadherin superfamily and physiological role of CELSR cadherins in relation to planar cell polarity.
Collapse
Affiliation(s)
- Shigetaka Nishiguchi
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki444-8787, Japan
| | - Rinshi S. Kasai
- Institute for Life and Medical Sciences, Kyoto University, Kyoto606-8507, Japan
- Institute for Glyco-core Research, Gifu University, Gifu501-1193, Japan
| | - Takayuki Uchihashi
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki444-8787, Japan
- Department of Physics, Nagoya University, Nagoya464-8602, Japan
- Institute for Glyco-core Research, Nagoya University, Nagoya464-8602, Japan
| |
Collapse
|
4
|
Srinivas CS, Singaraju GS, Kaur V, Das S, Ghosh SK, Sagar A, Kumar A, Bhatia T, Rakshit S. Transient interactions drive the lateral clustering of cadherin-23 on membrane. Commun Biol 2023; 6:293. [PMID: 36934176 PMCID: PMC10024700 DOI: 10.1038/s42003-023-04677-6] [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: 09/21/2022] [Accepted: 03/07/2023] [Indexed: 03/20/2023] Open
Abstract
Cis and trans-interactions among cadherins secure multicellularity. While the molecular structure of trans-interactions of cadherins is well understood, work to identify the molecular cues that spread the cis-interactions two-dimensionally is still ongoing. Here, we report that transient, weak, yet multivalent, and spatially distributed hydrophobic interactions that are involved in liquid-liquid phase separations of biomolecules in solution, alone can drive the lateral-clustering of cadherin-23 on a membrane. No specific cis-dimer interactions are required for the lateral clustering. In cells, the cis-clustering accelerates cell-cell adhesion and, thus, contributes to cell-adhesion kinetics along with strengthening the junction. Although the physiological connection of cis-clustering with rapid adhesion is yet to be explored, we speculate that the over-expression of cadherin-23 in M2-macrophages may facilitate faster attachments to circulatory tumor cells during metastasis.
Collapse
Affiliation(s)
- Cheerneni S Srinivas
- Department of Chemical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Gayathri S Singaraju
- Department of Chemical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Veerpal Kaur
- Department of Chemical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Sayan Das
- Department of Chemical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Sanat K Ghosh
- Department of Physical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Amin Sagar
- Department of Chemical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Anuj Kumar
- Department of Physical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
- Centre for Protein Science Design and Engineering, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Tripta Bhatia
- Department of Physical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Sabyasachi Rakshit
- Department of Chemical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India.
- Centre for Protein Science Design and Engineering, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India.
| |
Collapse
|
5
|
Wang J, Liu J, Luo M, Cui H, Zhang W, Zhao K, Dai H, Song F, Chen K, Yu Y, Zhou D, Li MJ, Yang H. Rational drug repositioning for coronavirus-associated diseases using directional mapping and side-effect inference. iScience 2022; 25:105348. [PMID: 36267550 PMCID: PMC9556799 DOI: 10.1016/j.isci.2022.105348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/02/2022] [Accepted: 10/11/2022] [Indexed: 02/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the pathogen of coronavirus disease 2019 (COVID-19), has infected hundreds of millions of people and caused millions of deaths. Looking for valid druggable targets with minimal side effects for the treatment of COVID-19 remains critical. After discovering host genes from multiscale omics data, we developed an end-to-end network method to investigate drug-host gene(s)-coronavirus (CoV) paths and the mechanism of action between the drug and the host factor in a directional network. We also inspected the potential side effect of the candidate drug on several common comorbidities. We established a catalog of host genes associated with three CoVs. Rule-based prioritization yielded 29 Food and Drug Administration (FDA)-approved drugs via accounting for the effects of drugs on CoVs, comorbidities, and drug-target confidence information. Seven drugs are currently undergoing clinical trials as COVID-19 treatment. This catalog of druggable host genes associated with CoVs and the prioritized repurposed drugs will provide a new sight in therapeutics discovery for severe COVID-19 patients.
Collapse
Affiliation(s)
- Jianhua Wang
- Department of Epidemiology and Biostatistics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China,Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China,Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jiaojiao Liu
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Menghan Luo
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China,Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hui Cui
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Wenwen Zhang
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China,Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ke Zhao
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China,Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hongji Dai
- Department of Epidemiology and Biostatistics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Fangfang Song
- Department of Epidemiology and Biostatistics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Ying Yu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Dongming Zhou
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China,Corresponding author
| | - Mulin Jun Li
- Department of Epidemiology and Biostatistics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China,Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China,Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China,Corresponding author
| | - Hongxi Yang
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China,Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China,Corresponding author
| |
Collapse
|
6
|
Multiple dimeric structures and strand-swap dimerization of E-cadherin in solution visualized by high-speed atomic force microscopy. Proc Natl Acad Sci U S A 2022; 119:e2208067119. [PMID: 35867820 PMCID: PMC9335211 DOI: 10.1073/pnas.2208067119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Classical cadherins play key roles in cell-cell adhesion. The adhesion process is thought to comprise mainly two steps: X-dimer and strand-swap (SS-) dimer formation of the extracellular domains (ectodomains) of cadherins. The dimerization mechanism of this two-step process has been investigated for type I cadherins, including E-cadherin, of classical cadherins, whereas other binding states also have been proposed, raising the possibility of additional binding processes required for the cadherin dimerization. However, technical limitations in observing single-molecule structures and their dynamics have precluded the investigation of the dynamic binding process of cadherin. Here, we used high-speed atomic force microscopy (HS-AFM) to observe full-length ectodomains of E-cadherin in solution and identified multiple dimeric structures that had not been reported previously. HS-AFM revealed that almost half of the cadherin dimers showed S- (or reverse S-) shaped conformations, which had more dynamic properties than the SS- and X-like dimers. The combined HS-AFM, mutational, and molecular modeling analyses showed that the S-shaped dimer was formed by membrane-distal ectodomains, while the binding interface was different from that of SS- and X-dimers. Furthermore, the formation of the SS-dimer from the S-shaped and X-like dimers was directly visualized, suggesting the processes of SS-dimer formation from S-shaped and X-dimers during cadherin dimerization.
Collapse
|
7
|
Thompson CJ, Vu VH, Leckband DE, Schwartz DK. Cadherin cis and trans interactions are mutually cooperative. Proc Natl Acad Sci U S A 2021; 118:e2019845118. [PMID: 33658369 PMCID: PMC7958404 DOI: 10.1073/pnas.2019845118] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Cadherin transmembrane proteins are responsible for intercellular adhesion in all biological tissues and modulate tissue morphogenesis, cell motility, force transduction, and macromolecular transport. The protein-mediated adhesions consist of adhesive trans interactions and lateral cis interactions. Although theory suggests cooperativity between cis and trans bonds, direct experimental evidence of such cooperativity has not been demonstrated. Here, the use of superresolution microscopy, in conjunction with intermolecular single-molecule Förster resonance energy transfer, demonstrated the mutual cooperativity of cis and trans interactions. Results further demonstrate the consequent assembly of large intermembrane junctions, using a biomimetic lipid bilayer cell adhesion model. Notably, the presence of cis interactions resulted in a nearly 30-fold increase in trans-binding lifetimes between epithelial-cadherin extracellular domains. In turn, the presence of trans interactions increased the lifetime of cis bonds. Importantly, comparison of trans-binding lifetimes of small and large cadherin clusters suggests that this cooperativity is primarily due to allostery. The direct quantitative demonstration of strong mutual cooperativity between cis and trans interactions at intermembrane adhesions provides insights into the long-standing controversy of how weak cis and trans interactions act in concert to create strong macroscopic cell adhesions.
Collapse
Affiliation(s)
- Connor J Thompson
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309
| | - Vinh H Vu
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - Deborah E Leckband
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - Daniel K Schwartz
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, CO 80309;
| |
Collapse
|
8
|
Chandran R, Kale G, Philippe JM, Lecuit T, Mayor S. Distinct actin-dependent nanoscale assemblies underlie the dynamic and hierarchical organization of E-cadherin. Curr Biol 2021; 31:1726-1736.e4. [PMID: 33607036 DOI: 10.1016/j.cub.2021.01.059] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 11/05/2020] [Accepted: 01/18/2021] [Indexed: 02/07/2023]
Abstract
Cadherins are transmembrane adhesion proteins required for the formation of cohesive tissues.1-4 Intracellular interactions of E-cadherin with the Catenin family proteins, α- and β-catenin, facilitate connections with the cortical actomyosin network. This is necessary for maintaining the integrity of cell-cell adhesion in epithelial tissues.5-11 The supra-molecular architecture of E-cadherin is an important feature of its adhesion function; cis and trans interactions of E-cadherin are deployed12-15 to form clusters, both in cis and trans.11,16-21 Studies in Drosophila embryo have also shown that Drosophila E-cadherin (dE-cad) is organized as finite-sized dynamic clusters that localize with actin patches at cell-cell junctions, in continuous exchange with the extra-junctional pool of dE-cad surrounding the clusters.11,19 Here, we use the ectopic expression of dE-cad in larval hemocytes, which lack endogenous dE-cad to recapitulate functional cell-cell junctions in a convenient model system. We find that, while dE-cad at cell-cell junctions in hemocytes exhibits a clustered trans-paired organization similar to that reported previously in embryonic epithelial tissue, extra-junctional dE-cad is also organized as relatively immobile nanoclusters as well as more loosely packed diffusive oligomers. Oligomers are promoted by cis interactions of the ectodomain, and their growth is counteracted by the activity of cortical actomyosin. Oligomers in turn promote assembly of dense nanoclusters that require cortical actomyosin activity. Thus, cortical actin activity remodels oligomers and generates nanoclusters. The requirement for dynamic actin in the organization of dE-cad at the nanoscale may provide a mechanism to dynamically tune junctional strength.
Collapse
Affiliation(s)
- Rumamol Chandran
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bellary Road, Bangalore 560065, India; Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Girish Kale
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bellary Road, Bangalore 560065, India
| | - Jean-Marc Philippe
- Aix Marseille Université & CNRS, IBDM - UMR7288 & Turing Centre for Living Systems, Campus de Luminy Case 907, 13288 Marseille, France
| | - Thomas Lecuit
- Aix Marseille Université & CNRS, IBDM - UMR7288 & Turing Centre for Living Systems, Campus de Luminy Case 907, 13288 Marseille, France; Collège de France, 11 Place Marcelin Berthelot, 75005 Paris, France
| | - Satyajit Mayor
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bellary Road, Bangalore 560065, India.
| |
Collapse
|
9
|
Thompson CJ, Su Z, Vu VH, Wu Y, Leckband DE, Schwartz DK. Cadherin clusters stabilized by a combination of specific and nonspecific cis-interactions. eLife 2020; 9:e59035. [PMID: 32876051 PMCID: PMC7505656 DOI: 10.7554/elife.59035] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022] Open
Abstract
We demonstrate a combined experimental and computational approach for the quantitative characterization of lateral interactions between membrane-associated proteins. In particular, weak, lateral (cis) interactions between E-cadherin extracellular domains tethered to supported lipid bilayers, were studied using a combination of dynamic single-molecule Förster Resonance Energy Transfer (FRET) and kinetic Monte Carlo (kMC) simulations. Cadherins are intercellular adhesion proteins that assemble into clusters at cell-cell contacts through cis- and trans- (adhesive) interactions. A detailed and quantitative understanding of cis-clustering has been hindered by a lack of experimental approaches capable of detecting and quantifying lateral interactions between proteins on membranes. Here single-molecule intermolecular FRET measurements of wild-type E-cadherin and cis-interaction mutants combined with simulations demonstrate that both nonspecific and specific cis-interactions contribute to lateral clustering on lipid bilayers. Moreover, the intermolecular binding and dissociation rate constants are quantitatively and independently determined, demonstrating an approach that is generalizable for other interacting proteins.
Collapse
Affiliation(s)
- Connor J Thompson
- Department of Chemical and Biological Engineering, University of Colorado BoulderBoulderUnited States
| | - Zhaoqian Su
- Department of Systems and Computational Biology, Albert Einstein College of MedicineBronxUnited States
| | - Vinh H Vu
- Department of Biochemistry and University of Illinois, Urbana-ChampaignUrbanaUnited States
| | - Yinghao Wu
- Department of Systems and Computational Biology, Albert Einstein College of MedicineBronxUnited States
| | - Deborah E Leckband
- Department of Biochemistry and University of Illinois, Urbana-ChampaignUrbanaUnited States
- Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana-ChampaignUrbanaUnited States
| | - Daniel K Schwartz
- Department of Chemical and Biological Engineering, University of Colorado BoulderBoulderUnited States
| |
Collapse
|
10
|
Thompson CJ, Vu VH, Leckband DE, Schwartz DK. Cadherin Extracellular Domain Clustering in the Absence of Trans-Interactions. J Phys Chem Lett 2019; 10:4528-4534. [PMID: 31335147 PMCID: PMC6815682 DOI: 10.1021/acs.jpclett.9b01500] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
While both cis and trans (adhesive)-interactions cooperate in the assembly of intercellular adhesions, computational simulations have predicted that two-dimensional confinement may promote cis-oligomerization, in the absence of trans-interactions. Here, single-molecule tracking of cadherin extracellular domains on supported lipid bilayers revealed the density-dependent formation of oligomers and cis-clusters in the absence of trans-interactions. Lateral oligomers were virtually eliminated by mutating a putative cis (lateral) binding interface. At low cadherin surface coverage, wild-type and mutant cadherin diffused rapidly, consistent with the motion of a lipid molecule within a cadherin-free supported bilayer and with cadherins diffusing as monomers. Although the diffusion of mutant cadherin did not change appreciably with increasing surface coverage, the average short-time diffusion coefficient of wild-type cadherin slowed significantly above a fractional surface coverage of ∼0.01 (∼1100 molecules/μm2). A detailed analysis of molecular trajectories suggested the presence of a broad size distribution of cis-cadherin oligomers. These findings verify predictions that two-dimensional confinement promotes cis-oligomerization, in the absence of trans-interactions.
Collapse
Affiliation(s)
- Connor J. Thompson
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder 80309, Colorado, United States
| | - Vinh H. Vu
- Department of Biochemistry, University of Illinois, Urbana–Champaign, Urbana 61801, Illinois, United States
| | - Deborah E. Leckband
- Department of Biochemistry, University of Illinois, Urbana–Champaign, Urbana 61801, Illinois, United States
- Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana–Champaign, Urbana 61801, Illinois, United States
| | - Daniel K. Schwartz
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder 80309, Colorado, United States
| |
Collapse
|
11
|
Chen J, Newhall J, Xie ZR, Leckband D, Wu Y. A Computational Model for Kinetic Studies of Cadherin Binding and Clustering. Biophys J 2017; 111:1507-1518. [PMID: 27705773 DOI: 10.1016/j.bpj.2016.08.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 08/02/2016] [Accepted: 08/30/2016] [Indexed: 12/20/2022] Open
Abstract
Cadherin is a cell-surface transmembrane receptor that mediates calcium-dependent cell-cell adhesion and is a major component of adhesive junctions. The formation of intercellular adhesive junctions is initiated by trans binding between cadherins on adjacent cells, which is followed by the clustering of cadherins via the formation of cis interactions between cadherins on the same cell membranes. Moreover, classical cadherins have multiple glycosylation sites along their extracellular regions. It was found that aberrant glycosylation affects the adhesive function of cadherins and correlates with metastatic phenotypes of several cancers. However, a mechanistic understanding of cadherin clustering during cell adhesion and the role of glycosylation in this process is still lacking. Here, we designed a kinetic model that includes multistep reaction pathways for cadherin clustering. We further applied a diffusion-reaction algorithm to numerically simulate the clustering process using a recently developed coarse-grained model. Using experimentally measured rates of trans binding between soluble E-cadherin extracellular domains, we conducted simulations of cadherin-mediated cell-cell binding kinetics, and the results are quantitatively comparable to experimental data from micropipette experiments. In addition, we show that incorporating cadherin clustering via cis interactions further increases intercellular binding. Interestingly, a two-phase kinetic profile was derived under the assumption that glycosylation regulates the kinetic rates of cis interactions. This two-phase profile is qualitatively consistent with experimental results from micropipette measurements. Therefore, our computational studies provide new, to our knowledge, insights into the molecular mechanism of cadherin-based cell adhesion.
Collapse
Affiliation(s)
- Jiawen Chen
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Jillian Newhall
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Zhong-Ru Xie
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Deborah Leckband
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Yinghao Wu
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
12
|
Qudrat A, Truong K. Engineering Synthetic Proteins to Generate Ca 2+ Signals in Mammalian Cells. ACS Synth Biol 2017; 6:582-590. [PMID: 28301940 DOI: 10.1021/acssynbio.6b00310] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The versatility of Ca2+ signals allows it to regulate diverse cellular processes such as migration, apoptosis, motility and exocytosis. In some receptors (e.g., VEGFR2), Ca2+ signals are generated upon binding their ligand(s) (e.g., VEGF-A). Here, we employed a design strategy to engineer proteins that generate a Ca2+ signal upon binding various extracellular stimuli by creating fusions of protein domains that oligomerize to the transmembrane domain and the cytoplasmic tail of the VEGFR2. To test the strategy, we created chimeric proteins that generate Ca2+ signals upon stimulation with various extracellular stimuli (e.g., rapamycin, EDTA or extracellular free Ca2+). By coupling these chimeric proteins that generate Ca2+ signals with proteins that respond to Ca2+ signals, we rewired, for example, dynamic cellular blebbing to increases in extracellular free Ca2+. Thus, using this design strategy, it is possible to engineer proteins to generate a Ca2+ signal to rewire a wide range of extracellular stimuli to a wide range of Ca2+-activated processes.
Collapse
Affiliation(s)
- Anam Qudrat
- Institute of Biomaterials
and Biomedical Engineering, University of Toronto, 164 College
Street, Toronto, Ontario M5S 3G9, Canada
| | - Kevin Truong
- Institute of Biomaterials
and Biomedical Engineering, University of Toronto, 164 College
Street, Toronto, Ontario M5S 3G9, Canada
- Edward
S. Rogers, Sr. Department of Electrical and Computer Engineering, University of Toronto, 10 King’s College Circle, Toronto, Ontario M5S 3G4, Canada
| |
Collapse
|
13
|
Disruption of cell adhesion by an antibody targeting the cell-adhesive intermediate (X-dimer) of human P-cadherin. Sci Rep 2017; 7:39518. [PMID: 28045038 PMCID: PMC5206748 DOI: 10.1038/srep39518] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 11/24/2016] [Indexed: 12/04/2022] Open
Abstract
Human P-cadherin is a cell adhesion protein of the family of classical cadherins, the overexpression of which is correlated with poor prognosis in various types of cancer. Antibodies inhibiting cell-cell adhesion mediated by P-cadherin show clear therapeutic effect, although the mechanistic basis explaining their effectiveness is still unclear. Based on structural, physicochemical, and functional analyses, we have elucidated the molecular mechanism of disruption of cell adhesion by antibodies targeting human P-cadherin. Herein we have studied three different antibodies, TSP5, TSP7, and TSP11, each recognizing a different epitope on the surface of the cell-adhesive domain (EC1). Although all these three antibodies recognized human P-cadherin with high affinity, only TSP7 disrupted cell adhesion. Notably, we demonstrated that TSP7 abolishes cell adhesion by disabling the so-called X-dimer (a kinetic adhesive intermediate), in addition to disrupting the strand-swap dimer (the final thermodynamic state). The inhibition of the X-dimer was crucial for the overall inhibitory effect, raising the therapeutic value of a kinetic intermediary not only for preventing, but also for reversing, cell adhesion mediated by a member of the classical cadherin family. These findings should help to design more innovative and effective therapeutic solutions targeting human P-cadherin.
Collapse
|
14
|
Jian MY, Liu Y, Li Q, Wolkowicz P, Alexeyev M, Zmijewski J, Creighton J. N-cadherin coordinates AMP kinase-mediated lung vascular repair. Am J Physiol Lung Cell Mol Physiol 2015; 310:L71-85. [PMID: 26545901 DOI: 10.1152/ajplung.00227.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/30/2015] [Indexed: 01/24/2023] Open
Abstract
Injury to the pulmonary circulation compromises endothelial barrier function and increases lung edema. Resolution of lung damage involves restoring barrier integrity, a process requiring reestablishment of endothelial cell-cell adhesions. However, mechanisms underlying repair in lung endothelium are poorly understood. In pulmonary microvascular endothelium, AMP kinase α1 (AMPKα1) stimulation enhances recovery of the endothelial barrier after LPS-induced vascular damage. AMPKα1 colocalizes to a discrete membrane compartment with the adhesion protein neuronal cadherin (N-cadherin). This study sought to determine N-cadherin's role in the repair process. Short-hairpin RNA against full-length N-cadherin or a C-terminally truncated N-cadherin, designed to disrupt the cadherin's interactions with intracellular proteins, were expressed in lung endothelium. Disruption of N-cadherin's intracellular domain caused translocation of AMPK away from the membrane and attenuated AMPK-mediated restoration of barrier function in LPS-treated endothelium. AMPK activity measurements indicated that lower basal AMPK activity in cells expressing the truncated N-cadherin compared with controls. Moreover, the AMPK stimulator 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) failed to increase AMPK activity in cells expressing the modified N-cadherin, indicating uncoupling of a functional association between AMPK and the cadherin. Isolated lung studies confirmed a physiologic role for this pathway in vivo. AMPK activation reversed LPS-induced increase in permeability, whereas N-cadherin inhibition hindered AMPK-mediated repair. Thus N-cadherin coordinates the vascular protective actions of AMPK through a functional link with the kinase. This study provides insight into intrinsic repair mechanisms in the lung and supports AMPK stimulation as a modality for treating vascular disease.
Collapse
Affiliation(s)
- Ming-Yuan Jian
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, Center for Lung Injury and Repair
| | - Yanping Liu
- Division of Endocrinology, Diabetes, and Metabolism
| | - Qian Li
- Division of Pediatric Neonatology, and
| | | | - Mikhail Alexeyev
- Department of Physiology and Cell Biology, Center for Lung Biology, University of South Alabama, Mobile
| | - Jaroslaw Zmijewski
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Judy Creighton
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, Center for Lung Injury and Repair,
| |
Collapse
|
15
|
Chen CS, Hong S, Indra I, Sergeeva AP, Troyanovsky RB, Shapiro L, Honig B, Troyanovsky SM. α-Catenin-mediated cadherin clustering couples cadherin and actin dynamics. J Cell Biol 2015; 210:647-61. [PMID: 26261181 PMCID: PMC4539995 DOI: 10.1083/jcb.201412064] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 07/13/2015] [Indexed: 12/14/2022] Open
Abstract
The function of the actin-binding domain of α-catenin, αABD, including its possible role in the direct anchorage of the cadherin-catenin complex to the actin cytoskeleton, has remained uncertain. We identified two point mutations on the αABD surface that interfere with αABD binding to actin and used them to probe the role of α-catenin-actin interactions in adherens junctions. We found that the junctions directly bound to actin via αABD were more dynamic than the junctions bound to actin indirectly through vinculin and that recombinant αABD interacted with cortical actin but not with actin bundles. This interaction resulted in the formation of numerous short-lived cortex-bound αABD clusters. Our data suggest that αABD clustering drives the continuous assembly of transient, actin-associated cadherin-catenin clusters whose disassembly is maintained by actin depolymerization. It appears then that such actin-dependent αABD clustering is a unique molecular mechanism mediating both integrity and reassembly of the cell-cell adhesive interface formed through weak cis- and trans-intercadherin interactions.
Collapse
Affiliation(s)
- Chi-Shuo Chen
- Department of Dermatology, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Soonjin Hong
- Department of Dermatology, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Indrajyoti Indra
- Department of Dermatology, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Alina P Sergeeva
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, NY 10032 Center for Computational Biology and Bioinformatics, Columbia University Medical Center, New York, NY 10032 Department of Systems Biology, Columbia University, New York, NY 10032
| | - Regina B Troyanovsky
- Department of Dermatology, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Lawrence Shapiro
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, NY 10032 Department of Systems Biology, Columbia University, New York, NY 10032
| | - Barry Honig
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center, New York, NY 10032 Center for Computational Biology and Bioinformatics, Columbia University Medical Center, New York, NY 10032 Department of Systems Biology, Columbia University, New York, NY 10032 Howard Hughes Medical Institute, Chevy Chase, MD 20815
| | - Sergey M Troyanovsky
- Department of Dermatology, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| |
Collapse
|
16
|
Romereim SM, Conoan NH, Chen B, Dudley AT. A dynamic cell adhesion surface regulates tissue architecture in growth plate cartilage. Development 2014; 141:2085-95. [PMID: 24764078 DOI: 10.1242/dev.105452] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The architecture and morphogenetic properties of tissues are founded in the tissue-specific regulation of cell behaviors. In endochondral bones, the growth plate cartilage promotes bone elongation via regulated chondrocyte maturation within an ordered, three-dimensional cell array. A key event in the process that generates this cell array is the transformation of disordered resting chondrocytes into clonal columns of discoid proliferative cells aligned with the primary growth vector. Previous analysis showed that column-forming chondrocytes display planar cell divisions, and the resulting daughter cells rearrange by ∼90° to align with the lengthening column. However, these previous studies provided limited information about the mechanisms underlying this dynamic process. Here we present new mechanistic insights generated by application of a novel time-lapse confocal microscopy method along with immunofluorescence and electron microscopy. We show that, during cell division, daughter chondrocytes establish a cell-cell adhesion surface enriched in cadherins and β-catenin. Rearrangement into columns occurs concomitant with expansion of this adhesion surface in a process more similar to cell spreading than to migration. Column formation requires cell-cell adhesion, as reducing cadherin binding via chelation of extracellular calcium inhibits chondrocyte rearrangement. Importantly, physical indicators of cell polarity, such as cell body alignment, are not prerequisites for oriented cell behavior. Our results support a model in which regulation of adhesive surface dynamics and cortical tension by extrinsic signaling modifies the thermodynamic landscape to promote organization of daughter cells in the context of the three-dimensional growth plate tissue.
Collapse
Affiliation(s)
- Sarah M Romereim
- Department of Genetics, Cell Biology, and Anatomy and the Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, 985965 Nebraska Medical Center, Omaha, NE 68198-5965, USA
| | | | | | | |
Collapse
|
17
|
Indra I, Troyanovsky R, Troyanovsky SM. Afadin controls cadherin cluster stability using clathrin-independent mechanism. Tissue Barriers 2014; 2:e28687. [PMID: 25045601 PMCID: PMC4092309 DOI: 10.4161/tisb.28687] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Revised: 03/23/2014] [Accepted: 03/27/2014] [Indexed: 11/19/2022] Open
Abstract
Afadin is an actin-binding protein that interacts with the intracellular region of the transmembrane proteins, nectins. In collaboration with other transmembrane proteins, cadherins, nectins form adherens junctions, a major type of cell-cell adhesive structures in the multicellular organisms. To elucidate the afadin function, we studied adherens junction defects induced by afadin depletion in epithelial A431 cells. We have found that the cells lacking afadin exhibit no abnormalities in morphology or in general dynamics of adherens junctions in the confluent cell cultures. The only observed difference is a slight increase in the rate of cadherin turnover in these junctions. However, afadin depletion strongly affects the assembly of new adherens junctions immediately after two cells touch one another: initiation of new junctions is significantly delayed, the growth of the nascent junctions stagnates, and their lifetime shortens. As a result, the afadin-depleted cells need much more time to establish the mature junctional structures. This defect is not caused by the clathrin-dependent endocytosis of cadherin clusters that was monitored using live-cell imaging of A431 cells co-expressing GFP-tagged E-cadherin and mCherry-tagged clathrin light chain. Taken together our data show that afadin reinforces adherens junctions and that this process is crucial for the fast formation of adherens junctions at the sites of new cell-cell contacts.
Collapse
Affiliation(s)
- Indrajyoti Indra
- Department of Dermatology; The Feinberg School of Medicine; Chicago, Illinois
| | - Regina Troyanovsky
- Department of Dermatology; The Feinberg School of Medicine; Chicago, Illinois
| | | |
Collapse
|
18
|
Takeda H. Effects of Cd2+ on cis-dimer structure of E-cadherin in living cells. Biochem Biophys Res Commun 2014; 444:467-72. [PMID: 24480437 DOI: 10.1016/j.bbrc.2014.01.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 01/17/2014] [Indexed: 10/25/2022]
Abstract
E-cadherin, a calcium (Ca(2+))-dependent cell-cell adhesion molecule, plays a key role in the maintenance of tissue integrity. We have previously demonstrated that E-cadherin functions in vivo as a cis-dimer through chemical cross-linking reagents. Ca(2+) plays an important role in the cis-dimer formation of cadherin. However, the molecular mechanisms by which Ca(2+) interacts with the binding sites that regulate cis-dimer structures have not been completely elucidated. As expected for a Ca(2+) antagonist, cadmium (Cd(2+)) disrupts cadherin function by displacing Ca(2+) from its binding sites on the cadherin molecules. We used Cd(2+) as a probe for investigating the role of Ca(2+) in the dynamics of the E-cadherin extracellular region that involve cis-dimer formation and adhesion. While cell-cell adhesion assembly was completely disrupted in the presence of Cd(2+), the amount of cis-dimers of E-cadherin that formed at the cell surface was not affected. In our "Cd(2+)-switch" experiments, we did not find that Cd(2+)-induced E-cadherin cis-dimer formation in EL cells when they were incubated in low-Ca(2+) medium. In the present study, we demonstrated for the first time the effects of Cd(2+) on the cis-dimer structure of E-cadherin in living cells using a chemical cross-link analysis.
Collapse
Affiliation(s)
- Hiroshi Takeda
- Department of Tissue Development and Regeneration, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, S-1, W-17, Chuo-ku, Sapporo 060-8556, Japan.
| |
Collapse
|
19
|
Xiaoxue W, Xi C, Zhibo X. Effects of botulinum toxin type A on expression of genes in keloid fibroblasts. Aesthet Surg J 2014; 34:154-9. [PMID: 23709452 DOI: 10.1177/1090820x13482938] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Invasive growth of fibroblast cells, which is regulated by multiple biological factors, is the key event in the pathophysiology of keloid scars. Recent studies have suggested that botulinum toxin type A (BoNT-A) could inhibit invasive growth of keloids. However, the molecular mechanisms are unknown. OBJECTIVE The authors explore the effect of BoNT-A on the expression of genes relevant to invasive growth in keloid fibroblasts. METHODS With 112 genes that were relevant to invasive growth, the authors utilized microarray analysis to study messenger RNA expression profiles in keloid fibroblasts treated with BoNT-A. Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to confirm the microarray results. RESULTS Analyses from microarray and qRT-PCR revealed that the S100A4 gene was upregulated and that the TGF-β1, VEGF, MMP-1, and PDGFA genes were downregulated in fibroblasts treated with BoNT-A. CONCLUSIONS The BoNT-A altered expression levels of S100A4, TGF-β1, VEGF, MMP-1, and PDGFA genes in keloid fibroblasts provide a useful clue for exploring the function of BoNT-A and finding a novel treatment for keloid scarring.
Collapse
Affiliation(s)
- Wang Xiaoxue
- Second Affiliated Hospital of the Harbin Medical University, Harbin City, China
| | | | | |
Collapse
|
20
|
Truong Quang BA, Mani M, Markova O, Lecuit T, Lenne PF. Principles of E-cadherin supramolecular organization in vivo. Curr Biol 2013; 23:2197-2207. [PMID: 24184100 DOI: 10.1016/j.cub.2013.09.015] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 07/29/2013] [Accepted: 09/05/2013] [Indexed: 12/16/2022]
Abstract
BACKGROUND E-cadherin plays a pivotal role in tissue morphogenesis by forming clusters that support intercellular adhesion and transmit tension. What controls E-cadherin mesoscopic organization in clusters is unclear. RESULTS We use 3D superresolution quantitative microscopy in Drosophila embryos to characterize the size distribution of E-cadherin nanometric clusters. The cluster size follows power-law distributions over three orders of magnitude with exponential decay at large cluster sizes. By exploring the predictions of a general theoretical framework including cluster fusion and fission events and recycling of E-cadherin, we identify two distinct active mechanisms setting the cluster-size distribution. Dynamin-dependent endocytosis targets large clusters only, thereby imposing a cutoff size. Moreover, interactions between E-cadherin clusters and actin filaments control the fission in a size-dependent manner. CONCLUSIONS E-cadherin clustering depends on key cortical regulators, which provide tunable and local control over E-cadherin organization. Our data provide the foundation for a quantitative understanding of how E-cadherin distribution affects adhesion and might regulate force transmission in vivo.
Collapse
Affiliation(s)
- Binh-An Truong Quang
- Developmental Biology Institute of Marseilles, UMR 7288 CNRS, Aix-Marseille Université, 13288 Marseille Cedex 9, France
| | - Madhav Mani
- Kavli Institute of Theoretical Physics, Santa Barbara, CA 93101, USA; University of California, Department of Physics, Santa Barbara, CA 93101, USA
| | - Olga Markova
- Developmental Biology Institute of Marseilles, UMR 7288 CNRS, Aix-Marseille Université, 13288 Marseille Cedex 9, France
| | - Thomas Lecuit
- Developmental Biology Institute of Marseilles, UMR 7288 CNRS, Aix-Marseille Université, 13288 Marseille Cedex 9, France
| | - Pierre-François Lenne
- Developmental Biology Institute of Marseilles, UMR 7288 CNRS, Aix-Marseille Université, 13288 Marseille Cedex 9, France.
| |
Collapse
|
21
|
Ritterson RS, Kuchenbecker KM, Michalik M, Kortemme T. Design of a photoswitchable cadherin. J Am Chem Soc 2013; 135:12516-9. [PMID: 23923816 PMCID: PMC3774674 DOI: 10.1021/ja404992r] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Indexed: 01/31/2023]
Abstract
There is a growing interest in engineering proteins whose function can be controlled with the spatial and temporal precision of light. Here, we present a novel example of a functional light-triggered switch in the Ca-dependent cell-cell adhesion protein E-cadherin, created using a mechanism-based design strategy. We report an 18-fold change in apparent Ca(2+) binding affinity upon illumination. Our results include a detailed examination of functional switching via linked changes in Ca(2+) binding and cadherin dimerization. This design opens avenues toward controllable tools that could be applied to many long-standing questions about cadherin's biological function in cell-cell adhesion and downstream signaling.
Collapse
Affiliation(s)
- Ryan S Ritterson
- Graduate Group in Biophysics, University of California, San Francisco, San Francisco, California 94158, USA.
| | | | | | | |
Collapse
|
22
|
Hong S, Troyanovsky RB, Troyanovsky SM. Binding to F-actin guides cadherin cluster assembly, stability, and movement. ACTA ACUST UNITED AC 2013; 201:131-43. [PMID: 23547031 PMCID: PMC3613698 DOI: 10.1083/jcb.201211054] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Binding of cadherin to F-actin cooperates with the cadherin cis-interface to stabilize cadherin adhesion clusters and is required for their directional movement. The cadherin extracellular region produces intercellular adhesion clusters through trans- and cis-intercadherin bonds, and the intracellular region connects these clusters to the cytoskeleton. To elucidate the interdependence of these binding events, cadherin adhesion was reconstructed from the minimal number of structural elements. F-actin–uncoupled adhesive clusters displayed high instability and random motion. Their assembly required a cadherin cis-binding interface. Coupling these clusters with F-actin through an α-catenin actin-binding domain (αABD) dramatically extended cluster lifetime and conferred direction to cluster motility. In addition, αABD partially lifted the requirement for the cis-interface for cluster assembly. Even more dramatic enhancement of cadherin clustering was observed if αABD was joined with cadherin through a flexible linker or if it was replaced with an actin-binding domain of utrophin. These data present direct evidence that binding to F-actin stabilizes cadherin clusters and cooperates with the cis-interface in cadherin clustering. Such cooperation apparently synchronizes extracellular and intracellular binding events in the process of adherens junction assembly.
Collapse
Affiliation(s)
- Soonjin Hong
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | | |
Collapse
|
23
|
Zhang Y, Jiang N, Zarnitsyna VI, Klopocki AG, McEver RP, Zhu C. P-selectin glycoprotein ligand-1 forms dimeric interactions with E-selectin but monomeric interactions with L-selectin on cell surfaces. PLoS One 2013; 8:e57202. [PMID: 23451187 PMCID: PMC3581448 DOI: 10.1371/journal.pone.0057202] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 01/18/2013] [Indexed: 11/19/2022] Open
Abstract
Interactions of selectins with cell surface glycoconjugates mediate the first step of the adhesion and signaling cascade that recruits circulating leukocytes to sites of infection or injury. P-selectin dimerizes on the surface of endothelial cells and forms dimeric bonds with P-selectin glycoprotein ligand-1 (PSGL-1), a homodimeric sialomucin on leukocytes. It is not known whether leukocyte L-selectin or endothelial cell E-selectin are monomeric or oligomeric. Here we used the micropipette technique to analyze two-dimensional binding of monomeric or dimeric L- and E-selectin with monomeric or dimeric PSGL-1. Adhesion frequency analysis demonstrated that E-selectin on human aortic endothelial cells supported dimeric interactions with dimeric PSGL-1 and monomeric interactions with monomeric PSGL-1. In contrast, L-selectin on human neutrophils supported monomeric interactions with dimeric or monomeric PSGL-1. Our work provides a new method to analyze oligomeric cross-junctional molecular binding at the interface of two interacting cells.
Collapse
Affiliation(s)
- Yan Zhang
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | | | | | | | | | | |
Collapse
|
24
|
Abstract
Members of the p120-catenin family associate with cadherins and regulate their stability at the plasma membrane. How p120-catenin limits cadherin endocytosis has long remained a mystery. In this issue, Nanes et al. (2012. J. Cell Biol. doi:10.1083/jcb.201205029) identify a conserved acidic motif within cadherins that acts as a physical platform for p120-catenin binding. However, in the absence of p120-catenin, the motif acts as an endocytic signal. These results provide new insight into p120-catenin's role as guardian of intercellular junction dynamics.
Collapse
Affiliation(s)
- Mirna Perez-Moreno
- BBVA Foundation-Cancer Cell Biology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid 28019, Spain.
| | | |
Collapse
|
25
|
Microenvironmental protection of CML stem and progenitor cells from tyrosine kinase inhibitors through N-cadherin and Wnt-β-catenin signaling. Blood 2013; 121:1824-38. [PMID: 23299311 DOI: 10.1182/blood-2012-02-412890] [Citation(s) in RCA: 214] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are highly effective in treatment of chronic myeloid leukemia (CML) but do not eliminate leukemia stem cells (LSCs), which remain a potential source of relapse. TKI treatment effectively inhibits BCR-ABL kinase activity in CML LSCs, suggesting that additional kinase-independent mechanisms contribute to LSC preservation. We investigated whether signals from the bone marrow (BM) microenvironment protect CML LSCs from TKI treatment. Coculture with human BM mesenchymal stromal cells (MSCs) significantly inhibited apoptosis and preserved CML stem/progenitor cells following TKI exposure, maintaining colony-forming ability and engraftment potential in immunodeficient mice. We found that the N-cadherin receptor plays an important role in MSC-mediated protection of CML progenitors from TKI. N-cadherin-mediated adhesion to MSCs was associated with increased cytoplasmic N-cadherin-β-catenin complex formation as well as enhanced β-catenin nuclear translocation and transcriptional activity. Increased exogenous Wnt-mediated β-catenin signaling played an important role in MSC-mediated protection of CML progenitors from TKI treatment. Our results reveal a close interplay between N-cadherin and the Wnt-β-catenin pathway in protecting CML LSCs during TKI treatment. Importantly, these results reveal novel mechanisms of resistance of CML LSCs to TKI treatment and suggest new targets for treatment designed to eradicate residual LSCs in CML patients.
Collapse
|
26
|
Wang Q, Lin JLC, Chan SY, Lin JJC. The Xin repeat-containing protein, mXinβ, initiates the maturation of the intercalated discs during postnatal heart development. Dev Biol 2012; 374:264-80. [PMID: 23261932 DOI: 10.1016/j.ydbio.2012.12.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 11/25/2012] [Accepted: 12/04/2012] [Indexed: 10/27/2022]
Abstract
The intercalated disc (ICD) is a unique structure to the heart and plays vital roles in communication and signaling among cardiomyocytes. ICDs are formed and matured during postnatal development through a profound redistribution of the intercellular junctions, as well as recruitment and assembly of more than 200 proteins at the termini of cardiomyocytes. The molecular mechanism underlying this process is not completely understood. The mouse orthologs (mXinα and mXinβ) of human cardiomyopathy-associated (CMYA)/Xin actin-binding repeat-containing protein (XIRP) genes (CMYA1/XIRP1 and CMYA3/XIRP2, respectively) encode proteins localized to ICDs. Ablation of mXinα results in adult late-onset cardiomyopathy with conduction defects and up-regulation of mXinβ. ICD structural defects are found in adult but not juvenile mXinα-null hearts. On the other hand, loss of mXinβ leads to ICD defects at postnatal day 16.5, a developmental stage when the heart is forming ICDs, suggesting mXinβ is required for ICD formation. Using quantitative Western blot, we showed in this study that mXinβ but not mXinα was uniquely up-regulated during the redistribution of intercellular junction from the lateral membrane of cardiomyocytes to their termini. In the absence of mXinβ, the intercellular junctions failed to be restricted to the termini of the cells, and the onset of such defect correlated with the peak expression of mXinβ. Immunofluorescence staining and subcellular fractionation showed that mXinβ preferentially associated with the forming ICDs, further suggesting that mXinβ functioned locally to promote ICD maturation. In contrast, the spatiotemporal expression profile of mXinα and the lack of more severe ICD defects in mXinα-/-;mXinβ-/- double knockout hearts than in mXinβ-/- hearts suggested that mXinα was not essential for the postnatal formation of ICDs. A two-step model for the development of ICD is proposed where mXinβ is essential for the redistribution of intercellular junction components from the lateral puncta to the cell termini.
Collapse
Affiliation(s)
- Qinchuan Wang
- Department of Biology, University of Iowa, Iowa City, IA 52242-1324, USA
| | | | | | | |
Collapse
|
27
|
Harrison OJ, Vendome J, Brasch J, Jin X, Hong S, Katsamba PS, Ahlsen G, Troyanovsky RB, Troyanovsky SM, Honig B, Shapiro L. Nectin ectodomain structures reveal a canonical adhesive interface. Nat Struct Mol Biol 2012; 19:906-15. [PMID: 22902367 PMCID: PMC3443293 DOI: 10.1038/nsmb.2366] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 07/19/2012] [Indexed: 11/21/2022]
Abstract
Nectins are immunoglobulin superfamily glycoproteins that mediate intercellular adhesion in many vertebrate tissues. Homophilic and heterophilic interactions between nectin family members help to mediate tissue patterning. We determined homophilic binding affinities and heterophilic specificities of all four nectins and the related protein nectin-like 5 from human and mouse, revealing a range of homophilic strengths and a defined heterophilic specificity pattern. To understand the molecular basis of adhesion and specificity, we determined crystal structures of natively glycosylated full ectodomains or adhesive fragments of nectins 1–4 and nectin-like 5. All crystal structures reveal dimeric nectins bound through a stereotyped interface previously proposed to represent a cis dimer. However, conservation of this interface and results of targeted cross-linking experiments show that this dimer likely represents the adhesive trans interaction. Its structure provides a simple molecular explanation for the adhesive binding specificity of nectins.
Collapse
Affiliation(s)
- Oliver J Harrison
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Boscher C, Zheng YZ, Lakshminarayan R, Johannes L, Dennis JW, Foster LJ, Nabi IR. Galectin-3 protein regulates mobility of N-cadherin and GM1 ganglioside at cell-cell junctions of mammary carcinoma cells. J Biol Chem 2012; 287:32940-52. [PMID: 22846995 DOI: 10.1074/jbc.m112.353334] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Galectin-3 binding to cell surface glycoproteins, including branched N-glycans generated by N-acetylglucosaminyltransferase V (Mgat5) activity, forms a multivalent, heterogeneous, and dynamic lattice. This lattice has been shown to regulate integrin and receptor tyrosine kinase signaling promoting tumor cell migration. N-cadherin is a homotypic cell-cell adhesion receptor commonly overexpressed in tumor cells that contributes to cell motility. Here we show that galectin-3 and N-cadherin interact and colocalize with the lipid raft marker GM1 ganglioside in cell-cell junctions of mammary epithelial cancer cells. Disruption of the lattice by deletion of Mgat5, siRNA depletion of galectin-3, or competitive inhibition with lactose stabilizes cell-cell junctions. It also reduces, in a p120-catenin-dependent manner, the dynamic pool of junctional N-cadherin. Proteomic analysis of detergent-resistant membranes (DRMs) revealed that the galectin lattice opposes entry of many proteins into DRM rafts. N-cadherin and catenins are present in DRMs; however, their DRM distribution is not significantly affected by lattice disruption. Galectin lattice integrity increases the mobile fraction of the raft marker, GM1 ganglioside binding cholera toxin B subunit Ctb, at cell-cell contacts in a p120-catenin-independent manner, but does not affect the mobility of either Ctb-labeled GM1 or GFP-coupled N-cadherin in nonjunctional regions. Our results suggest that the galectin lattice independently enhances lateral molecular diffusion by direct interaction with specific glycoconjugates within the adherens junction. By promoting exchange between raft and non-raft microdomains as well as molecular dynamics within junction-specific raft microdomains, the lattice may enhance turnover of N-cadherin and other glycoconjugates that determine junctional stability and rates of cell migration.
Collapse
Affiliation(s)
- Cécile Boscher
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | | | | | | | | | | | | |
Collapse
|
29
|
Hartsock A, Nelson WJ. Competitive regulation of E-cadherin juxtamembrane domain degradation by p120-catenin binding and Hakai-mediated ubiquitination. PLoS One 2012; 7:e37476. [PMID: 22693575 PMCID: PMC3365061 DOI: 10.1371/journal.pone.0037476] [Citation(s) in RCA: 38] [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: 02/20/2012] [Accepted: 04/23/2012] [Indexed: 12/31/2022] Open
Abstract
p120-Catenin binding to, and Hakai-mediated ubiquitination of the E-cadherin juxtamembrane domain (JMD) are thought to be involved in regulating E-cadherin internalization and degradation. However, the relationship between these two pathways is not understood. We targeted the E-cadherin JMD to mitochondria (WT-JMD) to isolate this domain from the plasma membrane and internalization, and to examine protein modifications and degradation. WT-JMD localized to mitochondria, but did not accumulate there except when proteasome activity was inhibited. We found WT-JMD was ubiquitinated, and arginine substitution of lysines at position 5 (K5R) and 83 (K83R) resulted in the stable accumulation of mutant JMD at mitochondria. p120-Catenin did not localize, or bind to WT-JMD even upon proteasome inhibition, whereas the K5,83R-JMD mutant bound and localized p120-catenin to mitochondria. Mutation of the p120-catenin binding site in combination with these lysine mutations inhibited p120-catenin binding, but did not decrease JMD stability or its accumulation at mitochondria. Thus, increased stability of JMD lysine mutants was due to inhibition of ubiquitination and not to p120-catenin binding. Finally, mutation of these critical lysines in full length E-cadherin had similar effects on protein stability as WT-JMD. Our results indicate that ubiquitination of the JMD inhibits p120-catenin binding, and targets E-cadherin for degradation.
Collapse
Affiliation(s)
- Andrea Hartsock
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California, United States of America
| | - W. James Nelson
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California, United States of America
- Department of Biology, Stanford University, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
30
|
Discovery and development of N-cadherin antagonists. Cell Tissue Res 2012; 348:309-13. [DOI: 10.1007/s00441-011-1320-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 12/21/2011] [Indexed: 10/14/2022]
|
31
|
Abstract
Classical cadherins are a family of transmembrane proteins that mediate cell-cell adhesion at adherens junctions. A complex chain of cis- and trans- interactions between cadherin ectodomains establishes a cadherin adhesive cluster. A principal adhesive interaction in such clusters is an exchange of β strands between the first extracellular cadherin domains (EC1). The structure of cadherin adhesive clusters can be modified by other adherens junction proteins including additional transmembrane proteins, nectins and various intracellular proteins that directly or indirectly interact with the intracellular cadherin region. These interactions determine the dynamics and stability of cadherin adhesive structures.
Collapse
Affiliation(s)
- Sergey Troyanovsky
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, 60611, Chicago, IL, USA,
| |
Collapse
|
32
|
New Insights into the Regulation of E-cadherin Distribution by Endocytosis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 295:63-108. [DOI: 10.1016/b978-0-12-394306-4.00008-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
33
|
Troyanovsky RB, Klingelhöfer J, Troyanovsky SM. α-Catenin contributes to the strength of E-cadherin-p120 interactions. Mol Biol Cell 2011; 22:4247-55. [PMID: 21937720 PMCID: PMC3216651 DOI: 10.1091/mbc.e11-03-0250] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Cadherin–catenin interactions play an important role in cadherin adhesion. In the cadherin complex, α-catenin contributes to the binding strength of another catenin, p120, to the same complex. The data suggest that α-catenin–p120 contact within the cadherin–catenin complex can regulate cadherin trafficking. Cadherin–catenin interactions play an important role in cadherin-mediated adhesion. Here we present strong evidence that in the cadherin–catenin complex α-catenin contributes to the binding strength of another catenin, p120, to the same complex. Specifically, we found that a β-catenin–uncoupled cadherin mutant interacts much more weakly with p120 than its full-size counterpart and that it is rapidly endocytosed from the surface of A-431 cells. We also showed that p120 overexpression stabilizes this mutant on the cell surface. Examination of the α-catenin–deficient MDA-MB-468 cells and their derivates in which α-catenin was reintroduced showed that α-catenin reinforces E-cadherin–p120 association. Finally, a cross-linking analysis of the cadherin–catenin complex indicated that a large loop located in the middle of the p120 arm-repeat domain is in close spatial vicinity to the amino-terminal VH1 domain of α-catenin. The six amino acid–long extension of this loop, caused by an alternative splicing, weakens p120 binding to cadherin. The data suggest that α-catenin–p120 contact within the cadherin–catenin complex can regulate cadherin trafficking.
Collapse
Affiliation(s)
- Regina B Troyanovsky
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | | |
Collapse
|
34
|
Fogel AI, Stagi M, Perez de Arce K, Biederer T. Lateral assembly of the immunoglobulin protein SynCAM 1 controls its adhesive function and instructs synapse formation. EMBO J 2011; 30:4728-38. [PMID: 21926970 DOI: 10.1038/emboj.2011.336] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Accepted: 08/18/2011] [Indexed: 11/09/2022] Open
Abstract
Synapses are specialized adhesion sites between neurons that are connected by protein complexes spanning the synaptic cleft. These trans-synaptic interactions can organize synapse formation, but their macromolecular properties and effects on synaptic morphology remain incompletely understood. Here, we demonstrate that the synaptic cell adhesion molecule SynCAM 1 self-assembles laterally via its extracellular, membrane-proximal immunoglobulin (Ig) domains 2 and 3. This cis oligomerization generates SynCAM oligomers with increased adhesive capacity and instructs the interactions of this molecule across the nascent and mature synaptic cleft. In immature neurons, cis assembly promotes the adhesive clustering of SynCAM 1 at new axo-dendritic contacts. Interfering with the lateral self-assembly of SynCAM 1 in differentiating neurons strongly impairs its synaptogenic activity. At later stages, the lateral oligomerization of SynCAM 1 restricts synaptic size, indicating that this adhesion molecule contributes to the structural organization of synapses. These results support that lateral interactions assemble SynCAM complexes within the synaptic cleft to promote synapse induction and modulate their structure. These findings provide novel insights into synapse development and the adhesive mechanisms of Ig superfamily members.
Collapse
Affiliation(s)
- Adam I Fogel
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | | | | | | |
Collapse
|
35
|
Calcium-dependent dynamics of cadherin interactions at cell-cell junctions. Proc Natl Acad Sci U S A 2011; 108:9857-62. [PMID: 21613566 DOI: 10.1073/pnas.1019003108] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cadherins play a key role in the dynamics of cell-cell contact formation and remodeling of junctions and tissues. Cadherin-cadherin interactions are gated by extracellular Ca(2+), which serves to rigidify the cadherin extracellular domains and promote trans junctional interactions. Here we describe the direct visualization and quantification of spatiotemporal dynamics of N-cadherin interactions across intercellular junctions in living cells using a genetically encodable FRET reporter system. Direct measurements of transjunctional cadherin interactions revealed a sudden, but partial, loss of homophilic interactions (τ = 1.17 ± 0.06 s(-1)) upon chelation of extracellular Ca(2+). A cadherin mutant with reduced adhesive activity (W2A) exhibited a faster, more substantial loss of homophilic interactions (τ = 0.86 ± 0.02 s(-1)), suggesting two types of native cadherin interactions--one that is rapidly modulated by changes in extracellular Ca(2+) and another with relatively stable adhesive activity that is Ca(2+) independent. The Ca(2+)-sensitive dynamics of cadherin interactions were transmitted to the cell interior where β-catenin translocated to N-cadherin at the junction in both cells. These data indicate that cadherins can rapidly convey dynamic information about the extracellular environment to both cells that comprise a junction.
Collapse
|
36
|
Hong S, Troyanovsky RB, Troyanovsky SM. Cadherin exits the junction by switching its adhesive bond. ACTA ACUST UNITED AC 2011; 192:1073-83. [PMID: 21422232 PMCID: PMC3063135 DOI: 10.1083/jcb.201006113] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Intercellular traction forces or lateral alignment of cadherin molecules can influence adherens junction dynamics by altering the cadherin dimerization interface. The plasticity of cell–cell adhesive structures is crucial to all normal and pathological morphogenetic processes. The molecular principles of this plasticity remain unknown. Here we study the roles of two dimerization interfaces, the so-called strand-swap and X dimer interfaces of E-cadherin, in the dynamic remodeling of adherens junctions using photoactivation, calcium switch, and coimmunoprecipitation assays. We show that the targeted inactivation of the X dimer interface blocks the turnover of catenin-uncoupled cadherin mutants in the junctions of A-431 cells. In contrast, the junctions formed by strand-swap dimer interface mutants exhibit high instability. Collectively, our data demonstrate that the strand-swap interaction is a principal cadherin adhesive bond that keeps cells in firm contact. However, to leave the adherens junction, cadherin reconfigures its adhesive bond from the strand swap to the X dimer type. Such a structural transition, controlled by intercellular traction forces or by lateral cadherin alignment, may be the key event regulating adherens junction dynamics.
Collapse
Affiliation(s)
- Soonjin Hong
- Department of Dermatology, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | | |
Collapse
|
37
|
Vunnam N, Flint J, Balbo A, Schuck P, Pedigo S. Dimeric states of neural- and epithelial-cadherins are distinguished by the rate of disassembly. Biochemistry 2011; 50:2951-61. [PMID: 21375242 PMCID: PMC3471160 DOI: 10.1021/bi2001246] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Epithelial- and neural-cadherins are specifically localized at synapses in neurons which can change the shape and contact surface on a time scale of seconds to months. We have focused our studies on the role of the extracellular domains of cadherins in the dynamics of synapses. The kinetics of dimer disassembly of the first two extracellular domains of E- and N-cadherin, ECAD12 and NCAD12, were studied with analytical size exclusion chromatography and sedimentation velocity. NCAD12 forms three different dimers that are distinguished by assembly conditions and kinetics of dissociation. ECAD12 dimer disassembles rapidly regardless of the calcium concentration, whereas the disassembly of NCAD12 dimers was strongly dependent on calcium concentration. In addition to the apo- and saturated-dimeric forms of NCAD12, there is a third dimeric form that is a slow exchange dimer. This third dimeric form for NCAD12, formed by decalcification of the calcium-saturated dimer, was kinetically trapped in apo-conditions and did not disassemble over a period of months. Sedimentation velocity experiments showed that this dimer, upon addition of calcium, had similar weighted averages as a calcium-saturated dimer. These studies provide evidence that the kinetics of dimer disassembly of the extracellular domains may be a major contributor to the morphological dynamics of synapses in vivo.
Collapse
Affiliation(s)
- Nagamani Vunnam
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS 38677
| | - Jon Flint
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS 38677
| | - Andrea Balbo
- Dynamics of Macromolecular Assembly, LBPS, NIBIB, National Institutes of Health, Bethesda, MD 20892
| | - Peter Schuck
- Dynamics of Macromolecular Assembly, LBPS, NIBIB, National Institutes of Health, Bethesda, MD 20892
| | - Susan Pedigo
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS 38677
| |
Collapse
|
38
|
De Felice B, Garbi C, Wilson RR, Santoriello M, Nacca M. Effect of selenocystine on gene expression profiles in human keloid fibroblasts. Genomics 2011; 97:265-76. [PMID: 21371550 DOI: 10.1016/j.ygeno.2011.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 01/19/2011] [Accepted: 02/24/2011] [Indexed: 01/09/2023]
Abstract
In this study, selenocystine, a nutritionally available selenoamino acid, was identified for the first time as a novel agent with anti proliferative activity on human keloids. The 20 μM concentration after 48 h treatment used here was the most effective to reduce keloid fibroblast growth. We analyzed the gene expression profile of selenocystine treatment response in keloid fibroblasts by the microarray system to characterize the effects of selenocystine on human keloids. The major alterations in keloid fibroblasts following selenocystine exposure included up-regulation of the genes encoding cell death and transcription factors. Prominent down-regulation of genes involved in development, cell adhesion and cytoskeleton, as well as extra cellular matrix genes, usually strongly up-regulated in keloids, resulted following selenocystine exposure. The range of the down-regulated genes and the degree of the decreased expression appeared to be correlated with the degree of the morphological alterations in selenocystine treated keloids.
Collapse
Affiliation(s)
- Bruna De Felice
- Department of Life Sciences, University of Naples II, Via Vivaldi 43, 81100 Caserta, Italy.
| | | | | | | | | |
Collapse
|
39
|
Harrison OJ, Jin X, Hong S, Bahna F, Ahlsen G, Brasch J, Wu Y, Vendome J, Felsovalyi K, Hampton CM, Troyanovsky RB, Ben-Shaul A, Frank J, Troyanovsky SM, Shapiro L, Honig B. The extracellular architecture of adherens junctions revealed by crystal structures of type I cadherins. Structure 2011; 19:244-56. [PMID: 21300292 PMCID: PMC3070544 DOI: 10.1016/j.str.2010.11.016] [Citation(s) in RCA: 306] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 11/02/2010] [Accepted: 11/03/2010] [Indexed: 11/24/2022]
Abstract
Adherens junctions, which play a central role in intercellular adhesion, comprise clusters of type I classical cadherins that bind via extracellular domains extended from opposing cell surfaces. We show that a molecular layer seen in crystal structures of E- and N-cadherin ectodomains reported here and in a previous C-cadherin structure corresponds to the extracellular architecture of adherens junctions. In all three ectodomain crystals, cadherins dimerize through a trans adhesive interface and are connected by a second, cis, interface. Assemblies formed by E-cadherin ectodomains coated on liposomes also appear to adopt this structure. Fluorescent imaging of junctions formed from wild-type and mutant E-cadherins in cultured cells confirm conclusions derived from structural evidence. Mutations that interfere with the trans interface ablate adhesion, whereas cis interface mutations disrupt stable junction formation. Our observations are consistent with a model for junction assembly involving strong trans and weak cis interactions localized in the ectodomain.
Collapse
Affiliation(s)
- Oliver J. Harrison
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
- Howard Hughes Medical Institute, Columbia University, New York, NY, USA
| | - Xiangshu Jin
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
- Howard Hughes Medical Institute, Columbia University, New York, NY, USA
| | - Soonjin Hong
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL, USA
| | - Fabiana Bahna
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
- Howard Hughes Medical Institute, Columbia University, New York, NY, USA
| | - Goran Ahlsen
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Julia Brasch
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Yinghao Wu
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
- Howard Hughes Medical Institute, Columbia University, New York, NY, USA
- Center for Computational Biology and Bioinformatics, Columbia University, New York, NY, USA
| | - Jeremie Vendome
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
- Howard Hughes Medical Institute, Columbia University, New York, NY, USA
- Center for Computational Biology and Bioinformatics, Columbia University, New York, NY, USA
| | - Klara Felsovalyi
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
- Howard Hughes Medical Institute, Columbia University, New York, NY, USA
- Center for Computational Biology and Bioinformatics, Columbia University, New York, NY, USA
| | - Cheri M. Hampton
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Regina B. Troyanovsky
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL, USA
| | - Avinoam Ben-Shaul
- Department of Physical Chemistry and the Fritz Haber Research Center, The Hebrew University, Jerusalem, Israel
| | - Joachim Frank
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
- Howard Hughes Medical Institute, Columbia University, New York, NY, USA
| | - Sergey M. Troyanovsky
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL, USA
| | - Lawrence Shapiro
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
- Edward S. Harkness Eye Institute, Columbia University, New York, NY, USA
| | - Barry Honig
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
- Howard Hughes Medical Institute, Columbia University, New York, NY, USA
- Center for Computational Biology and Bioinformatics, Columbia University, New York, NY, USA
| |
Collapse
|
40
|
McCrea PD, Gu D, Balda MS. Junctional music that the nucleus hears: cell-cell contact signaling and the modulation of gene activity. Cold Spring Harb Perspect Biol 2010; 1:a002923. [PMID: 20066098 DOI: 10.1101/cshperspect.a002923] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cell-cell junctions continue to capture the interest of cell and developmental biologists, with an emerging area being the molecular means by which junctional signals relate to gene activity in the nucleus. Although complexities often arise in determining the direct versus indirect nature of such signal transduction, it is clear that such pathways are essential for the function of tissues and that alterations may contribute to many pathological outcomes. This review assesses a variety of cell-cell junction-to-nuclear signaling pathways, and outlines interesting areas for further study.
Collapse
Affiliation(s)
- Pierre D McCrea
- Department of Biochemistry and Molecular Biology, University of Texas MD Anderson Cancer Center, Program in Genes and Development, University of Texas Graduate School of Biomedical Sciences, Houston, Texas 77030, USA.
| | | | | |
Collapse
|
41
|
Green KJ, Getsios S, Troyanovsky S, Godsel LM. Intercellular junction assembly, dynamics, and homeostasis. Cold Spring Harb Perspect Biol 2010; 2:a000125. [PMID: 20182611 DOI: 10.1101/cshperspect.a000125] [Citation(s) in RCA: 214] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Intercellular anchoring junctions are highly specialized regions of the plasma membrane where members of the cadherin family of transmembrane adhesion molecules on opposing cells interact through their extracellular domains, and through their cytoplasmic domains serve as a platform for organizing cytoskeletal anchors and remodelers. Here we focus on assembly of so-called "anchoring" or "adhering" junctions-adherens junctions (AJs) and desmosomes (DSMs), which associate with actin and intermediate filaments, respectively. We will examine how the assembly and function of AJs and DSMs are intimately connected during embryogenesis and in adult cells and tissues, and in some cases even form specialized "mixed" junctions. We will explore signaling and trafficking machineries that drive assembly and remodeling and how these mechanisms are co-opted in human disease.
Collapse
Affiliation(s)
- Kathleen J Green
- Northwestern University Feinberg School of Medicine, Department of Pathology, R.H. Lurie Comprehensive Cancer Center, 303 E. Chicago Ave. Chicago, Illinois 60611, USA.
| | | | | | | |
Collapse
|
42
|
Cooperativity between trans and cis interactions in cadherin-mediated junction formation. Proc Natl Acad Sci U S A 2010; 107:17592-7. [PMID: 20876147 DOI: 10.1073/pnas.1011247107] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Intercellullar junctions formed by cadherins, including desmosomes and adherens junctions, comprise two dimensional arrays of "trans" dimers formed between monomers emanating from opposing cell surfaces. Lateral "cis" interfaces between cadherins from the same cell surface have been proposed to play a role in cadherin clustering. Although the molecular details of cis interactions remain uncertain, they must define an anisotropic arrangement where binding is favorable only in certain orientations. Here we report Monte Carlo simulations performed on a 2D lattice constructed to account for the anisotropy in cadherin cis interactions. A crucial finding is that the "phase transition" between freely diffusing cadherin monomers and dimers and a condensed ordered 2D junction formed by dimers alone is a cooperative process involving both trans and cis interactions. Moreover, cis interactions, despite being too weak to be measured in solution, are critical to the formation of an ordered junction structure. We discuss these results in light of available experimental information on cadherin binding free energies that are transformed from their bulk solution values to interaction energies on a 2D lattice.
Collapse
|
43
|
|
44
|
Spontaneous assembly and active disassembly balance adherens junction homeostasis. Proc Natl Acad Sci U S A 2010; 107:3528-33. [PMID: 20133579 DOI: 10.1073/pnas.0911027107] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The homeostasis of adherens junctions was studied using E-cadherin and its two mutants tagged by the photoconvertible protein Dendra2 in epithelial A-431 cells and in CHO cells lacking endogenous cadherin. The first mutant contained point mutations of two elements, Lys738 and the dileucine motif that suppressed cadherin endocytosis. The second mutant contained, in addition, an extensive truncation that uncoupled the mutant from beta-catenin and p120. Surprisingly, the intact cadherin and its truncated mutant were recruited into the junctions with identical kinetics. The full-size cadherin was actively removed from the junctions by a process that was unaffected by the inactivation of its endocytic elements. The cadherin's apparent half-residence time in the junction was about 2 min. Cadherin clusters made of the truncated mutant exhibited much slower but ATP-independent junctional turnover. Taken together, our experiments showed that adherens junction homeostasis consists of three distinctive steps: cadherin spontaneous recruitment, its lateral catenin-dependent association, and its active release from the resulting clusters. The latter process, whose mechanism is not clear, may play an important role in various kinds of normal and abnormal morphogenesis.
Collapse
|
45
|
Blaschuk OW, Devemy E. Cadherins as novel targets for anti-cancer therapy. Eur J Pharmacol 2009; 625:195-8. [PMID: 19836380 DOI: 10.1016/j.ejphar.2009.05.033] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Revised: 05/06/2009] [Accepted: 05/18/2009] [Indexed: 12/12/2022]
Abstract
The cell adhesion molecules N-, VE- and OB-cadherin have been implicated as regulators of tumor growth and metastasis. We discuss evidence that N- and VE-cadherin play a key role in promoting blood vessel formation and stability, processes which are essential for tumor growth. Secondly, we describe the potential involvement of N- and OB-cadherin in the metastatic process. Finally, studies concerning the effects of the N-cadherin antagonist designated ADH-1 on tumor growth are presented. Collectively, these observations suggest that antagonists of N-, VE- and OB-cadherin would be useful as anti-cancer agents.
Collapse
Affiliation(s)
- Orest W Blaschuk
- Division of Urology, Department of Surgery, McGill University, Urology Research Laboratories, Royal Victoria Hospital, Room H6.15, 687 Pine Avenue West, Montreal, Quebec, Canada H3A 1A1.
| | | |
Collapse
|
46
|
Devemy E, Blaschuk OW. Identification of a novel dual E- and N-cadherin antagonist. Peptides 2009; 30:1539-47. [PMID: 19465078 DOI: 10.1016/j.peptides.2009.05.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Revised: 05/12/2009] [Accepted: 05/13/2009] [Indexed: 01/31/2023]
Abstract
E- and N-cadherin are related calcium-dependent cell adhesion molecules that exert an influence over multiple biological and disease processes. Antagonists of these cadherins can therefore be envisaged as therapeutically useful drugs. We have used phage display technology to discover such antagonists. A peptide phage library was screened against a chimeric protein composed of the human E-cadherin ectodomain fused to the Fc fragment of human immunoglobulin G1 (E-cad/Fc). All of the phage clones that were isolated also bound a chimeric protein composed of the human N-cadherin ectodomain fused to the Fc fragment of human immunoglobulin G1 (N-cad/Fc). A peptide displayed by several of the isolated phage clones was synthesized (H-SWELYYPLRANL-NH2) and found to bind both E- and N-cad/Fc chimeric proteins with affinities (K(D)) of 9.4 microM and 323 nM, respectively, as judged by surface plasmon resonance spectroscopy. This peptide was also capable of blocking the aggregation of E- and N-cad/Fc chimeric protein-coated beads, as well as the aggregation of MCF-7 and MDA-MB435 human breast cancer cells (these cells express E- and N-cadherin, respectively). Finally, we showed that the peptide disrupted MCF-7 and MDA-MB435 cell monolayers. The peptide, H-SWELYYPLRANL-NH(2) thus proved to be a biologically active, dual E- and N-cadherin antagonist. Such an antagonist has application in a wide variety of biological contexts.
Collapse
Affiliation(s)
- Emmanuelle Devemy
- Division of Urology, Department of Surgery, McGill University, Urology Research Laboratories, Royal Victoria Hospital, Room H6.15, 687 Pine Avenue West, Montreal, Quebec, Canada H3A 1A1.
| | | |
Collapse
|
47
|
Katsamba P, Carroll K, Ahlsen G, Bahna F, Vendome J, Posy S, Rajebhosale M, Price S, Jessell TM, Ben-Shaul A, Shapiro L, Honig BH. Linking molecular affinity and cellular specificity in cadherin-mediated adhesion. Proc Natl Acad Sci U S A 2009; 106:11594-9. [PMID: 19553217 PMCID: PMC2710653 DOI: 10.1073/pnas.0905349106] [Citation(s) in RCA: 190] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Indexed: 11/18/2022] Open
Abstract
Many cell-cell adhesive events are mediated by the dimerization of cadherin proteins presented on apposing cell surfaces. Cadherin-mediated processes play a central role in the sorting of cells into separate tissues in vivo, but in vitro assays aimed at mimicking this behavior have yielded inconclusive results. In some cases, cells that express different cadherins exhibit homotypic cell sorting, forming separate cell aggregates, whereas in other cases, intermixed aggregates are formed. A third pattern is observed for mixtures of cells expressing either N- or E-cadherin, which form distinct homotypic aggregates that adhere to one another through a heterotypic interface. The molecular basis of cadherin-mediated cell patterning phenomena is poorly understood, in part because the relationship between cellular adhesive specificity and intermolecular binding free energies has not been established. To clarify this issue, we have measured the dimerization affinities of N-cadherin and E-cadherin. These proteins are similar in sequence and structure, yet are able to mediate homotypic cell patterning behavior in a variety of tissues. N-cadherin is found to form homodimers with higher affinity than does E-cadherin and, unexpectedly, the N/E-cadherin heterophilic binding affinity is intermediate in strength between the 2 homophilic affinities. We can account for observed cell aggregation behaviors by using a theoretical framework that establishes a connection between molecular affinities and cell-cell adhesive specificity. Our results illustrate how graded differences between different homophilic and heterophilic cadherin dimerizaton affinities can result in homotypic cell patterning and, more generally, show how proteins that are closely related can, nevertheless, be responsible for highly specific cellular adhesive behavior.
Collapse
Affiliation(s)
- P. Katsamba
- Department of Biochemistry and Molecular Biophysics
- Howard Hughes Medical Institute, and
| | - K. Carroll
- Department of Biochemistry and Molecular Biophysics
| | - G. Ahlsen
- Department of Biochemistry and Molecular Biophysics
| | - F. Bahna
- Department of Biochemistry and Molecular Biophysics
- Howard Hughes Medical Institute, and
| | - J. Vendome
- Department of Biochemistry and Molecular Biophysics
- Howard Hughes Medical Institute, and
- Center for Computational Biology and Bioinformatics, Columbia University, Room 815, 1130 St. Nicholas Avenue, New York, NY 10032
| | - S. Posy
- Department of Biochemistry and Molecular Biophysics
- Howard Hughes Medical Institute, and
- Center for Computational Biology and Bioinformatics, Columbia University, Room 815, 1130 St. Nicholas Avenue, New York, NY 10032
| | - M. Rajebhosale
- Research Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - S. Price
- Research Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - T. M. Jessell
- Department of Biochemistry and Molecular Biophysics
- Howard Hughes Medical Institute, and
| | - A. Ben-Shaul
- Department of Physical Chemistry and the Fritz Haber Research Center, The Hebrew University, Jerusalem 91904, Israel; and
| | - L. Shapiro
- Department of Biochemistry and Molecular Biophysics
- Edward S. Harkness Eye Institute, Columbia University, New York, NY 10032
| | - Barry H. Honig
- Department of Biochemistry and Molecular Biophysics
- Howard Hughes Medical Institute, and
- Center for Computational Biology and Bioinformatics, Columbia University, Room 815, 1130 St. Nicholas Avenue, New York, NY 10032
| |
Collapse
|
48
|
Dawes AT. A mathematical model of alpha-catenin dimerization at adherens junctions in polarized epithelial cells. J Theor Biol 2009; 257:480-8. [PMID: 19135061 PMCID: PMC2689185 DOI: 10.1016/j.jtbi.2008.11.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2008] [Revised: 11/24/2008] [Accepted: 11/27/2008] [Indexed: 01/20/2023]
Abstract
The protein alpha-catenin is found as a monomer or homodimer. As a monomer, alpha-catenin can bind to beta-catenin, which localizes to the plasma membrane at the site of adherens junctions (AJs) in polarized epithelial cells. As a dimer, alpha-catenin can bind to actin filaments, affecting the organization of the actin cytoskeleton. At usual cytoplasmic concentrations, alpha-catenin is found predominantly in monomeric form. It is currently thought that alpha-catenin cannot simultaneously bind beta-catenin and homodimerize, and that the dynamics of binding and unbinding from beta-catenin, possibly coupled with lower diffusion near an AJ, are sufficient to locally accumulate alpha-catenin monomers and homodimers. Using a mathematical model of alpha-catenin dynamics, I show that alpha-catenin must transiently homodimerize while bound to beta-catenin in order for homodimers to form, even in the presence of a spatial diffusion gradient.
Collapse
Affiliation(s)
- Adriana T Dawes
- Center for Cell Dynamics, University of Washington, 620 University Road, Friday Harbor, WA 98250, USA.
| |
Collapse
|
49
|
Marrs GS, Theisen CS, Brusés JL. N-cadherin modulates voltage activated calcium influx via RhoA, p120-catenin, and myosin-actin interaction. Mol Cell Neurosci 2009; 40:390-400. [PMID: 19162191 PMCID: PMC2883866 DOI: 10.1016/j.mcn.2008.12.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Revised: 12/08/2008] [Accepted: 12/11/2008] [Indexed: 01/12/2023] Open
Abstract
N-cadherin is a transmembrane adhesion receptor that contributes to neuronal development and synapse formation through homophilic interactions that provide structural-adhesive support to contacts between cell membranes. In addition, N-cadherin homotypic binding may initiate cell signaling that regulates neuronal physiology. In this study, we investigated signaling capabilities of N-cadherin that control voltage activated calcium influx. Using whole-cell voltage clamp recording of isolated inward calcium currents in freshly isolated chick ciliary ganglion neurons we show that the juxtamembrane region of N-cadherin cytoplasmic domain regulates high-threshold voltage activated calcium currents by interacting with p120-catenin and activating RhoA. This regulatory mechanism requires myosin interaction with actin. Furthermore, N-cadherin homophilic binding enhanced voltage activated calcium current amplitude in dissociated neurons that have already developed mature synaptic contacts in vivo. The increase in calcium current amplitude was not affected by brefeldin A suggesting that the effect is caused via direct channel modulation and not by increasing channel expression. In contrast, homotypic N-cadherin interaction failed to regulate calcium influx in freshly isolated immature neurons. However, RhoA inhibitors enhanced calcium current amplitude in these immature neurons, suggesting that the inhibitory effect of RhoA on calcium entry is regulated during neuronal development and synapse maturation. These results indicate that N-cadherin modulates voltage activated calcium entry by a mechanism that involves RhoA activity and its downstream effects on the cytoskeleton, and suggest that N-cadherin provides support for synaptic maturation and sustained synaptic activity by facilitating voltage activated calcium influx.
Collapse
Affiliation(s)
| | - Christopher S. Theisen
- University of Kansas School of Medicine, Department of Anatomy and Cell Biology, Kansas City, KS 66160
| | - Juan L. Brusés
- University of Kansas School of Medicine, Department of Anatomy and Cell Biology, Kansas City, KS 66160
| |
Collapse
|
50
|
Abstract
Cadherins are a large family of cell-cell adhesion molecules that tether cytoskeletal networks of actin and intermediate filaments to the plasma membrane. This function of cadherins promotes tissue organization and integrity, as demonstrated by numerous disease states that are characterized by the loss of cadherin-based adhesion. However, plasticity in cell adhesion is often required in cellular processes such as tissue patterning during development and epithelial migration during wound healing. Recent work has revealed a pivotal role for various membrane trafficking pathways in regulating cellular transitions between quiescent adhesive states and more dynamic phenotypes. The regulation of cadherins by membrane trafficking is emerging as a key player in this balancing act, and studies are beginning to reveal how this process goes awry in the context of disease. This review summarizes the current understanding of how cadherins are routed and how the interface between cadherins and membrane trafficking pathways regulates cell surface adhesive potential. Particular emphasis is placed on the regulation of cadherin trafficking by catenins and the interplay between growth factor signaling pathways and cadherin endocytosis.
Collapse
Affiliation(s)
- Emmanuella Delva
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30332
- Biochemistry, Cell, and Development Biology Graduate Program, Emory University School of Medicine, Atlanta, GA 30332
| | - Andrew P. Kowalczyk
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30332
- Department of Dermatology, Emory University School of Medicine, Atlanta, GA 30332
| |
Collapse
|