1
|
Vu V, Sullivan B, Hebner E, Rahil Z, Zou Y, Leckband D. Cadherins and growth factor receptors - ligand-selective mechano-switches at cadherin junctions. J Cell Sci 2025; 138:JCS262279. [PMID: 39817537 PMCID: PMC11883276 DOI: 10.1242/jcs.262279] [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: 05/18/2024] [Accepted: 01/06/2025] [Indexed: 01/18/2025] Open
Abstract
This study investigated possible mechanisms underlying differences between heterophilic and homophilic cadherin adhesions that influence intercellular mechanics and multicellular organization. Results suggest that homophilic cadherin ligation selectively activates force transduction, such that resulting signaling and mechano-transduction amplitudes are independent of cadherin-binding affinities. Epithelial (E-) and neural (N-)cadherin cooperate with distinct growth factors to mechanically activate force transduction cascades. Prior results have demonstrated that E-cadherin and epidermal growth factor receptor form force-sensitive complexes at intercellular junctions. Here, we show that the reconstitution of N-cadherin force transduction requires the co-expression of N-cadherin and fibroblast growth factor receptor. Mechanical measurements further demonstrated that homophilic ligation initiates receptor tyrosine kinase-dependent force transduction cascades, but heterophilic cadherin ligands fail to activate signaling or generate stereotypical mechano-transduction signatures. The all-or-nothing contrast between mechano-transduction by heterophilic versus homophilic cadherin adhesions supersedes differences in cadherin adhesion strength. This mechano-selectivity impacts cell spreading and traction generation on cadherin substrates. Homophilic ligation appears to be a key that selectively unlocks cadherin mechano-transduction. These findings might reconcile the roles of cadherin recognition and cell mechanics in the organization of multicellular assemblies.
Collapse
Affiliation(s)
- Vinh Vu
- Department of Biochemistry, University of Illinois, 600 South Mathews Ave, Urbana, IL 61801, USA
| | - Brendan Sullivan
- Department of Biochemistry, University of Illinois, 600 South Mathews Ave, Urbana, IL 61801, USA
| | - Evan Hebner
- Department of Biochemistry, University of Illinois, 600 South Mathews Ave, Urbana, IL 61801, USA
| | - Zainab Rahil
- Department of Bioengineering, University of Illinois, 1402 W Green St., Urbana, IL 61801, USA
| | - Yubo Zou
- Department of Biochemistry, University of Illinois, 600 South Mathews Ave, Urbana, IL 61801, USA
| | - Deborah Leckband
- Department of Biochemistry, University of Illinois, 600 South Mathews Ave, Urbana, IL 61801, USA
- Department of Bioengineering, University of Illinois, 1402 W Green St., Urbana, IL 61801, USA
- Department of Chemical and Biomolecular Engineering, University of Illinois, 600 South Mathews Ave, Urbana, IL 61801, USA
- Department of Chemistry, University of Illinois, 600 South Mathews Ave, Urbana, IL 61801, USA
- Carl W. Woese Institute for Genomic Biology, 1206 West Gregory Drive, Urbana, IL 61801, USA
| |
Collapse
|
2
|
Oda H, Nishiguchi S, Song C, Murata K, Uchihashi T, Suzuki Y. Nanoscale Visualization of Drosophila E-cadherin Ectodomain Fragments and Their Interactions Using DNA Origami Nanoblocks. J Mol Biol 2025; 437:168875. [PMID: 39581222 DOI: 10.1016/j.jmb.2024.168875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/03/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024]
Abstract
The adhesive function of cell surface proteins can be visually assessed through direct observation; however, the underlying structures that mediate adhesion typically remain invisible at the nanoscale level. This hinders knowledge on the diversity of molecular architectures responsible for cell-cell adhesion. Drosophila E-cadherin (DE-cadherin), a classical cadherin with a unique domain structure, demonstrates adhesive function; however, it lacks a structural model that explains its adhesion mechanism. Here, we present a novel application of DNA origami technology to create a cell-free, flat environment in which full DE-cadherin ectodomains are anchored using SNAP-tags and biotin-streptavidin interactions. DNA origami was assembled into a 120 nm long block, bearing 5 or 14 biotin:streptavidin sites that were evenly spaced on one lateral face. DE-cadherin ectodomain fragments were attached via biotinylated SNAP-tags. These decorated DNA origami nanoblocks were subjected to transmission electron and high-speed atomic force microscopy, which revealed a hinge-like site that separated the membrane-distal and -proximal portions of the DE-cadherin ectodomain, suggesting a role in mechanical flexibility. We also observed interactions between DE-cadherin ectodomains via their membrane-distal portions on single DNA origami nanoblocks. We reconstituted an adhesion-like process via pairing DNA origami nanoblocks using DE-cadherin ectodomain interactions. Homophilic associations of functional DE-cadherin ectodomains between the paired DNA origami nanoblocks were visualized at the nanoscale, displaying strand-like molecular configurations, likely representing the extracellular cadherin repeats without regular arrays of structural elements. This study introduces a DNA origami-based platform for reconstituting and visualizing cadherin ectodomain interactions, with potential applications for a broader range of adhesion molecules.
Collapse
Affiliation(s)
- Hiroki Oda
- Laboratory of Evolutionary Cell and Developmental Biology, JT Biohistory Research Hall, 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan; Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama-cho, Toyonaka, Osaka 560-0043, Japan.
| | - Shigetaka Nishiguchi
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
| | - Chihong Song
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan; National Institute for Physiological Sciences, National Institutes of Natural Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), 38 Nishigonaka, Myodaiji, Okazaki, Aichi, 444-8585, Japan
| | - Kazuyoshi Murata
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan; National Institute for Physiological Sciences, National Institutes of Natural Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), 38 Nishigonaka, Myodaiji, Okazaki, Aichi, 444-8585, Japan
| | - Takayuki Uchihashi
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan; Department of Physics, Graduate School of Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi 464-8602, Japan
| | - Yuki Suzuki
- Department of Applied Chemistry, Graduate School of Engineering, Mie University, 1577 Kurimamachiya-cho, Tsu, Mie 514-8507, Japan
| |
Collapse
|
3
|
Vachharajani VT, DeJong MP, Dutta S, Chapman J, Ghosh E, Singharoy A, Dunn AR. PDZ Domains from the Junctional Proteins Afadin and ZO-1 Act as Mechanosensors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.09.24.559210. [PMID: 37961673 PMCID: PMC10634676 DOI: 10.1101/2023.09.24.559210] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Intercellular adhesion complexes must withstand mechanical forces to maintain tissue cohesion while also retaining the capacity for dynamic remodeling during tissue morphogenesis and repair. Many cell-cell adhesion complexes contain at least one PSD95/Dlg/ZO-1 (PDZ) domain situated between the adhesion molecule and the actin cytoskeleton. However, PDZ-mediated interactions are characteristically nonspecific, weak, and transient, with multiple binding partners per PDZ domain, micromolar dissociation constants, and bond lifetimes of seconds or less. Here, we demonstrate that the bonds between the PDZ domain of the cytoskeletal adaptor protein afadin and the intracellular domains of the adhesion molecules nectin-1 and JAM-A form molecular catch bonds that reinforce in response to mechanical load. In contrast, the bond between the PDZ3-SH3-GUK (PSG) domain of the cytoskeletal adaptor ZO-1 and the JAM-A intracellular domain becomes dramatically weaker in response to ∼2 pN of load, the amount generated by single molecules of the cytoskeletal motor protein myosin II. Thus, physiologically relevant forces can exert dramatic and opposite effects on the stability of two of the major linkages between cell-cell adhesion proteins and the F-actin cytoskeleton. Our data demonstrate that that PDZ domains can serve as force-responsive mechanical anchors at cell-cell adhesion complexes. More broadly, our findings suggest that mechanical force may serve as a previously unsuspected regulator of the hundreds of PDZ-ligand interactions present in animal cells.
Collapse
|
4
|
Petersen M, Reyes-Vigil F, Campo M, Brusés JL. Classical cadherins evolutionary constraints in primates is associated with their expression in the central nervous system. PLoS One 2024; 19:e0313428. [PMID: 39570883 PMCID: PMC11581309 DOI: 10.1371/journal.pone.0313428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024] Open
Abstract
Classical cadherins (CDH) comprise a family of single-pass transmembrane glycoproteins that contribute to tissue morphogenesis by regulating cell-cell adhesion, cytoskeletal dynamics, and cell signaling. CDH are grouped into type I (CDH 1, 2, 3, 4 and 15) and type II (CDH 5, 6, 7, 8, 9, 10, 11, 12, 18, 20, 22 and 24), based on the folding of the cadherin binding domain involved in trans-dimer formation. CDH are exclusively found in metazoans, and the origin and expansion of the gene family coincide with the emergence of multicellularity and vertebrates respectively. This study examined the evolutionary changes of CDH orthologs in primates and the factors that influence selective pressure to investigate the varying constraints exerted among CDH. Pairwise comparisons of the number of amino acid substitutions and of the ratio of non-synonymous substitutions per non-synonymous sites (dN) over synonymous substitutions per synonymous sites (dS), show that CDH2, CDH4, and most type II CDH have been under significantly higher negative selective pressure as compared to CDH1, CDH3, CDH5 and CDH19. Evaluation of gene essentiality as determined by the effect of germline deletion on animal viability, morphogenic phenotype, and reproductive fitness, show no correlation with the with extent of negative selection observed on CDH. Spearman's correlation analysis shows a positive correlation between CDH expression levels (E) in mouse and human tissues and their rate of evolution (R), as observed in most proteins expressed on the cell surface. However, CDH expression in the CNS show a significant E-R negative correlation, indicating that the strong negative selection exerted on CDH2, CDH4, and most type II CDH is associated with their expression in the CNS. CDH participate in a variety of cellular processes in the CNS including neuronal migration and functional assembly of neural circuits, which could profoundly influence animal fitness. Therefore, our findings suggest that the unusually high negative selective pressure exerted on CDH2, CDH4 and most type II CDH is due to their role in CNS formation and function and may have contributed to shape the evolution of the CNS in primates.
Collapse
Affiliation(s)
- Max Petersen
- Department of Natural Sciences, School of Health and Natural Sciences, Mercy University, Dobbs Ferry, New York, United States of America
| | - Fredy Reyes-Vigil
- Department of Natural Sciences, School of Health and Natural Sciences, Mercy University, Dobbs Ferry, New York, United States of America
| | - Marc Campo
- Department of Natural Sciences, School of Health and Natural Sciences, Mercy University, Dobbs Ferry, New York, United States of America
| | - Juan L. Brusés
- Department of Natural Sciences, School of Health and Natural Sciences, Mercy University, Dobbs Ferry, New York, United States of America
| |
Collapse
|
5
|
Kozlova I, Sytnyk V. Cell Adhesion Molecules as Modulators of the Epidermal Growth Factor Receptor. Cells 2024; 13:1919. [PMID: 39594667 PMCID: PMC11592701 DOI: 10.3390/cells13221919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Cell adhesion molecules (CAMs) are cell surface glycoproteins mediating interactions of cells with other cells and the extracellular matrix. By mediating the adhesion and modulating activity of other plasma membrane proteins, CAMs are involved in regulating a multitude of cellular processes, including growth, proliferation, migration, and survival of cells. In this review, we present evidence showing that various CAMs interact with the epidermal growth factor receptor (EGFR), a receptor tyrosine kinase inducing pro-proliferative and anti-apoptotic intracellular signaling in response to binding to several soluble ligands, including the epidermal growth factor. We discuss that CAMs are involved in regulating EGFR signaling by either potentiating or inhibiting the soluble ligand-dependent activation of EGFR. In addition, CAMs induce soluble ligand-independent forms of EGFR activity and regulate the levels of EGFR and its ligand-induced degradation. The CAM-dependent modulation of EGFR activity plays a key role in regulating the growth, proliferation, and survival of cells. Future research is needed to determine whether these processes can be targeted in both normal and cancerous cells by regulating interactions of EGFR with various CAMs.
Collapse
Affiliation(s)
| | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW 2052, Australia;
| |
Collapse
|
6
|
Farokhi E, Alaofi AL, Prasasty VD, Stephanie F, Laksitorini MD, Kuczera K, Siahaan TJ. Mechanism of the blood-brain barrier modulation by cadherin peptides. EXPLORATION OF DRUG SCIENCE 2024; 2:322-338. [PMID: 39118806 PMCID: PMC11309765 DOI: 10.37349/eds.2024.00049] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 03/21/2024] [Indexed: 08/10/2024]
Abstract
Aim This study was aimed at finding the binding site on the human E-cadherin for Ala-Asp-Thr Cyclic 5 (ADTC5), ADTC7, and ADTC9 peptides as blood-brain barrier modulator (BBBM) for determining their mechanism of action in modulating the blood-brain barrier (BBB). Methods ADTC7 and ADTC9 were derivatives of ADTC5 where the Val6 residue in ADTC5 was replaced by Glu6 and Tyr6 residues, respectively. The binding properties of ADTC5, ADTC7, and ADTC9 to the extracellular-1 (EC1) domain of E-cadherin were evaluated using chemical shift perturbation (CSP) method in the two dimensional (2D) 1H-15N-heteronuclear single quantum coherence (HSQC) nuclear magnetic resonance (NMR) spectroscopy. Molecular docking experiments were used to determine the binding sites of these peptides to the EC1 domain of E-cadherin. Results This study indicates that ADTC5 has the highest binding affinity to the EC1 domain of E-cadherin compared to ADTC7 and ADTC9, suggesting the importance of the Val6 residue as shown in our previous in vitro study. All three peptides have a similar binding site at the hydrophobic binding pocket where the domain swapping occurs. ADTC5 has a higher overlapping binding site with ADTC7 than that of ADTC9. Binding of ADTC5 on the EC1 domain influences the conformation of the EC1 C-terminal tail. Conclusions These peptides bind the domain swapping region of the EC1 domain to inhibit the trans-cadherin interaction that creates intercellular junction modulation to increase the BBB paracellular porosity.
Collapse
Affiliation(s)
- Elinaz Farokhi
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
- Current address: Analytical Department, Johnson & Johnson, San Diego, CA 92123, USA
| | - Ahmed L. Alaofi
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
- Current address: Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Vivitri D. Prasasty
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
- Current address: Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Filia Stephanie
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
| | - Marlyn D. Laksitorini
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
- Current address: School of Pharmacy, University of Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Krzysztof Kuczera
- Department of Chemistry, The University of Kansas, Lawrence, KS 66047, USA
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS 66047, USA
| | - Teruna J. Siahaan
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA
| |
Collapse
|
7
|
Castro-Hinojosa C, Del Sol-Fernández S, Moreno-Antolín E, Martín-Gracia B, Ovejero JG, de la Fuente JM, Grazú V, Fratila RM, Moros M. A Simple and Versatile Strategy for Oriented Immobilization of His-Tagged Proteins on Magnetic Nanoparticles. Bioconjug Chem 2023; 34:2275-2292. [PMID: 37882455 PMCID: PMC10739578 DOI: 10.1021/acs.bioconjchem.3c00417] [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: 09/22/2023] [Revised: 10/16/2023] [Accepted: 10/16/2023] [Indexed: 10/27/2023]
Abstract
Oriented and covalent immobilization of proteins on magnetic nanoparticles (MNPs) is particularly challenging as it requires both the functionality of the protein and the colloidal stability of the MNPs to be preserved. Here, we describe a simple, straightforward, and efficient strategy for MNP functionalization with proteins using metal affinity binding. Our method involves a single-step process where MNPs are functionalized using a preformed, ready-to-use nitrilotriacetic acid-divalent metal cation (NTA-M2+) complex and polyethylene glycol (PEG) molecules. As a proof-of-concept, we demonstrate the oriented immobilization of a recombinant cadherin fragment engineered with a hexahistidine tag (6His-tag) onto the MNPs. Our developed methodology is simple and direct, enabling the oriented bioconjugation of His-tagged cadherins to MNPs while preserving protein functionality and the colloidal stability of the MNPs, and could be extended to other proteins expressing a polyhistidine tag. When compared to the traditional method where NTA is first conjugated to the MNPs and afterward free metal ions are added to form the complex, this novel strategy results in a higher functionalization efficiency while avoiding MNP aggregation. Additionally, our method allows for covalent bonding of the cadherin fragments to the MNP surface while preserving functionality, making it highly versatile. Finally, our strategy not only ensures the correct orientation of the protein fragments on the MNPs but also allows for the precise control of their density. This feature enables the selective targeting of E-cadherin-expressing cells only when MNPs are decorated with a high density of cadherin fragments.
Collapse
Affiliation(s)
- Christian Castro-Hinojosa
- Instituto
de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, Zaragoza 50009, Spain
| | - Susel Del Sol-Fernández
- Instituto
de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, Zaragoza 50009, Spain
| | - Eduardo Moreno-Antolín
- Instituto
de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, Zaragoza 50009, Spain
| | - Beatriz Martín-Gracia
- Instituto
de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, Zaragoza 50009, Spain
| | - Jesús G. Ovejero
- Instituto
de Ciencia de Materiales de Madrid (ICMM/CSIC), Sor Juana Inés de la Cruz 3, Madrid 28049, Spain
- Department
of Dosimetry and Radioprotection, General
University Hospital Gregorio Marañón, Dr Esquerdo 46, Madrid 28007, Spain
| | - Jesús Martínez de la Fuente
- Instituto
de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, Zaragoza 50009, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
| | - Valeria Grazú
- Instituto
de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, Zaragoza 50009, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
| | - Raluca M. Fratila
- Instituto
de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, Zaragoza 50009, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
- Departamento
de Química Orgánica, Facultad de Ciencias, Universidad de Zaragoza, C/Pedro Cerbuna 12, Zaragoza 50009, Spain
| | - María Moros
- Instituto
de Nanociencia y Materiales de Aragón, INMA (CSIC-Universidad de Zaragoza), C/Pedro Cerbuna 12, Zaragoza 50009, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
| |
Collapse
|
8
|
Sivasankar S, Xie B. Engineering the Interactions of Classical Cadherin Cell-Cell Adhesion Proteins. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:343-349. [PMID: 37459190 PMCID: PMC10361579 DOI: 10.4049/jimmunol.2300098] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/30/2023] [Indexed: 07/20/2023]
Abstract
Classical cadherins are calcium-dependent cell-cell adhesion proteins that play key roles in the formation and maintenance of tissues. Deficiencies in cadherin adhesion are hallmarks of numerous cancers. In this article, we review recent biophysical studies on the regulation of cadherin structure and adhesion. We begin by reviewing distinct cadherin binding conformations, their biophysical properties, and their response to mechanical stimuli. We then describe biophysical guidelines for engineering Abs that can regulate adhesion by either stabilizing or destabilizing cadherin interactions. Finally, we review molecular mechanisms by which cytoplasmic proteins regulate the conformation of cadherin extracellular regions from the inside out.
Collapse
Affiliation(s)
- Sanjeevi Sivasankar
- Department of Biomedical Engineering, University of California, Davis, CA 95616
- Biophysics Graduate Group, University of California, Davis, CA 95616
| | - Bin Xie
- Biophysics Graduate Group, University of California, Davis, CA 95616
| |
Collapse
|
9
|
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
|
10
|
Al Zubi K, Mwafi N, Alrawashdeh HM, Al Sarireh F, Somkuwar A, Abdulmannan DM. The first reported case of CDH3-related hypotrichosis with juvenile macular dystrophy from Jordan: a case report. Ophthalmic Genet 2022; 43:420-424. [PMID: 35038959 DOI: 10.1080/13816810.2022.2028298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Pathogenic variants in the Cadherin 3 (CDH3) gene are responsible for the occurrence of Hypotrichosis with Juvenile Macular Dystrophy (HJMD) and Ectodermal Dysplasia, Ectrodactyly and Macular Dystrophy Syndrome (EEMS), both of which are rare autosomal recessive disorders characterized by hypotrichosis and progressive macular dystrophy. The CDH3 gene encodes for P-cadherin, a calcium-binding protein that is essential for cell-cell adhesion, which is expressed in the retinal pigment epithelial cells and hair follicles. MATERIALS AND METHODS Fundus examination of both eyes was done in addition to clinical investigation. Genomic DNA was extracted from a whole-blood sample and whole-exome sequencing (WES) was performed to identify the underlying etiology.All identified variants were evaluated for their pathogenicity and causality. RESULTS We present the first case of HJMD in a 23-year-old female patient from Jordan. The patient presented to our ophthalmology clinic with poor vision in both eyes. Gross examination revealed sparse scalp hair along with macular dystrophy on fundus exam in both eyes. HJMD was suspected and whole-exome sequencing (WES) confirmed the diagnosis with the identification of a homozygous frameshift deletion (p.Gly277AlafsTer20) localised in exon 7 of the CDH3 gene. CONCLUSION Blindness due to progressive macular degeneration is a common manifestation in numerous syndromic recessive disorders such as HJMD. Ophthalmologists should consider the importance of systemic manifestations and genetic testing for the confirmation of diagnosis.
Collapse
Affiliation(s)
- Khalid Al Zubi
- Special Surgery Department, Faculty of Medicine, Mutah University, Al-karak, Jordan
| | - Nesrin Mwafi
- Biochemistry and Molecular Biology Department, Faculty of Medicine, Mutah University, Al-karak, Jordan
| | | | - Fawaz Al Sarireh
- Special Surgery Department, Faculty of Medicine, Mutah University, Al-karak, Jordan
| | - Areej Somkuwar
- Ophthalmology Department, Al-Karak Governmental Hospital, Al-karak, Jordan
| | - Dina M Abdulmannan
- Ophthalmology Department, Faculty of Medicine, Umm Alqura University, Makkah, Saudi Arabia
| |
Collapse
|
11
|
Huene AL, Nicotra ML. Cell Aggregation Assays for Homophilic Interactions Between Cell Surface Proteins. Methods Mol Biol 2022; 2421:91-102. [PMID: 34870813 DOI: 10.1007/978-1-0716-1944-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Many proteins expressed on the cellular surface provide signaling and cell adhesion properties required for vital cellular functions. These binding interactions can occur between different but complementary proteins such as a ligand and receptor, or between the same protein acting as both ligand and receptor. The cell aggregation assay is a straightforward technique to identify homophilic interactions from such proteins. Here we describe the procedure for testing proteins via cell aggregation assays in HEK293T cells.
Collapse
Affiliation(s)
- Aidan L Huene
- Department of Surgery and Center for Evolutionary Biology and Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Matthew L Nicotra
- Departments of Surgery and Immunology, Thomas E. Starzl Transplantation Institute, and Center for Evolutionary Biology and Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
12
|
Aladin DMK, Chu YS, Shen S, Robinson RC, Dufour S, Viasnoff V, Borghi N, Thiery JP. Extracellular domains of E-cadherin determine key mechanical phenotypes of an epithelium through cell- and non-cell-autonomous outside-in signaling. PLoS One 2021; 16:e0260593. [PMID: 34937057 PMCID: PMC8694416 DOI: 10.1371/journal.pone.0260593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 11/14/2021] [Indexed: 11/18/2022] Open
Abstract
Cadherins control intercellular adhesion in most metazoans. In vertebrates, intercellular adhesion differs considerably between cadherins of type-I and type-II, predominantly due to their different extracellular regions. Yet, intercellular adhesion critically depends on actomyosin contractility, in which the role of the cadherin extracellular region is unclear. Here, we dissect the roles of the Extracellular Cadherin (EC) Ig-like domains by expressing chimeric E-cadherin with E-cadherin and cadherin-7 Ig-like domains in cells naturally devoid of cadherins. Using cell-cell separation, cortical tension measurement, tissue stretching and migration assays, we show that distinct EC repeats in the extracellular region of cadherins differentially modulate epithelial sheet integrity, cell-cell separation forces, and cell cortical tension with the Cdc42 pathway, which further differentially regulate epithelial tensile strength, ductility, and ultimately collective migration. Interestingly, dissipative processes rather than static adhesion energy mostly dominate cell-cell separation forces. We provide a framework for the emergence of epithelial phenotypes from cell mechanical properties dependent on EC outside-in signaling.
Collapse
Affiliation(s)
- Darwesh Mohideen Kaderbatcha Aladin
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
- BioSyM Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology (SMART), Singapore, Singapore
| | - Yeh Shiu Chu
- Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
| | - Shuo Shen
- Sinopharm, Zhengdian, Jiangxia District, Wuhan, Hubei, China
| | | | - Sylvie Dufour
- IMRB, Université Paris Est Créteil, INSERM, Créteil, France
- * E-mail: (NB); (VV); (SD); (JPT)
| | - Virgile Viasnoff
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- UMI 3639 CNRS, Singapore
- * E-mail: (NB); (VV); (SD); (JPT)
| | - Nicolas Borghi
- Institut Jacques Monod, Université de Paris, CNRS, Paris, France
- * E-mail: (NB); (VV); (SD); (JPT)
| | - Jean Paul Thiery
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
- Guangzhou Laboratory, International Bioisland, Guangzhou, Haizhu District, China
- * E-mail: (NB); (VV); (SD); (JPT)
| |
Collapse
|
13
|
Yui A, Caaveiro JMM, Kuroda D, Nakakido M, Nagatoishi S, Goda S, Maruno T, Uchiyama S, Tsumoto K. Mechanism of dimerization and structural features of human LI-cadherin. J Biol Chem 2021; 297:101054. [PMID: 34364873 PMCID: PMC8427231 DOI: 10.1016/j.jbc.2021.101054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/02/2021] [Accepted: 08/04/2021] [Indexed: 11/24/2022] Open
Abstract
Liver intestine (LI)-cadherin is a member of the cadherin superfamily, which encompasses a group of Ca2+-dependent cell-adhesion proteins. The expression of LI-cadherin is observed on various types of cells in the human body, such as normal small intestine and colon cells, and gastric cancer cells. Because its expression is not observed on normal gastric cells, LI-cadherin is a promising target for gastric cancer imaging. However, because the cell adhesion mechanism of LI-cadherin has remained unknown, rational design of therapeutic molecules targeting this cadherin has been hampered. Here, we have studied the homodimerization mechanism of LI-cadherin. We report the crystal structure of the LI-cadherin homodimer containing its first four extracellular cadherin repeats (EC1-4). The EC1-4 homodimer exhibited a unique architecture different from that of other cadherins reported so far, driven by the interactions between EC2 of one protein chain and EC4 of the second protein chain. The crystal structure also revealed that LI-cadherin possesses a noncanonical calcium ion-free linker between the EC2 and EC3 domains. Various biochemical techniques and molecular dynamics simulations were employed to elucidate the mechanism of homodimerization. We also showed that the formation of the homodimer observed in the crystal structure is necessary for LI-cadherin-dependent cell adhesion by performing cell aggregation assays. Taken together, our data provide structural insights necessary to advance the use of LI-cadherin as a target for imaging gastric cancer.
Collapse
Affiliation(s)
- Anna Yui
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Jose M M Caaveiro
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan; Department of Global Healthcare, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.
| | - Daisuke Kuroda
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan; Medical Device Development and Regulation Research Center, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Makoto Nakakido
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | | | - Shuichiro Goda
- Graduate School of Science and Engineering, Soka University, Tokyo, Japan
| | - Takahiro Maruno
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Susumu Uchiyama
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Kouhei Tsumoto
- Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, Japan; Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
14
|
Li D, March ME, Fortugno P, Cox LL, Matsuoka LS, Monetta R, Seiler C, Pyle LC, Bedoukian EC, Sánchez-Soler MJ, Caluseriu O, Grand K, Tam A, Aycinena ARP, Camerota L, Guo Y, Sleiman P, Callewaert B, Kumps C, Dheedene A, Buckley M, Kirk EP, Turner A, Kamien B, Patel C, Wilson M, Roscioli T, Christodoulou J, Cox TC, Zackai EH, Brancati F, Hakonarson H, Bhoj EJ. Pathogenic variants in CDH11 impair cell adhesion and cause Teebi hypertelorism syndrome. Hum Genet 2021; 140:1061-1076. [PMID: 33811546 DOI: 10.1007/s00439-021-02274-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/04/2021] [Indexed: 11/28/2022]
Abstract
Teebi hypertelorism syndrome (THS; OMIM 145420) is a rare craniofacial disorder characterized by hypertelorism, prominent forehead, short nose with broad or depressed nasal root. Some cases of THS have been attributed to SPECC1L variants. Homozygous variants in CDH11 truncating the transmembrane and intracellular domains have been implicated in Elsahy-Waters syndrome (EWS; OMIM 211380) with hypertelorism. We report THS due to CDH11 heterozygous missense variants on 19 subjects from 9 families. All affected residues in the extracellular region of Cadherin-11 (CHD11) are highly conserved across vertebrate species and classical cadherins. Six of the variants that cluster around the EC2-EC3 and EC3-EC4 linker regions are predicted to affect Ca2+ binding that is required for cadherin stability. Two of the additional variants [c.164G > C, p.(Trp55Ser) and c.418G > A, p.(Glu140Lys)] are also notable as they are predicted to directly affect trans-homodimer formation. Immunohistochemical study demonstrates that CDH11 is strongly expressed in human facial mesenchyme. Using multiple functional assays, we show that five variants from the EC1, EC2-EC3 linker, and EC3 regions significantly reduced the cell-substrate trans adhesion activity and one variant from EC3-EC4 linker results in changes in cell morphology, focal adhesion, and migration, suggesting dominant negative effect. Characteristic features in this cohort included depressed nasal root, cardiac and umbilical defects. These features distinguished this phenotype from that seen in SPECC1L-related hypertelorism syndrome and CDH11-related EWS. Our results demonstrate heterozygous variants in CDH11, which decrease cell-cell adhesion and increase cell migratory behavior, cause a form of THS, as termed CDH11-related THS.
Collapse
Affiliation(s)
- Dong Li
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Michael E March
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Paola Fortugno
- Laboratory of Molecular and Cell Biology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Rome, Italy.,Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Liza L Cox
- Departments of Oral and Craniofacial Sciences and Pediatrics, University of Missouri-Kansas City School of Dentistry, Kansas City, MO, 64108, USA
| | - Leticia S Matsuoka
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rosanna Monetta
- Laboratory of Molecular and Cell Biology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Rome, Italy.,Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Christoph Seiler
- Zebrafish Core Facility, The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Louise C Pyle
- Individualized Medical Genetics Center, Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Emma C Bedoukian
- Individualized Medical Genetics Center, Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - María José Sánchez-Soler
- Sección de Genética Médica, Servicio de Pediatría, Hospital Clínico Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, España
| | - Oana Caluseriu
- Department of Medical Genetics, University of Alberta, Edmonton, AB, T6G 2H7, Canada.,The Stollery Pediatric Hospital, Edmonton, AB, T6G 2H7, Canada
| | - Katheryn Grand
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Allison Tam
- Division of Medical Genetics, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Alicia R P Aycinena
- Division of Medical Genetics, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Letizia Camerota
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Yiran Guo
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Patrick Sleiman
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bert Callewaert
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Candy Kumps
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Annelies Dheedene
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Michael Buckley
- NSW Health Pathology Genomics Laboratory, Prince of Wales Hospital, Randwick, NSW, Australia
| | - Edwin P Kirk
- NSW Health Pathology Genomics Laboratory, Prince of Wales Hospital, Randwick, NSW, Australia.,Centre for Clinical Genetics, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Anne Turner
- Centre for Clinical Genetics, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Benjamin Kamien
- Genetic Services of Western Australia, King Edward Memorial Hospital, Perth, Australia
| | - Chirag Patel
- Genetic Health Queensland, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Meredith Wilson
- Department of Clinical Genetics, Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Tony Roscioli
- NSW Health Pathology Genomics Laboratory, Prince of Wales Hospital, Randwick, NSW, Australia.,Centre for Clinical Genetics, Sydney Children's Hospital, Randwick, NSW, Australia.,Neuroscience Research Australia and Prince of Wales Clinical School, University of New South Wales, Kensington, NSW, Australia
| | - John Christodoulou
- Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Australia.,Discipline of Child and Adolescent Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Timothy C Cox
- Departments of Oral and Craniofacial Sciences and Pediatrics, University of Missouri-Kansas City School of Dentistry, Kansas City, MO, 64108, USA
| | - Elaine H Zackai
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Francesco Brancati
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.,Institute of Translational Pharmacology, National Research Council, Rome, Italy.,IRCCS San Raffaele Pisana, Rome, Italy
| | - Hakon Hakonarson
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elizabeth J Bhoj
- Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA. .,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. .,Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
15
|
Ichikawa T, Stuckenholz C, Davidson LA. Non-junctional role of Cadherin3 in cell migration and contact inhibition of locomotion via domain-dependent, opposing regulation of Rac1. Sci Rep 2020; 10:17326. [PMID: 33060598 PMCID: PMC7567069 DOI: 10.1038/s41598-020-73862-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 09/07/2020] [Indexed: 11/08/2022] Open
Abstract
Classical cadherins are well-known adhesion molecules responsible for physically connecting neighboring cells and signaling this cell-cell contact. Recent studies have suggested novel signaling roles for "non-junctional" cadherins (NJCads); however, the function of cadherin signaling independent of cell-cell contacts remains unknown. In this study, mesendodermal cells and tissues from gastrula stage Xenopus laevis embryos demonstrate that deletion of extracellular domains of Cadherin3 (Cdh3; formerly C-cadherin in Xenopus) disrupts contact inhibition of locomotion. In both bulk Rac1 activity assays and spatio-temporal FRET image analysis, the extracellular and cytoplasmic Cdh3 domains disrupt NJCad signaling and regulate Rac1 activity in opposing directions. Stabilization of the cytoskeleton counteracted this regulation in single cell migration assays. Our study provides novel insights into adhesion-independent signaling by Cadherin3 and its role in regulating single and collective cell migration.
Collapse
Affiliation(s)
- Takehiko Ichikawa
- Department of Bioengineering, University of Pittsburgh, 3501 Fifth Avenue, 5059-BST3, Pittsburgh, PA, 15260, USA
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, 920-1192, Japan
| | - Carsten Stuckenholz
- Department of Bioengineering, University of Pittsburgh, 3501 Fifth Avenue, 5059-BST3, Pittsburgh, PA, 15260, USA
| | - Lance A Davidson
- Department of Bioengineering, University of Pittsburgh, 3501 Fifth Avenue, 5059-BST3, Pittsburgh, PA, 15260, USA.
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, 15260, USA.
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
16
|
Richardson T, Wiegand C, Adisa F, Ravikumar K, Candiello J, Kumta P, Banerjee I. Engineered peptide modified hydrogel platform for propagation of human pluripotent stem cells. Acta Biomater 2020; 113:228-239. [PMID: 32603868 DOI: 10.1016/j.actbio.2020.06.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/24/2022]
Abstract
Human pluripotent stem cells (hPSCs) have enormous potential to alleviate cell needs for regenerative medicine, however these cells require expansion in cell colonies to maintain cell-cell contact, thus limiting the scalability needed to meet the demands of cell therapy. While the use of a Rho-associated protein kinase (ROCK) inhibitor will allow for culture of single cell hPSCs, typically only 50% of cells are recovered after dissociation. When hPSCs lose cell-cell contact through E-cadherin, dissociation induced apoptosis occurs. In this study, we hypothesized that the extracellular E-cadherin domain of hPSCs will bind to synthetic E-cadherin peptides presented on a hydrogel substrate, mimicking the required cell-cell contact and thereby retaining single-cell viability and clonogenicity. Hence, the objective of this study was to functionalize alginate hydrogels with synthetic peptides mimicking E-cadherin and evaluate peptide performance in promoting cell attachment, viability, maintaining pluripotency, and differentiation potential. We observed that alginate conjugated with synthetic E-cadherin peptides not only supported initial cell attachment with high viability, but also supported hPSC propagation and high fold expansion. hPSCs propagated on the peptide modified substrates maintained the hPSC characteristic pluripotency and differentiation potential, characterized by both spontaneous and directed differentiation. STATEMENT OF SIGNIFICANCE: Human pluripotent stem cells (hPSCs) have enormous potential to alleviate cell needs for regenerative medicine and cell therapy. However, scalable culture of hPSCs is challenged by its need for maintenance of cell-cell contact, dissociation of which triggers the apoptotic pathway. Hence hPSCs are commonly maintained as colonies over Matrigel coated culture plates. Furthermore, use of xenogenic and undefined Matrigel compromises the translational potential of hPSCs. In this work we have developed a completely defined substrate to enable adherent culture of hPSCs as single cells. This substrate prevents apoptosis of the single cells and allows significant fold expansion of hPSCs while maintaining pluripotency and differentiation potential. The developed substrate is expected to be a cost-effective and translatable alternative to Matrigel.
Collapse
Affiliation(s)
- Thomas Richardson
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, United States
| | - Connor Wiegand
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, United States
| | - Fatimah Adisa
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, United States
| | - K Ravikumar
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, United States
| | - Joe Candiello
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, United States
| | - Prashant Kumta
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, United States; Department of Bioengineering, University of Pittsburgh, United States; McGowan Institute for Regenerative Medicine, United States
| | - Ipsita Banerjee
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, United States; Department of Bioengineering, University of Pittsburgh, United States; McGowan Institute for Regenerative Medicine, United States.
| |
Collapse
|
17
|
Mendonsa AM, Bandyopadhyay C, Gumbiner BM. p120-catenin phosphorylation status alters E-cadherin mediated cell adhesion and ability of tumor cells to metastasize. PLoS One 2020; 15:e0235337. [PMID: 32589661 PMCID: PMC7319294 DOI: 10.1371/journal.pone.0235337] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 06/12/2020] [Indexed: 01/06/2023] Open
Abstract
p120-catenin is considered to be a tumor suppressor because it stabilizes E-cadherin levels at the cell surface. p120-catenin phosphorylation is increased in several types of cancer, but the role of phosphorylation in cancer is unknown. The phosphorylation state of p120-catenin is important in controlling E-cadherin homophilic binding strength which maintains epithelial junctions. Because decreased cell-cell adhesion is associated with increased cancer metastasis we hypothesize that p120-catenin phosphorylation at specific Serine and Threonine residues alters the E-cadherin binding strength between tumor cells and thereby affect the ability of tumor cells to leave the primary tumor and metastasize to distant sites. In this study we show that expression of the p120-catenin phosphorylation dead mutant, by converting six Serine and Threonine sites to Alanine, leads to enhanced E-cadherin adhesive binding strength in tumor cells. We observed a decrease in the ability of tumor cells expressing the p120-catenin phosphorylation mutant to migrate and invade using in-vitro models of cancer progression. Further, tumor cells expressing the phosphorylation mutant form of p120-catenin demonstrated a decrease in ability to metastasize to the lungs using an in-vivo orthotopic mammary fat pad injection model of breast cancer development and metastasis. This suggests that regulation of p120-catenin phosphorylation at the cell surface is important in mediating cell-adhesion, thereby impacting cancer progression and metastasis.
Collapse
Affiliation(s)
- Alisha M. Mendonsa
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Chirosree Bandyopadhyay
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Barry M. Gumbiner
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- Department of Biochemistry, University of Washington, Seattle, Washington, United States of America
- Department of Pediatrics, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
18
|
Comparative effects of N-cadherin protein and peptide fragments on mesenchymal stem cell mechanotransduction and paracrine function. Biomaterials 2020; 239:119846. [DOI: 10.1016/j.biomaterials.2020.119846] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 02/04/2020] [Accepted: 02/04/2020] [Indexed: 12/16/2022]
|
19
|
Arora P, Dongre S, Raman R, Sonawane M. Stepwise polarisation of developing bilayered epidermis is mediated by aPKC and E-cadherin in zebrafish. eLife 2020; 9:49064. [PMID: 31967543 PMCID: PMC6975926 DOI: 10.7554/elife.49064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 12/13/2019] [Indexed: 12/20/2022] Open
Abstract
The epidermis, a multilayered epithelium, surrounds and protects the vertebrate body. It develops from a bilayered epithelium formed of the outer periderm and underlying basal epidermis. How apicobasal polarity is established in the developing epidermis has remained poorly understood. We show that both the periderm and the basal epidermis exhibit polarised distribution of adherens junctions in zebrafish. aPKC, an apical polarity regulator, maintains the robustness of polarisation of E-cadherin- an adherens junction component- in the periderm. E-cadherin in one layer controls the localisation of E-cadherin in the second layer in a layer non-autonomous manner. Importantly, E-cadherin controls the localisation and levels of Lgl, a basolateral polarity regulator, in a layer autonomous as well non-autonomous manner. Since periderm formation from the enveloping layer precedes the formation of the basal epidermis, our analyses suggest that peridermal polarity, initiated by aPKC, is transduced in a stepwise manner by E-cadherin to the basal layer.
Collapse
Affiliation(s)
- Prateek Arora
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Shivali Dongre
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Renuka Raman
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Mahendra Sonawane
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
20
|
S-nitrosylation and its role in breast cancer angiogenesis and metastasis. Nitric Oxide 2019; 87:52-59. [PMID: 30862477 DOI: 10.1016/j.niox.2019.03.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 01/23/2019] [Accepted: 03/06/2019] [Indexed: 12/24/2022]
Abstract
S-nitrosylation, the modification by nitric oxide of free sulfhydryl groups in cysteines, has become an important regulatory mechanism in carcinogenesis and metastasis. S-nitrosylation of targets in tumor cells contributes to metastasis regulating epithelial to mesenchymal transition, migration and invasion. In the tumor environment, the role of S-nitrosylation in endothelium has not been addressed; however, the evidence points out that S-nitrosylation of endothelial proteins may regulate angiogenesis, adhesion of tumor cells to the endothelium, intra and extravasation of tumor cells and contribute to metastasis.
Collapse
|
21
|
Tiwari P, Mrigwani A, Kaur H, Kaila P, Kumar R, Guptasarma P. Structural-Mechanical and Biochemical Functions of Classical Cadherins at Cellular Junctions: A Review and Some Hypotheses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1112:107-138. [DOI: 10.1007/978-981-13-3065-0_9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
22
|
Gaber A, Kim SJ, Kaake RM, Benčina M, Krogan N, Šali A, Pavšič M, Lenarčič B. EpCAM homo-oligomerization is not the basis for its role in cell-cell adhesion. Sci Rep 2018; 8:13269. [PMID: 30185875 PMCID: PMC6125409 DOI: 10.1038/s41598-018-31482-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 08/20/2018] [Indexed: 01/01/2023] Open
Abstract
Cell-surface tumor marker EpCAM plays a key role in proliferation, differentiation and adhesion processes in stem and epithelial cells. It is established as a cell-cell adhesion molecule, forming intercellular interactions through homophilic association. However, the mechanism by which such interactions arise has not yet been fully elucidated. Here, we first show that EpCAM monomers do not associate into oligomers that would resemble an inter-cellular homo-oligomer, capable of mediating cell-cell adhesion, by using SAXS, XL-MS and bead aggregation assays. Second, we also show that EpCAM forms stable dimers on the surface of a cell with pre-formed cell-cell contacts using FLIM-FRET; however, no inter-cellular homo-oligomers were detectable. Thus, our study provides clear evidence that EpCAM indeed does not function as a homophilic cell adhesion molecule and therefore calls for a significant revision of its role in both normal and cancerous tissues. In the light of this, we strongly support the previously suggested name Epithelial Cell Activating Molecule instead of the Epithelial Cell Adhesion Molecule.
Collapse
Affiliation(s)
- Aljaž Gaber
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, Ljubljana, SI 1000, Slovenia
| | - Seung Joong Kim
- Department of Bioengineering and Therapeutic Sciences, Department of Pharmaceutical Chemistry, California Institute for Quantitative Biosciences, University of California, San Francisco, 1700 4th Street, Suite 503B, San Francisco, CA, 94158, USA
| | - Robyn M Kaake
- J. David Gladstone Institutes, San Francisco, CA, 94158, USA
| | - Mojca Benčina
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, Ljubljana, SI 1000, Slovenia
| | - Nevan Krogan
- J. David Gladstone Institutes, San Francisco, CA, 94158, USA
- Quantitative Biosciences Institute, QBI, Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Andrej Šali
- Department of Bioengineering and Therapeutic Sciences, Department of Pharmaceutical Chemistry, California Institute for Quantitative Biosciences, University of California, San Francisco, 1700 4th Street, Suite 503B, San Francisco, CA, 94158, USA
| | - Miha Pavšič
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, Ljubljana, SI 1000, Slovenia.
| | - Brigita Lenarčič
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, Ljubljana, SI 1000, Slovenia.
- Department of Biochemistry, Molecular and Structural Biology, Institute Jožef Stefan, Jamova 39, Ljubljana, SI 1000, Slovenia.
| |
Collapse
|
23
|
Light SEW, Jontes JD. δ-Protocadherins: Organizers of neural circuit assembly. Semin Cell Dev Biol 2017; 69:83-90. [PMID: 28751249 DOI: 10.1016/j.semcdb.2017.07.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/21/2017] [Accepted: 07/21/2017] [Indexed: 02/08/2023]
Abstract
The δ-protocadherins comprise a small family of homophilic cell adhesion molecules within the larger cadherin superfamily. They are essential for neural development as mutations in these molecules give rise to human neurodevelopmental disorders, such as schizophrenia and epilepsy, and result in behavioral defects in animal models. Despite their importance to neural development, a detailed understanding of their mechanisms and the ways in which their loss leads to changes in neural function is lacking. However, recent results have begun to reveal roles for the δ-protocadherins in both regulation of neurogenesis and lineage-dependent circuit assembly, as well as in contact-dependent motility and selective axon fasciculation. These evolutionarily conserved mechanisms could have a profound impact on the robust assembly of the vertebrate nervous system. Future work should be focused on unraveling the molecular mechanisms of the δ-protocadherins and understanding how this family functions broadly to regulate neural development.
Collapse
Affiliation(s)
- Sarah E W Light
- Department of Neuroscience, Neuroscience Graduate Program, Ohio State University, 1060 Carmack Rd., 113 Rightmire Hall, Columbus, OH 43210, United States
| | - James D Jontes
- Department of Neuroscience, Neuroscience Graduate Program, Ohio State University, 1060 Carmack Rd., 113 Rightmire Hall, Columbus, OH 43210, United States.
| |
Collapse
|
24
|
Bays JL, Campbell HK, Heidema C, Sebbagh M, DeMali KA. Linking E-cadherin mechanotransduction to cell metabolism through force-mediated activation of AMPK. Nat Cell Biol 2017; 19:724-731. [PMID: 28553939 PMCID: PMC5494977 DOI: 10.1038/ncb3537] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 04/24/2017] [Indexed: 02/08/2023]
Abstract
The response of cells to mechanical force is a major determinant of cell behaviour and is an energetically costly event. How cells derive energy to resist mechanical force is unknown. Here, we show that application of force to E-cadherin stimulates liver kinase B1 (LKB1) to activate AMP-activated protein kinase (AMPK), a master regulator of energy homeostasis. LKB1 recruits AMPK to the E-cadherin mechanotransduction complex, thereby stimulating actomyosin contractility, glucose uptake and ATP production. The increase in ATP provides energy to reinforce the adhesion complex and actin cytoskeleton so that the cell can resist physiological forces. Together, these findings reveal a paradigm for how mechanotransduction and metabolism are linked and provide a framework for understanding how diseases involving contractile and metabolic disturbances arise.
Collapse
Affiliation(s)
- Jennifer L Bays
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | - Hannah K Campbell
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | - Christy Heidema
- Interdisciplinary Graduate Program in Molecular and Cellular Biology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | - Michael Sebbagh
- Centre de Recherche en Cancérologie de Marseille, Aix Marseille University UM105, Institut Paoli Calmettes, UMR7258 CNRS, U1068 INSERM, Cell Polarity, Cell signalling and Cancer-Equipe labellisée Ligue Contre le Cancer, Marseille 13273, France
| | - Kris A DeMali
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA.,Interdisciplinary Graduate Program in Molecular and Cellular Biology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| |
Collapse
|
25
|
Harada Y, Takayama F, Tanabe K, Ni J, Hayashi Y, Yamamoto K, Wu Z, Nakanishi H. Overexpression of Cathepsin E Interferes with Neuronal Differentiation of P19 Embryonal Teratocarcinoma Cells by Degradation of N-cadherin. Cell Mol Neurobiol 2017; 37:437-443. [PMID: 27116544 PMCID: PMC11482233 DOI: 10.1007/s10571-016-0376-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/16/2016] [Indexed: 11/24/2022]
Abstract
Cathepsin E (CatE), an aspartic protease, has a limited distribution in certain cell types such as gastric cells. CatE is not detectable in the normal brain, whereas it is increasingly expressed in damaged neurons and activated microglia of the pathological brain. Neurons expressing high levels of CatE showed apparent morphological changes, including a marked shrinkage of the cytoplasmic region and beading of neurites, suggesting neuronal damage. The intracellular level of CatE in neurons is strictly regulated at both transcriptional and translational levels. Although the up-regulation of CatE may cause pathological changes in neurons, little information is available about the precise outcome of the increased expression of CatE in neurons. In this study, we have attempted to clarify the outcome of up-regulated CatE gene expression in neurons using the P19 cell neuronal differentiation after the overexpression of CatE. We unexpectedly found that the overexpression of CatE interfered with neuronal differentiation of P19 cells through an impairment of cell aggregate formation. Pepstatin A, an aspartic protease inhibitor, restored the impaired cell aggregation of P19/CatE cells. The small number of P19 cells differentiated into neurons had abnormal morphology characterized by their fusiform cell bodies with short processes. Furthermore, CatE proteolytically cleaved the extracellular domain of N-cadherin. These observations suggest that the overexpression of CatE interferes with neuronal differentiation of P19 cells through an impairment of cell aggregate formation, possibly through proteolytic degradation of N-cadherin.
Collapse
Affiliation(s)
- Yuka Harada
- Department of Aging Science and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Fumiko Takayama
- Department of Aging Science and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka, 812-8582, Japan
| | | | - Junjun Ni
- Department of Aging Science and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yoshinori Hayashi
- Department of Aging Science and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Kenji Yamamoto
- Proteolysis Research Laboratory, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Zhou Wu
- Department of Aging Science and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Hiroshi Nakanishi
- Department of Aging Science and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka, 812-8582, Japan.
| |
Collapse
|
26
|
Turkowski KL, Tester DJ, Bos JM, Haugaa KH, Ackerman MJ. Whole exome sequencing with genomic triangulation implicatesCDH2-encoded N-cadherin as a novel pathogenic substrate for arrhythmogenic cardiomyopathy. CONGENIT HEART DIS 2017; 12:226-235. [DOI: 10.1111/chd.12462] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 03/01/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Kari L. Turkowski
- Mayo Clinic Graduate School of Biomedical Sciences; Mayo Clinic; Rochester Minnesota USA
| | - David J. Tester
- Department of Molecular Pharmacology & Experimental Therapeutics; Windland Smith Rice Sudden Death Genomics Laboratory; Mayo Clinic; Rochester Minnesota USA
- Department of Cardiovascular Diseases, Division of Heart Rhythm Services; Mayo Clinic; Rochester Minnesota USA
| | - J. Martijn Bos
- Department of Molecular Pharmacology & Experimental Therapeutics; Windland Smith Rice Sudden Death Genomics Laboratory; Mayo Clinic; Rochester Minnesota USA
- Department of Pediatric and Adolescent Medicine, Division of Pediatric Cardiology; Mayo Clinic; Rochester Minnesota USA
| | - Kristina H. Haugaa
- Center for Cardiological Innovation, Department of Cardiology; Institute for Surgical Research, Oslo University Hospital, Rikshospitalet, Oslo Norway and University of Oslo; Oslo Norway
| | - Michael J. Ackerman
- Department of Molecular Pharmacology & Experimental Therapeutics; Windland Smith Rice Sudden Death Genomics Laboratory; Mayo Clinic; Rochester Minnesota USA
- Department of Cardiovascular Diseases, Division of Heart Rhythm Services; Mayo Clinic; Rochester Minnesota USA
- Department of Pediatric and Adolescent Medicine, Division of Pediatric Cardiology; Mayo Clinic; Rochester Minnesota USA
| |
Collapse
|
27
|
Priest AV, Shafraz O, Sivasankar S. Biophysical basis of cadherin mediated cell-cell adhesion. Exp Cell Res 2017; 358:10-13. [PMID: 28300566 DOI: 10.1016/j.yexcr.2017.03.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 03/09/2017] [Indexed: 10/20/2022]
Abstract
Classical cadherin transmembrane cell-cell adhesion proteins play essential roles in tissue morphogenesis and in mediating tissue integrity. Cadherin ectodomains from opposing cells interact to form load-bearing trans dimers that mechanically couple cells. Cell-cell adhesion is believed to be strengthened by cis clustering of cadherins on the same cell surface. This review summarizes biophysical studies of the structure, interaction kinetics and biomechanics of classical cadherin ectodomains. We first discuss the structure and equilibrium binding kinetics of classical cadherin trans and cis dimers. We then discuss how mechanical stimuli alters the kinetics of cadherin interaction and tunes adhesion. Finally, we highlight open questions on the role of mechanical forces in influencing cadherin structure, function and organization on the cell surface.
Collapse
Affiliation(s)
- Andrew Vae Priest
- Department of Physics and Astronomy, Iowa State University, Ames, IA 50011, USA
| | - Omer Shafraz
- Department of Physics and Astronomy, Iowa State University, Ames, IA 50011, USA
| | - Sanjeevi Sivasankar
- Department of Physics and Astronomy, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
28
|
Kadirvelu K, Fathima NN. Self-assembly of keratin peptides: Its implication on the performance of electrospun PVA nanofibers. Sci Rep 2016; 6:36558. [PMID: 27812004 PMCID: PMC5109905 DOI: 10.1038/srep36558] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 10/18/2016] [Indexed: 02/07/2023] Open
Abstract
Drawing inspiration from the field of designer self-assembling materials, this work is aimed to focus on the self-assembling nature of extracted peptides. Hair keratin, a proteinacious reject in tanning industry has been chosen since they have been extracted and used for wide range of applications. Keratin source was subjected to five hydrolysis treatments (viz., sulphitolysis, β-mercaptoethanol, ionic liquid, thioglycolic acid and alkali) and assayed for functional groups. This was followed by the prediction of secondary structure using circular dichroism, determining the microstructural level to which the extracted peptide has self-assembled. Sulphitolysis and thioglycolic acid based hydrolysates exist in monomeric conformation, whereas β-mercaptoethanol based hydrolysate exhibited dimeric conformation. The subsequent part of the study is to incorporate these peptides into the nanofibers to study the structural implication of keratin peptides on its characteristics. Accordingly, the peptides were electrospun with PVA and subjected to morphological, mechanical, thermal and biological characterizations. Monomeric nanofiber mat has high tensile strength of around 5.5 MPa and offered lower mass transport resistance, whereas dimeric mat has high Tm of around 290 °C and was more biocompatible. These results help in understanding the extraction-structure-function aspect of the hydrolysates stressing the role of extraction methods on the choice of application.
Collapse
Affiliation(s)
- Kavitha Kadirvelu
- Chemical laboratory, CSIR-CLRI, Adyar, Chennai -600020, Tamil Nadu, India
| | | |
Collapse
|
29
|
Izaguirre MF, Casco VH. E-cadherin roles in animal biology: A perspective on thyroid hormone-influence. Cell Commun Signal 2016; 14:27. [PMID: 27814736 PMCID: PMC5097364 DOI: 10.1186/s12964-016-0150-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 10/26/2016] [Indexed: 01/15/2023] Open
Abstract
The establishment, remodeling and maintenance of tissular architecture during animal development, and even across juvenile to adult life, are deeply regulated by a delicate interplay of extracellular signals, cell membrane receptors and intracellular signal messengers. It is well known that cell adhesion molecules (cell-cell and cell-extracellular matrix) play a critical role in these processes. Particularly, adherens junctions (AJs) mediated by E-cadherin and catenins determine cell-cell contact survival and epithelia function. Consequently, this review seeks to encompass the complex and prolific knowledge about E-cadherin roles during physiological and pathological states, particularly focusing on the influence exerted by the thyroid hormone (TH).
Collapse
Affiliation(s)
- María Fernanda Izaguirre
- Laboratorio de Microscopia Aplicada a Estudios Moleculares y Celulares, Facultad de Ingeniería (Bioingeniería-Bioinformática), Universidad Nacional de Entre Ríos, Ruta 11, Km 10, Oro Verde, Entre Ríos, Argentina
| | - Victor Hugo Casco
- Laboratorio de Microscopia Aplicada a Estudios Moleculares y Celulares, Facultad de Ingeniería (Bioingeniería-Bioinformática), Universidad Nacional de Entre Ríos, Ruta 11, Km 10, Oro Verde, Entre Ríos, Argentina.
| |
Collapse
|
30
|
Vega L JCM, Lee MK, Qin EC, Rich M, Lee KY, Kim DH, Chung HJ, Leckband DE, Kong H. Three Dimensional Conjugation of Recombinant N-Cadherin to a Hydrogel for In Vitro Anisotropic Neural Growth. J Mater Chem B 2016; 4:6803-6811. [PMID: 28503305 DOI: 10.1039/c6tb01814a] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Living cells are extensively being studied to build functional tissues that are useful for both fundamental and applied bioscience studies. Increasing evidence suggests that cell-cell adhesion controlled by intercellular cadherin junction plays important roles in the quality of the resulting engineered tissue. These findings prompted efforts to interrogate biological effects of cadherin at a molecular scale; however, few efforts were made to harness the effects of cadherin on cells cultured in an in vivo-like three dimensional matrix. To this end, this study reports a hydrogel matrix three dimensionally functionalized with a controlled number of Fc-tagged recombinant N-cadherins (N-Cad-Fc). To retain the desired conformation of N-Cad, these cadherins were immobilized and oriented to the gel by anti-Fc-antibodies chemically coupled to gels. The gels were processed to present N-Cad-Fc in uniaxially aligned microchannels or randomly oriented micropores. Culturing cortical cells in the functionalized gels generated a large fraction of neurons that are functional as indicated by increased intracellular calcium ion concentrations with the microchanneled gel. In contrast, direct N-Cad-Fc immobilization to microchannel or micropore walls of the gel limited the growth of neurons and increased the glial to neuron ratio. The results of this study will be highly useful to organize a wide array of cadherin molecules in a series of biomaterials used for three-dimensional cell culture and to regulate phenotypic activities of tissue-forming cells in an elaborate manner.
Collapse
Affiliation(s)
- Johana C M Vega L
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Min Kyung Lee
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Ellen C Qin
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Max Rich
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Kwan Young Lee
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Dong Hyun Kim
- Department of Human and Culture Convergence Technology R&BD Group, Korea Institute of Industrial Technology, Ansan-si Gyeonggi-do, 426-910 South Korea
| | - Hee Jung Chung
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Deborah E Leckband
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hyunjoon Kong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
31
|
Petrova YI, Schecterson L, Gumbiner BM. Roles for E-cadherin cell surface regulation in cancer. Mol Biol Cell 2016; 27:3233-3244. [PMID: 27582386 PMCID: PMC5170857 DOI: 10.1091/mbc.e16-01-0058] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 08/24/2016] [Indexed: 12/11/2022] Open
Abstract
Loss of E-cadherin expression often occurs in tumors, but many metastases retain E-cadherin. Regulation of the adhesive activity of E-cadherin at the cell surface is important for metastasis of mammary tumor cells, and cancer-associated missense mutations in E-cadherin selectively affect the mechanism of cell surface regulation. The loss of E-cadherin expression in association with the epithelial–mesenchymal transition (EMT) occurs frequently during tumor metastasis. However, metastases often retain E-cadherin expression, an EMT is not required for metastasis, and metastases can arise from clusters of tumor cells. We demonstrate that the regulation of the adhesive activity of E-cadherin present at the cell surface by an inside-out signaling mechanism is important in cancer. First, we find that the metastasis of an E-cadherin–expressing mammary cell line from the mammary gland to the lung depends on reduced E-cadherin adhesive function. An activating monoclonal antibody to E-cadherin that induces a high adhesive state significantly reduced the number of cells metastasized to the lung without affecting the growth in size of the primary tumor in the mammary gland. Second, we find that many cancer-associated germline missense mutations in the E-cadherin gene in patients with hereditary diffuse gastric cancer selectively affect the mechanism of inside-out cell surface regulation without inhibiting basic E-cadherin adhesion function. This suggests that genetic deficits in E-cadherin cell surface regulation contribute to cancer progression. Analysis of these mutations also provides insights into the molecular mechanisms underlying cadherin regulation at the cell surface.
Collapse
Affiliation(s)
- Yuliya I Petrova
- Department of Obstetrics and Gynecology, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Leslayann Schecterson
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101
| | - Barry M Gumbiner
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101 .,Departments of Pediatrics and Biochemistry, University of Washington School of Medicine, Seattle, WA 98195
| |
Collapse
|
32
|
Guequén A, Carrasco R, Zamorano P, Rebolledo L, Burboa P, Sarmiento J, Boric MP, Korayem A, Durán WN, Sánchez FA. S-nitrosylation regulates VE-cadherin phosphorylation and internalization in microvascular permeability. Am J Physiol Heart Circ Physiol 2016; 310:H1039-44. [PMID: 26921435 PMCID: PMC4867340 DOI: 10.1152/ajpheart.00063.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 02/17/2016] [Indexed: 11/22/2022]
Abstract
The adherens junction complex, composed mainly of vascular endothelial (VE)-cadherin, β-catenin, p120, and γ-catenin, is the main element of the endothelial barrier in postcapillary venules.S-nitrosylation of β-catenin and p120 is an important step in proinflammatory agents-induced hyperpermeability. We investigated in vitro and in vivo whether or not VE-cadherin isS-nitrosylated using platelet-activating factor (PAF) as agonist. We report that PAF-stimulates S-nitrosylation of VE-cadherin, which disrupts its association with β-catenin. In addition, based on inhibition of nitric oxide production, our results strongly suggest that S-nitrosylation is required for VE-cadherin phosphorylation on tyrosine and for its internalization. Our results unveil an important mechanism to regulate phosphorylation of junctional proteins in association with S-nitrosylation.
Collapse
Affiliation(s)
- Anita Guequén
- Instituto de Inmunología, Universidad Austral de Chile, Valdivia, Chile
| | - Rodrigo Carrasco
- Instituto de Inmunología, Universidad Austral de Chile, Valdivia, Chile
| | - Patricia Zamorano
- Instituto de Inmunología, Universidad Austral de Chile, Valdivia, Chile
| | - Lorena Rebolledo
- Instituto de Inmunología, Universidad Austral de Chile, Valdivia, Chile
| | - Pia Burboa
- Instituto de Inmunología, Universidad Austral de Chile, Valdivia, Chile
| | - José Sarmiento
- Instituto de Fisiología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Mauricio P Boric
- Departamento de Fisiología, P. Universidad Católica de Chile, Santiago, Chile; and
| | - Adam Korayem
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey
| | - Walter N Durán
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey
| | - Fabiola A Sánchez
- Instituto de Inmunología, Universidad Austral de Chile, Valdivia, Chile;
| |
Collapse
|
33
|
Identification of E-cadherin signature motifs functioning as cleavage sites for Helicobacter pylori HtrA. Sci Rep 2016; 6:23264. [PMID: 26983597 PMCID: PMC4794652 DOI: 10.1038/srep23264] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 03/02/2016] [Indexed: 12/19/2022] Open
Abstract
The cell adhesion protein and tumour suppressor E-cadherin exhibits important functions in the prevention of gastric cancer. As a class-I carcinogen, Helicobacter pylori (H. pylori) has developed a unique strategy to interfere with E-cadherin functions. In previous studies, we have demonstrated that H. pylori secretes the protease high temperature requirement A (HtrA) which cleaves off the E-cadherin ectodomain (NTF) on epithelial cells. This opens cell-to-cell junctions, allowing bacterial transmigration across the polarised epithelium. Here, we investigated the molecular mechanism of the HtrA-E-cadherin interaction and identified E-cadherin cleavage sites for HtrA. Mass-spectrometry-based proteomics and Edman degradation revealed three signature motifs containing the [VITA]-[VITA]-x-x-D-[DN] sequence pattern, which were preferentially cleaved by HtrA. Based on these sites, we developed a substrate-derived peptide inhibitor that selectively bound and inhibited HtrA, thereby blocking transmigration of H. pylori. The discovery of HtrA-targeted signature sites might further explain why we detected a stable 90 kDa NTF fragment during H. pylori infection, but also additional E-cadherin fragments ranging from 105 kDa to 48 kDa in in vitro cleavage experiments. In conclusion, HtrA targets E-cadherin signature sites that are accessible in in vitro reactions, but might be partially masked on epithelial cells through functional homophilic E-cadherin interactions.
Collapse
|
34
|
Miyamoto Y, Sakane F, Hashimoto K. N-cadherin-based adherens junction regulates the maintenance, proliferation, and differentiation of neural progenitor cells during development. Cell Adh Migr 2015; 9:183-92. [PMID: 25869655 DOI: 10.1080/19336918.2015.1005466] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
This review addresses our current understanding of the regulatory mechanism by which N-cadherin, a classical cadherin, affects neural progenitor cells (NPCs) during development. N-cadherin is responsible for the integrity of adherens junctions (AJs), which develop in the sub-apical region of NPCs in the neural tube and brain cortex. The apical domain, which contains the sub-apical region, is involved in the switching from symmetric proliferative division to asymmetric neurogenic division of NPCs. In addition, N-cadherin-based AJ is deeply involved in the apico-basal polarity of NPCs and the regulation of Wnt-β-catenin, hedgehog (Hh), and Notch signaling. In this review, we discuss the roles of N-cadherin in the maintenance, proliferation, and differentiation of NPCs through components of AJ, β-catenin and αE-catenin.
Collapse
Key Words
- AJ, adherens junction
- EC, extracellular
- Fox, forkhead box
- Frz, frizzled
- GFAP, glial fibrillary acidic protein
- GSK3β, glycogen synthase kinase 3β
- Hes, hairly/enhancer of split
- Hh, hedgehog
- IP, intermediate progenitor
- KO, knockout
- LEF, lymphocyte enhancer factor
- N-cadherin
- NPC, neural progenitor cell
- Par, partition defective complex protein
- Ptc, Pached
- Smo, smoothened
- Sox2, sry (sex determining region Y)-box containing gene 2
- TA cell, transient amplifying cell; ZO-1, Zonula Occludens-1.
- TCF, T-cell factor
- aPKC, atypical protein kinase C
- adherens junction
- apico-basal polarity
- iPSC, induced pluripotent stem cell
- neural progenitor cells
- ngn2, neurogenin 2
- shRNA, short hairpin RNA
- β-catenin
Collapse
Affiliation(s)
- Yasunori Miyamoto
- a The Graduate School of Humanities and Sciences; Ochanomizu University ; Tokyo , Japan
| | | | | |
Collapse
|
35
|
Amyloid precursor protein dimerization and synaptogenic function depend on copper binding to the growth factor-like domain. J Neurosci 2014; 34:11159-72. [PMID: 25122912 DOI: 10.1523/jneurosci.0180-14.2014] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Accumulating evidence suggests that the copper-binding amyloid precursor protein (APP) has an essential synaptic function. APP synaptogenic function depends on trans-directed dimerization of the extracellular E1 domain encompassing a growth factor-like domain (GFLD) and a copper-binding domain (CuBD). Here we report the 1.75 Å crystal structure of the GFLD in complex with a copper ion bound with high affinity to an extended hairpin loop at the dimerization interface. In coimmunoprecipitation assays copper binding promotes APP interaction, whereas mutations in the copper-binding sites of either the GFLD or CuBD result in a drastic reduction in APP cis-orientated dimerization. We show that copper is essential and sufficient to induce trans-directed dimerization of purified APP. Furthermore, a mixed culture assay of primary neurons with HEK293 cells expressing different APP mutants revealed that APP potently promotes synaptogenesis depending on copper binding to the GFLD. Together, these findings demonstrate that copper binding to the GFLD of APP is required for APP cis-/trans-directed dimerization and APP synaptogenic function. Thus, neuronal activity or disease-associated changes in copper homeostasis likely go along with altered APP synaptic function.
Collapse
|
36
|
Giant cadherins Fat and Dachsous self-bend to organize properly spaced intercellular junctions. Proc Natl Acad Sci U S A 2014; 111:16011-6. [PMID: 25355906 DOI: 10.1073/pnas.1418990111] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The cadherins Fat and Dachsous regulate cell polarity and proliferation via their heterophilic interactions at intercellular junctions. Their ectodomains are unusually large because of repetitive extracellular cadherin (EC) domains, which raises the question of how they fit in regular intercellular spaces. Cadherins typically exhibit a linear topology through the binding of Ca(2+) to the linker between the EC domains. Our electron-microscopic observations of mammalian Fat4 and Dachsous1 ectodomains, however, revealed that, although their N-terminal regions exhibit a linear configuration, the C-terminal regions are kinked with multiple hairpin-like bends. Notably, certain EC-EC linkers in Fat4 and Dachsous1 lost Ca(2+)-binding amino acids. When such non-Ca(2+)-binding linkers were substituted for a normal linker in E-cadherin, the mutant E-cadherins deformed more extensively than the wild-type molecule. To simulate cadherin structures with non-Ca(2+)-binding linkers, we used an elastic network model and confirmed that bent configurations can be generated by deformation of non-Ca(2+)-binding linkers. These findings suggest that Fat and Dachsous self-bend due to the loss of Ca(2+)-binding amino acids from specific EC-EC linkers, and can therefore adapt to confined spaces.
Collapse
|
37
|
Emond MR, Jontes JD. Bead aggregation assays for the characterization of putative cell adhesion molecules. J Vis Exp 2014:e51762. [PMID: 25350770 DOI: 10.3791/51762] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cell-cell adhesion is fundamental to multicellular life and is mediated by a diverse array of cell surface proteins. However, the adhesive interactions for many of these proteins are poorly understood. Here we present a simple, rapid method for characterizing the adhesive properties of putative homophilic cell adhesion molecules. Cultured HEK293 cells are transfected with DNA plasmid encoding a secreted, epitope-tagged ectodomain of a cell surface protein. Using functionalized beads specific for the epitope tag, the soluble, secreted fusion protein is captured from the culture medium. The coated beads can then be used directly in bead aggregation assays or in fluorescent bead sorting assays to test for homophilic adhesion. If desired, mutagenesis can then be used to elucidate the specific amino acids or domains required for adhesion. This assay requires only small amounts of expressed protein, does not require the production of stable cell lines, and can be accomplished in 4 days.
Collapse
|
38
|
Leonardi E, Sartori S, Vecchi M, Bettella E, Polli R, Palma LD, Boniver C, Murgia A. Identification of four novel PCDH19 Mutations and prediction of their functional impact. Ann Hum Genet 2014; 78:389-98. [PMID: 25227595 DOI: 10.1111/ahg.12082] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 07/10/2014] [Indexed: 11/29/2022]
Abstract
The PCDH19 gene encodes protocadherin-19, a transmembrane protein with six cadherin (EC) domains, containing adhesive interfaces likely to be involved in neuronal connection. Over a hundred mostly private mutations have been identified in girls with epilepsy, with or without intellectual disability (ID). Furthermore, transmitting hemizygous males are devoid of seizures or ID, making it difficult to establish the pathogenic nature of newly identified variants. Here, we describe an integrated approach to evaluate the pathogenicity of four novel PCDH19 mutations. Segregation analysis has been complemented with an in silico analysis of mutation effects at the protein level. Using sequence information, we compared different computational prediction methods. We used homology modeling to build structural models of two PCDH19 EC-domains, and compared wild-type and mutant models to identify differences in residue interactions or biochemical properties of the model surfaces. Our analysis suggests different molecular effects of the novel mutations in exerting their pathogenic role. Two of them interfere with or alter functional residues predicted to mediate ligand or protein binding, one alters the EC-domain folding stability; the frame-shift mutation produces a truncated protein lacking the intracellular domain. Interestingly, the girl carrying the putative loss of function mutation presents the most severe phenotype.
Collapse
Affiliation(s)
- Emanuela Leonardi
- Molecular Genetics of Neurodevelopment, Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Stefano Sartori
- Pediatric Neurology Unit, Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Marilena Vecchi
- Pediatric Neurophysiology Unit, Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Elisa Bettella
- Molecular Genetics of Neurodevelopment, Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Roberta Polli
- Molecular Genetics of Neurodevelopment, Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Luca De Palma
- Pediatric Neurophysiology Unit, Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Clementina Boniver
- Pediatric Neurophysiology Unit, Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Alessandra Murgia
- Molecular Genetics of Neurodevelopment, Department of Women's and Children's Health, University of Padua, Padua, Italy
| |
Collapse
|
39
|
Vega L. JCM, Lee MK, Jeong JH, Smith CE, Lee KY, Chung HJ, Leckband DE, Kong H. Recapitulating Cell–Cell Adhesion Using N-Cadherin Biologically Tethered to Substrates. Biomacromolecules 2014; 15:2172-9. [DOI: 10.1021/bm500335w] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | | | - Jae Hyun Jeong
- Department
of Chemical Engineering, Soongsil University, Seoul, Korea
| | | | | | | | | | - Hyunjoon Kong
- Department
of Chemical Engineering, Soongsil University, Seoul, Korea
| |
Collapse
|
40
|
Shahbazi MN, Megias D, Epifano C, Akhmanova A, Gundersen GG, Fuchs E, Perez-Moreno M. CLASP2 interacts with p120-catenin and governs microtubule dynamics at adherens junctions. ACTA ACUST UNITED AC 2014; 203:1043-61. [PMID: 24368809 PMCID: PMC3871427 DOI: 10.1083/jcb.201306019] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The microtubule plus end–binding protein CLASP2 localizes to adherens junctions via direct interaction with p120-catenin and is required for adherens junction stability. Classical cadherins and their connections with microtubules (MTs) are emerging as important determinants of cell adhesion. However, the functional relevance of such interactions and the molecular players that contribute to tissue architecture are still emerging. In this paper, we report that the MT plus end–binding protein CLASP2 localizes to adherens junctions (AJs) via direct interaction with p120-catenin (p120) in primary basal mouse keratinocytes. Reductions in the levels of p120 or CLASP2 decreased the localization of the other protein to cell–cell contacts and altered AJ dynamics and stability. These features were accompanied by decreased MT density and altered MT dynamics at intercellular junction sites. Interestingly, CLASP2 was enriched at the cortex of basal progenitor keratinocytes, in close localization to p120. Our findings suggest the existence of a new mechanism of MT targeting to AJs with potential functional implications in the maintenance of proper cell–cell adhesion in epidermal stem cells.
Collapse
Affiliation(s)
- Marta N Shahbazi
- Epithelial Cell Biology Laboratory, BBVA (Banco Bilbao Vizcaya Argentaria) Foundation-CNIO (Spanish National Cancer Research Center) Cancer Cell Biology Program; and 2 Confocal Microscopy Unit, Biotechnology Program; CNIO, 28029 Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
41
|
Langenhan J, Dworschak J, Saschenbrecker S, Komorowski L, Schlumberger W, Stöcker W, Westermann J, Recke A, Zillikens D, Schmidt E, Probst C. Specific immunoadsorption of pathogenic autoantibodies in pemphigus requires the entire ectodomains of desmogleins. Exp Dermatol 2014; 23:253-9. [DOI: 10.1111/exd.12355] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2014] [Indexed: 02/05/2023]
Affiliation(s)
- Jana Langenhan
- Institute of Experimental Immunology; Euroimmun AG; Lübeck Germany
| | - Jenny Dworschak
- Department of Dermatology; University of Lübeck; Lübeck Germany
| | | | - Lars Komorowski
- Institute of Experimental Immunology; Euroimmun AG; Lübeck Germany
| | | | - Winfried Stöcker
- Institute of Experimental Immunology; Euroimmun AG; Lübeck Germany
| | | | - Andreas Recke
- Department of Dermatology; University of Lübeck; Lübeck Germany
| | | | - Enno Schmidt
- Department of Dermatology; University of Lübeck; Lübeck Germany
| | - Christian Probst
- Institute of Experimental Immunology; Euroimmun AG; Lübeck Germany
| |
Collapse
|
42
|
Fichtner D, Lorenz B, Engin S, Deichmann C, Oelkers M, Janshoff A, Menke A, Wedlich D, Franz CM. Covalent and density-controlled surface immobilization of E-cadherin for adhesion force spectroscopy. PLoS One 2014; 9:e93123. [PMID: 24675966 PMCID: PMC3968077 DOI: 10.1371/journal.pone.0093123] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 03/02/2014] [Indexed: 11/18/2022] Open
Abstract
E-cadherin is a key cell-cell adhesion molecule but the impact of receptor density and the precise contribution of individual cadherin ectodomains in promoting cell adhesion are only incompletely understood. Investigating these mechanisms would benefit from artificial adhesion substrates carrying different cadherin ectodomains at defined surface density. We therefore developed a quantitative E-cadherin surface immobilization protocol based on the SNAP-tag technique. Extracellular (EC) fragments of E-cadherin fused to the SNAP-tag were covalently bound to self-assembled monolayers (SAM) of thiols carrying benzylguanine (BG) head groups. The adhesive functionality of the different E-cadherin surfaces was then assessed using cell spreading assays and single-cell (SCSF) and single-molecule (SMSF) force spectroscopy. We demonstrate that an E-cadherin construct containing only the first and second outmost EC domain (E1-2) is not sufficient for mediating cell adhesion and yields only low single cadherin-cadherin adhesion forces. In contrast, a construct containing all five EC domains (E1-5) efficiently promotes cell spreading and generates strong single cadherin and cell adhesion forces. By varying the concentration of BG head groups within the SAM we determined a lateral distance of 5–11 nm for optimal E-cadherin functionality. Integrating the results from SCMS and SMSF experiments furthermore demonstrated that the dissolution of E-cadherin adhesion contacts involves a sequential unbinding of individual cadherin receptors rather than the sudden rupture of larger cadherin receptor clusters. Our method of covalent, oriented and density-controlled E-cadherin immobilization thus provides a novel and versatile platform to study molecular mechanisms underlying cadherin-mediated cell adhesion under defined experimental conditions.
Collapse
Affiliation(s)
- Dagmar Fichtner
- Karlsruhe Institute of Technology (KIT), DFG-Center for Functional Nanostructures, Karlsruhe, Germany
| | - Bärbel Lorenz
- University of Göttingen, Institute of Physical Chemistry, Göttingen, Germany
| | - Sinem Engin
- Karlsruhe Institute of Technology (KIT), DFG-Center for Functional Nanostructures, Karlsruhe, Germany
| | - Christina Deichmann
- Karlsruhe Institute of Technology (KIT), DFG-Center for Functional Nanostructures, Karlsruhe, Germany
| | - Marieelen Oelkers
- University of Göttingen, Institute of Physical Chemistry, Göttingen, Germany
| | - Andreas Janshoff
- University of Göttingen, Institute of Physical Chemistry, Göttingen, Germany
| | - Andre Menke
- Justus-Liebig-University Gieβen, Molecular Oncology of Solid Tumors, Gieβen, Germany
| | - Doris Wedlich
- Karlsruhe Institute of Technology (KIT), DFG-Center for Functional Nanostructures, Karlsruhe, Germany
| | - Clemens M. Franz
- Karlsruhe Institute of Technology (KIT), DFG-Center for Functional Nanostructures, Karlsruhe, Germany
- * E-mail:
| |
Collapse
|
43
|
Kurrle N, Völlner F, Eming R, Hertl M, Banning A, Tikkanen R. Flotillins directly interact with γ-catenin and regulate epithelial cell-cell adhesion. PLoS One 2013; 8:e84393. [PMID: 24391950 PMCID: PMC3877284 DOI: 10.1371/journal.pone.0084393] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 11/14/2013] [Indexed: 12/23/2022] Open
Abstract
Flotillin-1 and flotillin-2 are two homologous, membrane raft associated proteins. Although it has been reported that flotillins are involved in cell adhesion processes and play a role during breast cancer progression, thus making them interesting future therapeutic targets, their precise function has not been well elucidated. The present study investigates the function of these proteins in cell-cell adhesion in non-malignant cells. We have used the non-malignant epithelial MCF10A cells to study the interaction network of flotillins within cell-cell adhesion complexes. RNA interference was used to examine the effect of flotillins on the structure of adherens junctions and on the association of core proteins, such as E-cadherin, with membrane rafts. We here show that the cadherin proteins of the adherens junction associate with flotillin-2 in MCF10A cells and in various human cell lines. In vitro, flotillin-1 and flotillin-2 directly interact with γ-catenin which is so far the only protein known to be present both in the adherens junction and the desmosome. Mapping of the interaction domain within the γ-catenin sequence identified the Armadillo domains 6-8, especially ARM domain 7, to be important for the association with flotillins. Furthermore, depletion of flotillins significantly influenced the morphology of the adherens junction in human epithelial MCF10A cells and altered the association of E-cadherin and γ-catenin with membrane rafts. Taken together, these observations suggest a functional role for flotillins, especially flotillin-2, in cell-cell adhesion in non-malignant epithelial cells.
Collapse
Affiliation(s)
- Nina Kurrle
- Institute of Biochemistry, Medical Faculty, Justus Liebig University, Giessen, Germany
| | - Frauke Völlner
- Institute of Biochemistry, Medical Faculty, Justus Liebig University, Giessen, Germany
| | - Rüdiger Eming
- Department of Dermatology and Allergology, Phillips University, Marburg, Germany
| | - Michael Hertl
- Department of Dermatology and Allergology, Phillips University, Marburg, Germany
| | - Antje Banning
- Institute of Biochemistry, Medical Faculty, Justus Liebig University, Giessen, Germany
| | - Ritva Tikkanen
- Institute of Biochemistry, Medical Faculty, Justus Liebig University, Giessen, Germany
- * E-mail:
| |
Collapse
|
44
|
Lee YC, Bilen MA, Yu G, Lin SC, Huang CF, Ortiz A, Cho H, Song JH, Satcher RL, Kuang J, Gallick GE, Yu-Lee LY, Huang W, Lin SH. Inhibition of cell adhesion by a cadherin-11 antibody thwarts bone metastasis. Mol Cancer Res 2013; 11:1401-11. [PMID: 23913163 PMCID: PMC3834228 DOI: 10.1158/1541-7786.mcr-13-0108] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
UNLABELLED Cadherin-11 (CDH11) is a member of the cadherin superfamily mainly expressed in osteoblasts but not in epithelial cells. However, prostate cancer cells with a propensity for bone metastasis express high levels of cadherin-11 and reduced levels of E-cadherin. Downregulation of cadherin-11 inhibits interaction of prostate cancer cells with osteoblasts in vitro and homing of prostate cancer cells to bone in an animal model of metastasis. These findings indicate that targeting cadherin-11 may prevent prostate cancer bone metastasis. To explore this possibility, a panel of 21 monoclonal antibodies (mAb) was generated against the extracellular (EC) domain of cadherin-11. Two antibodies, mAbs 2C7 and 1A5, inhibited cadherin-11-mediated cell-cell aggregation in vitro using L-cells transfected with cadherin-11. Both antibodies demonstrated specificity to cadherin-11, and neither antibody recognized E-cadherin or N-cadherin on C4-2B or PC3 cells, respectively. Furthermore, mAb 2C7 inhibited cadherin-11-mediated aggregation between the highly metastatic PC3-mm2 cells and MC3T3-E1 osteoblasts. Mechanistically, a series of deletion mutants revealed a unique motif, aa 343-348, in the cadherin-11 EC3 domain that is recognized by mAb 2C7 and that this motif coordinated cell-cell adhesion. Importantly, administration of mAb 2C7 in a prophylactic setting effectively prevented metastasis of PC3-mm2 cells to bone in an in vivo mouse model. These results show that targeting the extracellular domain of cadherin-11 can limit cellular adhesion and metastatic dissemination of prostate cancer cells. IMPLICATIONS Monotherapy using a cadherin-11 antibody is a suitable option for the prevention of bone metastases.
Collapse
Affiliation(s)
- Yu-Chen Lee
- Departments of Translational Molecular Pathology, Genitourinary Medical Oncology, Orthopaedic Oncology, Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Mehmet Asim Bilen
- Departments of Translational Molecular Pathology, Genitourinary Medical Oncology, Orthopaedic Oncology, Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Guoyu Yu
- Departments of Translational Molecular Pathology, Genitourinary Medical Oncology, Orthopaedic Oncology, Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Song-Chang Lin
- Departments of Translational Molecular Pathology, Genitourinary Medical Oncology, Orthopaedic Oncology, Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Chih-Fen Huang
- Departments of Translational Molecular Pathology, Genitourinary Medical Oncology, Orthopaedic Oncology, Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
- Department of Pharmacy at National Taiwan University Hospital, Taipei, Taiwan
| | - Angelica Ortiz
- Departments of Translational Molecular Pathology, Genitourinary Medical Oncology, Orthopaedic Oncology, Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Hyojin Cho
- Departments of Translational Molecular Pathology, Genitourinary Medical Oncology, Orthopaedic Oncology, Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Jian H. Song
- Departments of Translational Molecular Pathology, Genitourinary Medical Oncology, Orthopaedic Oncology, Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Robert L. Satcher
- Departments of Translational Molecular Pathology, Genitourinary Medical Oncology, Orthopaedic Oncology, Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Jian Kuang
- Departments of Translational Molecular Pathology, Genitourinary Medical Oncology, Orthopaedic Oncology, Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Gary E. Gallick
- Departments of Translational Molecular Pathology, Genitourinary Medical Oncology, Orthopaedic Oncology, Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Li-Yuan Yu-Lee
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | | | - Sue-Hwa Lin
- Departments of Translational Molecular Pathology, Genitourinary Medical Oncology, Orthopaedic Oncology, Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| |
Collapse
|
45
|
Sivasankar S. Tuning the kinetics of cadherin adhesion. J Invest Dermatol 2013; 133:2318-2323. [PMID: 23812234 PMCID: PMC3773255 DOI: 10.1038/jid.2013.229] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 04/19/2013] [Accepted: 04/24/2013] [Indexed: 12/17/2022]
Abstract
Cadherins are Ca(2+)-dependent cell-cell adhesion proteins that maintain the structural integrity of the epidermis; their principle function is to resist mechanical force. This review summarizes the biophysical mechanisms by which classical cadherins tune adhesion and withstand mechanical stress. We first relate the structure of classical cadherins to their equilibrium binding properties. We then review the role of mechanical perturbations in tuning the kinetics of cadherin adhesion. In particular, we highlight recent studies that show that cadherins form three types of adhesive bonds: catch bonds, which become longer lived and lock in the presence of tensile force; slip bonds, which become shorter lived when pulled; and ideal bonds, which are insensitive to tugging.
Collapse
Affiliation(s)
- Sanjeevi Sivasankar
- Department of Physics and Astronomy, Iowa State University, Ames, Iowa, USA; Ames Laboratory, United States Department of Energy, Ames, Iowa, USA.
| |
Collapse
|
46
|
Biochemical and biophysical origins of cadherin selectivity and adhesion strength. Curr Opin Cell Biol 2012; 24:614-9. [DOI: 10.1016/j.ceb.2012.06.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 06/25/2012] [Accepted: 06/28/2012] [Indexed: 11/21/2022]
|
47
|
Becker SF, Langhe R, Huang C, Wedlich D, Kashef J. Giving the right tug for migration: Cadherins in tissue movements. Arch Biochem Biophys 2012; 524:30-42. [DOI: 10.1016/j.abb.2012.02.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 02/16/2012] [Accepted: 02/17/2012] [Indexed: 01/01/2023]
|
48
|
Tabdili H, Barry AK, Langer MD, Chien YH, Shi Q, Lee KJ, Lu S, Leckband DE. Cadherin point mutations alter cell sorting and modulate GTPase signaling. J Cell Sci 2012; 125:3299-309. [PMID: 22505612 PMCID: PMC3516376 DOI: 10.1242/jcs.087395] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2012] [Indexed: 01/18/2023] Open
Abstract
This study investigated the impact of cadherin binding differences on both cell sorting and GTPase activation. The use of N-terminal domain point mutants of Xenopus C-cadherin enabled us to quantify binding differences and determine their effects on cadherin-dependent functions without any potential complications arising as a result of differences in cytodomain interactions. Dynamic cell-cell binding measurements carried out with the micropipette manipulation technique quantified the impact of these mutations on the two-dimensional binding affinities and dissociation rates of cadherins in the native context of the cell membrane. Pairwise binding affinities were compared with in vitro cell-sorting specificity and ligation-dependent GTPase signaling. Two-dimensional affinity differences greater than five-fold correlated with cadherin-dependent in vitro cell segregation, but smaller differences failed to induce cell sorting. Comparison of the binding affinities with GTPase signaling amplitudes further demonstrated that differential binding also proportionally modulates intracellular signaling. These results show that differential cadherin affinities have broader functional consequences than merely controlling cell-cell cohesion.
Collapse
Affiliation(s)
- Hamid Tabdili
- Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Adrienne K. Barry
- Department of Biochemistry, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Matthew D. Langer
- Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Yuan-Hung Chien
- Department of Biochemistry, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Quanming Shi
- Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Keng Jin Lee
- Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Shaoying Lu
- Department of Bioengineering, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Deborah E. Leckband
- Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana-Champaign, IL 61801, USA
- Department of Biochemistry, University of Illinois, Urbana-Champaign, IL 61801, USA
- Department of Chemistry, University of Illinois, Urbana-Champaign, IL 61801, USA
| |
Collapse
|
49
|
Abstract
Cadherins are Ca(2+)-dependent cell-cell adhesion molecules that play critical roles in animal morphogenesis. Various cadherin-related molecules have also been identified, which show diverse functions, not only for the regulation of cell adhesion but also for that of cell proliferation and planar cell polarity. During the past decade, understanding of the roles of these molecules in the nervous system has significantly progressed. They are important not only for the development of the nervous system but also for its functions and, in turn, for neural disorders. In this review, we discuss the roles of cadherins and related molecules in neural development and function in the vertebrate brain.
Collapse
Affiliation(s)
- Shinji Hirano
- Department of Neurobiology and Anatomy, Kochi Medical School, Okoh-cho Kohasu, Nankoku-City 783–8505, Japan.
| | | |
Collapse
|
50
|
Leckband D, Sivasankar S. Cadherin recognition and adhesion. Curr Opin Cell Biol 2012; 24:620-7. [PMID: 22770731 DOI: 10.1016/j.ceb.2012.05.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2012] [Accepted: 05/23/2012] [Indexed: 01/17/2023]
Abstract
Classical cadherins are the principle adhesive proteins at cohesive intercellular junctions, and are essential proteins for morphogenesis and tissue homeostasis. Because subtype-dependent differences in cadherin adhesion are at the heart of cadherin functions, several structural and biophysical approaches have been used to elucidate relationships between cadherin structures, biophysical properties of cadherin bonds, and cadherin-dependent cell functions. Some experimental approaches appeared to provide conflicting views of the cadherin binding mechanism. However, recent structural and biophysical data, as well as computer simulations generated new insights into classical cadherin binding that increasingly reconcile diverse experimental findings. This review summarizes these recent findings, and highlights both the consistencies and remaining challenges needed to generate a comprehensive model of cadherin interactions that is consistent with all available experimental data.
Collapse
Affiliation(s)
- Deborah Leckband
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States.
| | | |
Collapse
|