1
|
Zhu H, Liu X, Zhang J, Zhao G, Wang J, Zhang H, Liu Y, Guo H, Yang J, Wang Z, Lu TJ, Xu F, Lin M. Cadherin dynamics and cortical tension in remodeling cell-cell adhesion during EMT. Biophys J 2025:S0006-3495(25)00280-2. [PMID: 40329531 DOI: 10.1016/j.bpj.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/30/2025] [Accepted: 05/01/2025] [Indexed: 05/08/2025] Open
Abstract
Epithelial-to-mesenchymal transition (EMT), a key process in cancer metastasis and fibrosis, disrupts cellular adhesion by replacing epithelial E-cadherin with mesenchymal N-cadherin. While, how the shift from E-cadherin to N-cadherin impacts molecular-scale adhesion mechanics and cluster dynamics-and how these changes weaken adhesion under varying mechanical and environmental conditions-remains poorly understood, limiting our ability to target EMT-driven pathological adhesion dynamics. Here, we developed a unified lattice-clutch model to investigate cadherin clustering, cortical tension, and adhesion strength during EMT. Using atomic force microscopy experiments, we measured the mechanical properties of single cadherin trans-bonds and cadherin-mediated cell-cell and cell-matrix adhesions across varying conditions. Our results demonstrate that N-cadherin trans-bonds are mechanically weaker than E-cadherin trans-bonds, leading to reduced adhesion strength during EMT. Computational modeling and experimental validation further revealed that EMT impairs cadherin clustering and cortical tension regulation, which collectively weaken both cell-cell and cell-matrix adhesions, particularly on stiff substrates. These findings highlight how EMT disrupts adhesion strength at multiple scales-from individual cadherin bonds to collective cluster dynamics. Our study elucidates how EMT-driven changes in cadherin type weaken adhesion strength and mechanotransduction, providing insights into cellular adhesion mechanics and potential therapeutic strategies for targeting EMT-associated diseases such as cancer metastasis and tissue remodeling.
Collapse
Affiliation(s)
- Hongyuan Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| | - Xiaoxi Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| | - Jiayu Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| | - Guoqing Zhao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| | - Jin Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| | - Huan Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| | - Yan Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Out-patient Department, School of Stomatology, The Fourth Military Medical University Xi'an, Shaanxi, China
| | - Hui Guo
- Department of Medical Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Jin Yang
- Phase I Clinical Trial Research Center, The First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, P.R. China; Department of Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, P.R. China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Tian Jian Lu
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, P.R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China
| | - Min Lin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, P.R. China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, P.R. China.
| |
Collapse
|
2
|
Alibrandi S, Rinaldi C, Vinci SL, Conti A, Donato L, Scimone C, Sidoti A, D’Angelo R. Mechanotransduction in Development: A Focus on Angiogenesis. BIOLOGY 2025; 14:346. [PMID: 40282211 PMCID: PMC12024848 DOI: 10.3390/biology14040346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025]
Abstract
Cells respond to external mechanical cues and transduce these forces into biological signals. This process is known as mechanotransduction and requires a group of proteins called mechanosensors. This peculiar class of receptors include extracellular matrix proteins, plasma membrane proteins, the cytoskeleton and the nuclear envelope. These cell components are responsive to a wide spectrum of physical cues including stiffness, tensile force, hydrostatic pressure and shear stress. Among mechanotransducers, the Transient Receptor Potential (TRP) and the PIEZO family members are mechanosensitive ion channels, coupling force transduction with intracellular cation transport. Their activity contributes to embryo development, tissue remodeling and repair, and cell homeostasis. In particular, vessel development is driven by hemodynamic cues such as flow direction and shear stress. Perturbed mechanotransduction is involved in several pathological vascular phenotypes including hereditary hemorrhagic telangiectasia. This review is conceived to summarize the most recent findings of mechanotransduction in development. We first collected main features of mechanosensitive proteins. However, we focused on the role of mechanical cues during development. Mechanosensitive ion channels and their function in vascular development are also discussed, with a focus on brain vessel morphogenesis.
Collapse
Affiliation(s)
- Simona Alibrandi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Istituto Euro-Mediterraneo di Scienza e Tecnologia (I.E.ME.S.T.), Street Michele Miraglia 20, 90139 Palermo, Italy
| | - Carmela Rinaldi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
| | - Sergio Lucio Vinci
- Neuroradiology Unit, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
| | - Alfredo Conti
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, Street Altura 3, 40123 Bologna, Italy
- Department of Biomedical and NeuroMotor Sciences (DiBiNeM), Alma Mater Studiorum—University of Bologna, 40127 Bologna, Italy
| | - Luigi Donato
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Istituto Euro-Mediterraneo di Scienza e Tecnologia (I.E.ME.S.T.), Street Michele Miraglia 20, 90139 Palermo, Italy
| | - Concetta Scimone
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
| | - Antonina Sidoti
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Istituto Euro-Mediterraneo di Scienza e Tecnologia (I.E.ME.S.T.), Street Michele Miraglia 20, 90139 Palermo, Italy
| | - Rosalia D’Angelo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
| |
Collapse
|
3
|
Liu S, Meng Y, Lan X, Li R, Kanchanawong P. Ground-state pluripotent stem cells are characterized by Rac1-dependent cadherin-enriched F-actin complexes. J Cell Sci 2025; 138:JCS263811. [PMID: 39886806 DOI: 10.1242/jcs.263811] [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: 08/17/2023] [Accepted: 01/24/2025] [Indexed: 02/01/2025] Open
Abstract
Pluripotent stem cells (PSCs) exhibit extraordinary differentiation potential and are thus highly valuable cellular model systems. However, although different PSC types corresponding to distinct stages of embryogenesis have been in common use, aspects of their cellular architecture and mechanobiology remain insufficiently understood. Here, we investigated how the actin cytoskeleton is regulated in different pluripotency states. We observed a drastic reorganization during the transition from ground-state naïve mouse embryonic stem cells (mESCs) into converted prime epiblast stem cells (EpiSCs). mESCs are characterized by prominent actin-enriched cortical structures that contain cadherin-based cell-cell junctional components, despite not locating at cell-cell junctions. We term these structures 'non-junctional cadherin complexes' (NJCCs) and show that they are under low mechanical tension, depend on the ectodomain but not the cytoplasmic domain of E-cadherin, and exhibit minimal Ca2+ dependence. Active Rac1 was identified as a negative regulator that promotes β-catenin dissociation and NJCC fragmentation. Our data suggests that NJCCs might arise from the cis-association of E-cadherin ectodomain, with potential roles in ground-state pluripotency, and could serve as structural markers to distinguish heterogeneous population of pluripotent stem cells.
Collapse
Affiliation(s)
- Shiying Liu
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Republic of Singapore
| | - Yue Meng
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Republic of Singapore
| | - Xi Lan
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Republic of Singapore
| | - Rong Li
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Republic of Singapore
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore 117558, Republic of Singapore
| | - Pakorn Kanchanawong
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Republic of Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
| |
Collapse
|
4
|
Morano NC, Lopez DH, Meltzer H, Sergeeva AP, Katsamba PS, Rostam KD, Gupta HP, Becker JE, Bornstein B, Cosmanescu F, Schuldiner O, Honig B, Mann RS, Shapiro L. Members of the DIP and Dpr adhesion protein families use cis inhibition to shape neural development in Drosophila. PLoS Biol 2025; 23:e3003030. [PMID: 40029885 DOI: 10.1371/journal.pbio.3003030] [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: 10/02/2024] [Revised: 03/19/2025] [Accepted: 01/22/2025] [Indexed: 03/21/2025] Open
Abstract
In Drosophila, two interacting adhesion protein families, Defective proboscis responses (Dprs) and Dpr interacting proteins (DIPs), coordinate the assembly of neural networks. While intercellular DIP::Dpr interactions have been well characterized, DIPs and Dprs are often co-expressed within the same cells, raising the question as to whether they also interact in cis. We show, in cultured cells and in vivo, that DIP-α and DIP-δ can interact in cis with their ligands, Dpr6/10 and Dpr12, respectively. When co-expressed in cis with their cognate partners, these Dprs regulate the extent of trans binding, presumably through competitive cis interactions. We demonstrate the neurodevelopmental effects of cis inhibition in fly motor neurons and in the mushroom body. We further show that a long disordered region of DIP-α at the C-terminus is required for cis but not trans interactions, likely because it alleviates geometric constraints on cis binding. Thus, the balance between cis and trans interactions plays a role in controlling neural development.
Collapse
Affiliation(s)
- Nicholas C Morano
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
- Aaron Diamond AIDS Research Center, Columbia University, New York, New York, United States of America
| | - Davys H Lopez
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
- Department of Genetics and Development, Columbia University, New York, New York, United States of America
| | - Hagar Meltzer
- Department of Molecular Cell Biology and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Alina P Sergeeva
- Department of Systems Biology, Columbia University, New York, New York, United States of America
| | - Phinikoula S Katsamba
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
| | - Kevin D Rostam
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York, United States of America
| | - Himanshu Pawankumar Gupta
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
| | - Jordan E Becker
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
- Aaron Diamond AIDS Research Center, Columbia University, New York, New York, United States of America
| | - Bavat Bornstein
- Department of Molecular Cell Biology and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Filip Cosmanescu
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
| | - Oren Schuldiner
- Department of Molecular Cell Biology and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Barry Honig
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
- Department of Systems Biology, Columbia University, New York, New York, United States of America
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York, United States of America
- Department of Medicine, Columbia University, New York, New York, United States of America
| | - Richard S Mann
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
- Department of Systems Biology, Columbia University, New York, New York, United States of America
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York, United States of America
| | - Lawrence Shapiro
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
- Aaron Diamond AIDS Research Center, Columbia University, New York, New York, United States of America
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York, United States of America
| |
Collapse
|
5
|
Troyanovsky RB, Indra I, Troyanovsky SM. Actin-dependent α-catenin oligomerization contributes to adherens junction assembly. Nat Commun 2025; 16:1801. [PMID: 39979305 PMCID: PMC11842732 DOI: 10.1038/s41467-025-57079-z] [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: 03/04/2024] [Accepted: 02/07/2025] [Indexed: 02/22/2025] Open
Abstract
Classic cadherins, specifically E-cadherin in most epithelial cells, are transmembrane adhesion receptors, whose intracellular region interacts with proteins, termed catenins, forming the cadherin-catenin complex (CCC). The cadherin ectodomain generates 2D adhesive clusters (E-clusters) through cooperative trans and cis interactions, while catenins anchor the E-clusters to the actin cytoskeleton. How these two types of interactions are coordinated in the formation of specialized cell-cell adhesions, adherens junctions (AJ), remains unclear. Here, we focus on the role of the actin-binding domain of α-catenin (αABD) by showing that the interaction of the αABD with actin generates actin-bound linear CCC oligomers (CCC/actin strands) incorporating up to six CCCs. This actin-driven CCC oligomerization, which is cadherin ectodomain independent, preferentially occurs along the actin cortex enriched with key basolateral proteins, myosin-1c, scribble, and DLG1. In cell-cell contacts, the CCC/actin strands integrate with the E-clusters giving rise to the composite oligomers, E/actin clusters. Targeted inactivation of strand formation by point mutations emphasizes the importance of this oligomerization process for blocking intercellular protrusive membrane activity and for coupling AJs with the actomyosin-derived tensional forces.
Collapse
Affiliation(s)
- Regina B Troyanovsky
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Indrajyoti Indra
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Sergey M Troyanovsky
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL, 60611, USA.
- Department of Cell & Developmental Biology, The Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
6
|
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
|
7
|
Perin C, Cretin G, Gelly JC. Hierarchical Analysis of Protein Structures: From Secondary Structures to Protein Units and Domains. Methods Mol Biol 2025; 2870:357-370. [PMID: 39543044 DOI: 10.1007/978-1-0716-4213-9_18] [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: 11/17/2024]
Abstract
The three-dimensional structure of proteins is traditionally organized into hierarchical levels, specifically secondary structures and domains. However, different studies suggest the existence of intermediate levels, such as Protein Units (PUs), which provide a refined understanding of protein architecture. PUs, characterized by their compactness and independence, serve as an intermediate organizational level, bridging the gap between secondary structures and domains. This new view not only enhances our comprehension of protein structure, folding, and evolutionary mechanisms but also provides a robust methodology for identifying and categorizing protein domains. Based on the concept of PUs, alternative structural partitioning solutions can be proposed that address the structural ambiguity of proteins, leading to more meaningful domain identification.
Collapse
Affiliation(s)
- Charlotte Perin
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France
| | | | | |
Collapse
|
8
|
Mariuzza RA, Singh P, Karade SS, Shahid S, Sharma VK. Recognition of Self and Viral Ligands by NK Cell Receptors. Immunol Rev 2025; 329:e13435. [PMID: 39748148 PMCID: PMC11695704 DOI: 10.1111/imr.13435] [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: 11/26/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025]
Abstract
Natural killer (NK) cells are essential elements of the innate immune response against tumors and viral infections. NK cell activation is governed by NK cell receptors that recognize both cellular (self) and viral (non-self) ligands, including MHC, MHC-related, and non-MHC molecules. These diverse receptors belong to two distinct structural families, the C-type lectin superfamily and the immunoglobulin superfamily. NK receptors include Ly49s, KIRs, LILRs, and NKG2A/CD94, which bind MHC class I (MHC-I) molecules, and NKG2D, which binds MHC-I paralogs such MICA and ULBP. Other NK receptors recognize tumor-associated antigens (NKp30, NKp44, NKp46), cell-cell adhesion proteins (KLRG1, CD96), or genetically coupled C-type lectin-like ligands (NKp65, NKR-P1). Additionally, cytomegaloviruses have evolved various immunoevasins, such as m157, m12, and UL18, which bind NK receptors and act as decoys to enable virus-infected cells to escape NK cell-mediated lysis. We review the remarkable progress made in the past 25 years in determining structures of representatives of most known NK receptors bound to MHC, MHC-like, and non-MHC ligands. Together, these structures reveal the multiplicity of solutions NK receptors have developed to recognize these molecules, and thereby mediate crucial interactions for regulating NK cytolytic activity by self and viral ligands.
Collapse
Affiliation(s)
- Roy A. Mariuzza
- W. M. Keck Laboratory for Structural BiologyUniversity of Maryland Institute for Bioscience and Biotechnology ResearchRockvilleMarylandUSA
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkMarylandUSA
| | - Pragya Singh
- W. M. Keck Laboratory for Structural BiologyUniversity of Maryland Institute for Bioscience and Biotechnology ResearchRockvilleMarylandUSA
- College of Natural and Mathematical SciencesUniversity of MarylandCollege ParkMarylandUSA
| | - Sharanbasappa S. Karade
- W. M. Keck Laboratory for Structural BiologyUniversity of Maryland Institute for Bioscience and Biotechnology ResearchRockvilleMarylandUSA
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkMarylandUSA
| | - Salman Shahid
- W. M. Keck Laboratory for Structural BiologyUniversity of Maryland Institute for Bioscience and Biotechnology ResearchRockvilleMarylandUSA
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkMarylandUSA
| | - Vijay Kumar Sharma
- W. M. Keck Laboratory for Structural BiologyUniversity of Maryland Institute for Bioscience and Biotechnology ResearchRockvilleMarylandUSA
- Department of Cell Biology and Molecular GeneticsUniversity of MarylandCollege ParkMarylandUSA
| |
Collapse
|
9
|
Zheng Y, Zhao J, Nie X, Chitrakar B, Gao J, Sang Y. Mutual adhesion of Lactobacillus spp. to intestinal cells: A review of perspectives on surface layer proteins and cell surface receptors. Int J Biol Macromol 2024; 282:137031. [PMID: 39476894 DOI: 10.1016/j.ijbiomac.2024.137031] [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: 08/11/2024] [Revised: 10/22/2024] [Accepted: 10/27/2024] [Indexed: 11/10/2024]
Abstract
The bacterial ability to adhere and colonize in the gut is a key prerequisite to become a probiotic. Lactobacillus spp. surface layer proteins (SLPs) play an important role for such functions in the human body. Interestingly, all SLPs in spite of their structural variation promote adhesion and colonization. A clear understanding about the binding sites of SLPs with the host and their binding modes would help to precisely reveal the process of Lactobacillus spp.-host interaction. Therefore, in this paper, we have sorted out the Lactobacillus spp. SLPs and their adhesion sites in human intestinal cells. Such SLPs included surface layer protein, motif proteins, binding proteins and moonlighting proteins, while enterocyte adhesion receptors included transmembrane glycoproteins and extracellular matrix proteins. We also summarized the tools to assess the adhesion by Lactobacillus spp. Finally, we recommended that three-dimensional cell models and intestinal microarrays could be major tools for assessing adhesion in the future.
Collapse
Affiliation(s)
- Yixin Zheng
- Hebei Agricultural University, No.2596, Lekai South Street, Baoding, Hebei 86-071000, China
| | - Jinrong Zhao
- Hebei Agricultural University, No.2596, Lekai South Street, Baoding, Hebei 86-071000, China
| | - Xinyu Nie
- Hebei Agricultural University, No.2596, Lekai South Street, Baoding, Hebei 86-071000, China
| | - Bimal Chitrakar
- Hebei Agricultural University, No.2596, Lekai South Street, Baoding, Hebei 86-071000, China
| | - Jie Gao
- Hebei Agricultural University, No.2596, Lekai South Street, Baoding, Hebei 86-071000, China.
| | - Yaxin Sang
- Hebei Agricultural University, No.2596, Lekai South Street, Baoding, Hebei 86-071000, China
| |
Collapse
|
10
|
Chen T, Karedla N, Enderlein J. Observation of E-cadherin adherens junction dynamics with metal-induced energy transfer imaging and spectroscopy. Commun Biol 2024; 7:1596. [PMID: 39613901 DOI: 10.1038/s42003-024-07281-4] [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: 06/07/2024] [Accepted: 11/15/2024] [Indexed: 12/01/2024] Open
Abstract
Epithelial cadherin (E-cad) mediated cell-cell junctions play a crucial role in the establishment and maintenance of tissues and organs. In this study, we employed metal-induced energy transfer imaging and spectroscopy to investigate variations in intermembrane distance during adhesion between two model membranes adorned with E-cad. By correlating the measured intermembrane distances with the distinct E-cad junction states, we probed the dynamic behavior and diversity of E-cad junctions across different binding pathways. Our observations led to the identification of a transient intermediate state referred to as the X-dimeric state and enabled a detailed analysis of its kinetics. We discovered that the formation of the X-dimer leads to significant membrane displacement, subsequently impacting the formation of other X-dimers. These direct experimental insights into the subtle dynamics of E-cad-modified membranes and the resultant changes in intermembrane distance provide perspectives on the assembly of E-cad junctions between cells. This knowledge enhances our comprehension of tissue and organ development and may serve as a foundation for the development of innovative therapeutic strategies for diseases linked to cell-cell adhesion abnormalities.
Collapse
Affiliation(s)
- Tao Chen
- Third Institute of Physics-Biophysics, Georg August University, Göttingen, Germany.
| | - Narain Karedla
- The Rosalind Franklin Institute, Didcot, UK
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Jörg Enderlein
- Third Institute of Physics-Biophysics, Georg August University, Göttingen, Germany.
- Cluster of Excellence 'Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells' (MBExC), Universitätsmedizin Göttingen, Göttingen, Germany.
| |
Collapse
|
11
|
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
|
12
|
Strutt H, Meshram D, Manning E, Madathil ACK, Strutt D. Fat-Dachsous planar polarity function requires two distinct heterophilic cadherin-cadherin binding interactions. Cell Rep 2024; 43:114722. [PMID: 39302834 PMCID: PMC11497213 DOI: 10.1016/j.celrep.2024.114722] [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: 04/17/2024] [Revised: 07/24/2024] [Accepted: 08/20/2024] [Indexed: 09/22/2024] Open
Abstract
Fat and Dachsous are evolutionarily conserved atypical cadherins that regulate polarized cell behaviors. In the Drosophila wing, they interact heterophilically between neighboring cells, localize asymmetrically to opposite cell ends, and control wing shape by regulating oriented cell rearrangements and divisions. Fat and Dachsous have 34 and 27 cadherin repeats, respectively, and previous work has identified trans interactions between their first four cadherin repeats. Here, we identify a second heterophilic binding site in their C-terminal cadherin repeats and show the conservation of this binding site in human Fat4 and Dachsous1. We provide evidence that both N- and C-terminal binding sites regulate the stability of Fat-Dachsous binding interactions and show that the N-terminal binding sites are partly dispensable for Fat-Dachsous function in vivo. Finally, we provide in vivo confirmation that the N-terminal repeats interact in an anti-parallel manner. We propose that multiple binding sites promote the clustering of Fat and Dachsous into a lattice-like array.
Collapse
Affiliation(s)
- Helen Strutt
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK.
| | - Dipak Meshram
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Elizabeth Manning
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | | | - David Strutt
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK.
| |
Collapse
|
13
|
Göz M, Pohl G, Steinecker SM, Walhorn V, Milting H, Anselmetti D. Arrhythmogenic cardiomyopathy-related cadherin variants affect desmosomal binding kinetics. J Mol Cell Cardiol 2024; 195:36-44. [PMID: 39079569 DOI: 10.1016/j.yjmcc.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/09/2024] [Accepted: 07/25/2024] [Indexed: 08/05/2024]
Abstract
Cadherins are calcium dependent adhesion proteins that establish and maintain the intercellular mechanical contact by bridging the gap between adjacent cells. Desmoglein-2 (Dsg2) and desmocollin-2 (Dsc2) are tissue specific cadherin isoforms of the cell-cell contact in cardiac desmosomes. Mutations in the DSG2-gene and in the DSC2-gene are related to arrhythmogenic right ventricular cardiomyopathy (ARVC) a rare but severe heart muscle disease. Here, several possible homophilic and heterophilic binding interactions of wild-type Dsg2, wild-type Dsc2, as well as one Dsg2- and two Dsc2-variants, each associated with ARVC, are investigated. Using single molecule force spectroscopy (SMFS) with atomic force microscopy (AFM) and applying Jarzynski's equality the kinetics and thermodynamics of Dsg2/Dsc2 interaction can be determined. The free energy landscape of Dsg2/Dsc2 dimerization exposes a high activation energy barrier, which is in line with the proposed strand-swapping binding motif. Although the binding motif is not affected by any of the mutations, the binding kinetics of the interactions differ significantly from the wild-type. While wild-type cadherins exhibit an average complex lifetime of approx. 0.3 s interactions involving a variant consistently show - lifetimes that are substantially larger. The lifetimes of the wild-type interactions give rise to the picture of a dynamic adhesion interface consisting of continuously dissociating and (re)associating molecular bonds, while the delayed binding kinetics of interactions involving an ARVC-associated variant might be part of the pathogenesis. Our data provide a comprehensive and consistent thermodynamic and kinetic description of cardiac cadherin binding, allowing detailed insight into the molecular mechanisms of cell adhesion.
Collapse
Affiliation(s)
- Manuel Göz
- Department of Physics, Experimental Biophysics and Applied Nanoscience, Bielefeld University, Universitätsstraße 25, Bielefeld, Germany
| | - Greta Pohl
- Erich & Hanna Klessmann Institute for Cardiovascular Research and Development, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstraße 11, Bad Oeynhausen, Germany
| | - Sylvia M Steinecker
- Department of Physics, Experimental Biophysics and Applied Nanoscience, Bielefeld University, Universitätsstraße 25, Bielefeld, Germany
| | - Volker Walhorn
- Department of Physics, Experimental Biophysics and Applied Nanoscience, Bielefeld University, Universitätsstraße 25, Bielefeld, Germany.
| | - Hendrik Milting
- Erich & Hanna Klessmann Institute for Cardiovascular Research and Development, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstraße 11, Bad Oeynhausen, Germany
| | - Dario Anselmetti
- Department of Physics, Experimental Biophysics and Applied Nanoscience, Bielefeld University, Universitätsstraße 25, Bielefeld, Germany
| |
Collapse
|
14
|
Wiseglass G, Boni N, Smorodinsky-Atias K, Rubinstein R. Clustered protocadherin cis-interactions are required for combinatorial cell-cell recognition underlying neuronal self-avoidance. Proc Natl Acad Sci U S A 2024; 121:e2319829121. [PMID: 38976736 PMCID: PMC11260096 DOI: 10.1073/pnas.2319829121] [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: 11/11/2023] [Accepted: 06/04/2024] [Indexed: 07/10/2024] Open
Abstract
In the developing human brain, only 53 stochastically expressed clustered protocadherin (cPcdh) isoforms enable neurites from individual neurons to recognize and self-avoid while simultaneously maintaining contact with neurites from other neurons. Cell assays have demonstrated that self-recognition occurs only when all cPcdh isoforms perfectly match across the cell boundary, with a single mismatch in the cPcdh expression profile interfering with recognition. It remains unclear, however, how a single mismatched isoform between neighboring cells is sufficient to block erroneous recognitions. Using systematic cell aggregation experiments, we show that abolishing cPcdh interactions on the same membrane (cis) results in a complete loss of specific combinatorial binding between cells (trans). Our computer simulations demonstrate that the organization of cPcdh in linear array oligomers, composed of cis and trans interactions, enhances self-recognition by increasing the concentration and stability of cPcdh trans complexes between the homotypic membranes. Importantly, we show that the presence of mismatched isoforms between cells drastically diminishes the concentration and stability of the trans complexes. Overall, we provide an explanation for the role of the cPcdh assembly arrangements in neuronal self/non-self-discrimination underlying neuronal self-avoidance.
Collapse
Affiliation(s)
- Gil Wiseglass
- Department of Biochemistry and Molecular Biology, School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv6997801, Israel
| | - Nadir Boni
- Department of Biochemistry and Molecular Biology, School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv6997801, Israel
| | - Karina Smorodinsky-Atias
- Department of Biochemistry and Molecular Biology, School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv6997801, Israel
| | - Rotem Rubinstein
- Department of Biochemistry and Molecular Biology, School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv6997801, Israel
| |
Collapse
|
15
|
Morano NC, Lopez DH, Meltzer H, Sergeeva AP, Katsamba PS, Rostam KD, Gupta HP, Becker JE, Bornstein B, Cosmanescu F, Schuldiner O, Honig B, Mann RS, Shapiro L. Cis inhibition of co-expressed DIPs and Dprs shapes neural development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583391. [PMID: 38895375 PMCID: PMC11185508 DOI: 10.1101/2024.03.04.583391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
In Drosophila , two interacting adhesion protein families, Dprs and DIPs, coordinate the assembly of neural networks. While intercellular DIP/Dpr interactions have been well characterized, DIPs and Dprs are often co-expressed within the same cells, raising the question as to whether they also interact in cis . We show, in cultured cells and in vivo, that DIP-α and DIP-δ can interact in cis with their ligands, Dpr6/10 and Dpr12, respectively. When co-expressed in cis with their cognate partners, these Dprs regulate the extent of trans binding, presumably through competitive cis interactions. We demonstrate the neurodevelopmental effects of cis inhibition in fly motor neurons and in the mushroom body. We further show that a long disordered region of DIP-α at the C-terminus is required for cis but not trans interactions, likely because it alleviates geometric constraints on cis binding. Thus, the balance between cis and trans interactions plays a role in controlling neural development.
Collapse
|
16
|
Campàs O, Noordstra I, Yap AS. Adherens junctions as molecular regulators of emergent tissue mechanics. Nat Rev Mol Cell Biol 2024; 25:252-269. [PMID: 38093099 DOI: 10.1038/s41580-023-00688-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2023] [Indexed: 03/28/2024]
Abstract
Tissue and organ development during embryogenesis relies on the collective and coordinated action of many cells. Recent studies have revealed that tissue material properties, including transitions between fluid and solid tissue states, are controlled in space and time to shape embryonic structures and regulate cell behaviours. Although the collective cellular flows that sculpt tissues are guided by tissue-level physical changes, these ultimately emerge from cellular-level and subcellular-level molecular mechanisms. Adherens junctions are key subcellular structures, built from clusters of classical cadherin receptors. They mediate physical interactions between cells and connect biochemical signalling to the physical characteristics of cell contacts, hence playing a fundamental role in tissue morphogenesis. In this Review, we take advantage of the results of recent, quantitative measurements of tissue mechanics to relate the molecular and cellular characteristics of adherens junctions, including adhesion strength, tension and dynamics, to the emergent physical state of embryonic tissues. We focus on systems in which cell-cell interactions are the primary contributor to morphogenesis, without significant contribution from cell-matrix interactions. We suggest that emergent tissue mechanics is an important direction for future research, bridging cell biology, developmental biology and mechanobiology to provide a holistic understanding of morphogenesis in health and disease.
Collapse
Affiliation(s)
- Otger Campàs
- Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany.
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
- Center for Systems Biology Dresden, Dresden, Germany.
| | - Ivar Noordstra
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia
| | - Alpha S Yap
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia.
| |
Collapse
|
17
|
Rabino A, Awadia S, Ali N, Edson A, Garcia-Mata R. The Scribble/SGEF/Dlg1 complex regulates the stability of apical junctions in epithelial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.26.586884. [PMID: 38585765 PMCID: PMC10996629 DOI: 10.1101/2024.03.26.586884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
SGEF, a RhoG specific GEF, can form a ternary complex with the Scribble polarity complex proteins Scribble and Dlg1, which regulates the formation and maintenance of adherens junctions and barrier function of epithelial cells. Notably, silencing SGEF results in a dramatic downregulation of the expression of both E-cadherin and ZO-1. However, the molecular mechanisms involved in the regulation of this pathway are not known. Here, we describe a novel signaling pathway governed by the Scribble/SGEF/Dlg1 complex. Our results show that an intact ternary complex is required to maintain the stability of the apical junctions, the expression of ZO-1, and TJ permeability. In contrast, only SGEF is necessary to regulate E-cadherin expression. The absence of SGEF destabilizes the E-cadherin/catenin complex at the membrane, triggering a positive feedback loop that exacerbates the phenotype through the repression of E-cadherin transcription in a process that involves the internalization of E-cadherin by endocytosis, β-catenin signaling and the transcriptional repressor Slug.
Collapse
Affiliation(s)
- Agustin Rabino
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Sahezeel Awadia
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Nabaa Ali
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Amber Edson
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| | - Rafael Garcia-Mata
- Department of Biological Sciences, University of Toledo, Toledo, OH, USA
| |
Collapse
|
18
|
Troyanovsky RB, Indra I, Troyanovsky SM. Characterization of early and late events of adherens junction assembly. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583373. [PMID: 38496678 PMCID: PMC10942379 DOI: 10.1101/2024.03.04.583373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Cadherins are transmembrane adhesion receptors. Cadherin ectodomains form adhesive 2D clusters through cooperative trans and cis interactions, whereas its intracellular region interacts with specific cytosolic proteins, termed catenins, to anchor the cadherin-catenin complex (CCC) to the actin cytoskeleton. How these two types of interactions are coordinated in the formation of specialized cell-cell adhesions, adherens junctions (AJ), remains unclear. We focus here on the role of the actin-binding domain of α-catenin (αABD) by showing that the interaction of αABD with actin generates actin-bound CCC oligomers (CCC/actin strands) incorporating up to six CCCs. The strands are primarily formed on the actin-rich cell protrusions. Once in cell-cell interface, the strands become involved in cadherin ectodomain clustering. Such combination of the extracellular and intracellular oligomerizations gives rise to the composite oligomers, trans CCC/actin clusters. To mature, these clusters then rearrange their actin filaments using several redundant pathways, two of which are characterized here: one depends on the α-catenin-associated protein, vinculin and the second one depends on the unstructured C-terminus of αABD. Thus, AJ assembly proceeds through spontaneous formation of trans CCC/actin clusters and their successive reorganization.
Collapse
Affiliation(s)
- Regina B Troyanovsky
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611
| | - Indrajyoti Indra
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611
| | - Sergey M Troyanovsky
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611
- Department of Cell & Developmental Biology, The Feinberg School of Medicine, Chicago, IL 60614
| |
Collapse
|
19
|
Tsai YX, Chang NE, Reuter K, Chang HT, Yang TJ, von Bülow S, Sehrawat V, Zerrouki N, Tuffery M, Gecht M, Grothaus IL, Colombi Ciacchi L, Wang YS, Hsu MF, Khoo KH, Hummer G, Hsu STD, Hanus C, Sikora M. Rapid simulation of glycoprotein structures by grafting and steric exclusion of glycan conformer libraries. Cell 2024; 187:1296-1311.e26. [PMID: 38428397 DOI: 10.1016/j.cell.2024.01.034] [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: 10/25/2022] [Revised: 10/18/2023] [Accepted: 01/22/2024] [Indexed: 03/03/2024]
Abstract
Most membrane proteins are modified by covalent addition of complex sugars through N- and O-glycosylation. Unlike proteins, glycans do not typically adopt specific secondary structures and remain very mobile, shielding potentially large fractions of protein surface. High glycan conformational freedom hinders complete structural elucidation of glycoproteins. Computer simulations may be used to model glycosylated proteins but require hundreds of thousands of computing hours on supercomputers, thus limiting routine use. Here, we describe GlycoSHIELD, a reductionist method that can be implemented on personal computers to graft realistic ensembles of glycan conformers onto static protein structures in minutes. Using molecular dynamics simulation, small-angle X-ray scattering, cryoelectron microscopy, and mass spectrometry, we show that this open-access toolkit provides enhanced models of glycoprotein structures. Focusing on N-cadherin, human coronavirus spike proteins, and gamma-aminobutyric acid receptors, we show that GlycoSHIELD can shed light on the impact of glycans on the conformation and activity of complex glycoproteins.
Collapse
Affiliation(s)
- Yu-Xi Tsai
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Ning-En Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Klaus Reuter
- Max Planck Computing and Data Facility, 85748 Garching, Germany
| | - Hao-Ting Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Tzu-Jing Yang
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Sören von Bülow
- Department of Theoretical Biophysics, Max Planck Institute for Biophysics, 60438 Frankfurt, Germany
| | - Vidhi Sehrawat
- Department of Theoretical Biophysics, Max Planck Institute for Biophysics, 60438 Frankfurt, Germany; Malopolska Centre of Biotechnology, Jagiellonian University, 31-007 Kraków, Poland
| | - Noémie Zerrouki
- Institute of Psychiatry and Neurosciences of Paris, Inserm UMR1266, Université Paris-Cité, 75014 Paris, France
| | - Matthieu Tuffery
- Institute of Psychiatry and Neurosciences of Paris, Inserm UMR1266, Université Paris-Cité, 75014 Paris, France
| | - Michael Gecht
- Department of Theoretical Biophysics, Max Planck Institute for Biophysics, 60438 Frankfurt, Germany
| | - Isabell Louise Grothaus
- Hybrid Materials Interfaces Group, Faculty of Production Engineering, Bremen Center for Computational Materials Science and MAPEX Center for Materials and Processes, University of Bremen, 28359 Bremen, Germany
| | - Lucio Colombi Ciacchi
- Hybrid Materials Interfaces Group, Faculty of Production Engineering, Bremen Center for Computational Materials Science and MAPEX Center for Materials and Processes, University of Bremen, 28359 Bremen, Germany
| | - Yong-Sheng Wang
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Min-Feng Hsu
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Kay-Hooi Khoo
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan
| | - Gerhard Hummer
- Department of Theoretical Biophysics, Max Planck Institute for Biophysics, 60438 Frankfurt, Germany; Institute of Biophysics, Goethe University, 60438 Frankfurt, Germany
| | - Shang-Te Danny Hsu
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan; International Institute for Sustainability with Knotted Chiral Meta Matter (WPI-SKCM(2)), Hiroshima University, Hiroshima 739-8526, Japan.
| | - Cyril Hanus
- Institute of Psychiatry and Neurosciences of Paris, Inserm UMR1266, Université Paris-Cité, 75014 Paris, France; GHU Psychiatrie et Neurosciences de Paris, 75014 Paris, France.
| | - Mateusz Sikora
- Department of Theoretical Biophysics, Max Planck Institute for Biophysics, 60438 Frankfurt, Germany; Malopolska Centre of Biotechnology, Jagiellonian University, 31-007 Kraków, Poland.
| |
Collapse
|
20
|
Pereira J, Melo S, Ferreira RM, Carneiro P, Yang V, Maia AF, Carvalho J, Figueiredo C, Machado JC, Morais-de-Sá E, Seruca R, Figueiredo J. E-cadherin variants associated with oral facial clefts trigger aberrant cell motility in a REG1A-dependent manner. Cell Commun Signal 2024; 22:152. [PMID: 38414029 PMCID: PMC10898076 DOI: 10.1186/s12964-024-01532-x] [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: 01/10/2024] [Accepted: 02/13/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Germline mutations of E-cadherin contribute to hereditary diffuse gastric cancer (HDGC) and congenital malformations, such as oral facial clefts (OFC). However, the molecular mechanisms through which E-cadherin loss-of-function triggers distinct clinical outcomes remain unknown. We postulate that E-cadherin-mediated disorders result from abnormal interactions with the extracellular matrix and consequent aberrant intracellular signalling, affecting the coordination of cell migration. METHODS Herein, we developed in vivo and in vitro models of E-cadherin mutants associated with either OFC or HDGC. Using a Drosophila approach, we addressed the impact of the different variants in cell morphology and migration ability. By combining gap closure migration assays and time-lapse microscopy, we further investigated the migration pattern of cells expressing OFC or HDGC variants. The adhesion profile of the variants was evaluated using high-throughput ECM arrays, whereas RNA sequencing technology was explored for identification of genes involved in aberrant cell motility. RESULTS We have demonstrated that cells expressing OFC variants exhibit an excessive motility performance and irregular leading edges, which prevent the coordinated movement of the epithelial monolayer. Importantly, we found that OFC variants promote cell adhesion to a wider variety of extracellular matrices than HDGC variants, suggesting higher plasticity in response to different microenvironments. We unveiled a distinct transcriptomic profile in the OFC setting and pinpointed REG1A as a putative regulator of this outcome. Consistent with this, specific RNAi-mediated inhibition of REG1A shifted the migration pattern of OFC expressing cells, leading to slower wound closure with coordinated leading edges. CONCLUSIONS We provide evidence that E-cadherin variants associated with OFC activate aberrant signalling pathways that support dynamic rearrangements of cells towards improved adaptability to the microenvironment. This proficiency results in abnormal tissue shaping and movement, possibly underlying the development of orofacial malformations.
Collapse
Affiliation(s)
- Joana Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of Porto University, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Soraia Melo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of Porto University, Porto, Portugal
| | - Rui M Ferreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of Porto University, Porto, Portugal
| | - Patrícia Carneiro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of Porto University, Porto, Portugal
| | - Vítor Yang
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- IBMC - Institute for Molecular and Cell Biology, University of Porto, Porto, Portugal
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - André F Maia
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- IBMC - Institute for Molecular and Cell Biology, University of Porto, Porto, Portugal
| | - João Carvalho
- CFisUC, Department of Physics, University of Coimbra, Coimbra, Portugal
| | - Ceu Figueiredo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of Porto University, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - José Carlos Machado
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of Porto University, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Eurico Morais-de-Sá
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- IBMC - Institute for Molecular and Cell Biology, University of Porto, Porto, Portugal
| | - Raquel Seruca
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of Porto University, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Joana Figueiredo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen, 208, Porto, 4200-135, Portugal.
- IPATIMUP - Institute of Molecular Pathology and Immunology of Porto University, Porto, Portugal.
- Faculty of Medicine, University of Porto, Porto, Portugal.
| |
Collapse
|
21
|
Leckband D, Schwartz DK, Wu Y. Computational and experimental approaches to quantify protein binding interactions under confinement. Biophys J 2024; 123:424-434. [PMID: 38245831 PMCID: PMC10912910 DOI: 10.1016/j.bpj.2024.01.018] [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: 09/18/2023] [Revised: 01/03/2024] [Accepted: 01/17/2024] [Indexed: 01/22/2024] Open
Abstract
Crowded environments and confinement alter the interactions of adhesion proteins confined to membranes or narrow, crowded gaps at adhesive contacts. Experimental approaches and theoretical frameworks were developed to quantify protein binding constants in these environments. However, recent predictions and the complexity of some protein interactions proved challenging to address with prior experimental or theoretical approaches. This perspective highlights new methods developed by these authors that address these challenges. Specifically, single-molecule fluorescence resonance energy transfer and single-molecule tracking measurements were developed to directly image the binding/unbinding rates of membrane-tethered cadherins. Results identified predicted cis (lateral) interactions, which control cadherin clustering on membranes but were not detected in solution. Kinetic Monte Carlo simulations, based on a realistic model of cis cadherin interactions, were developed to extract binding/unbinding rate constants from heterogeneous single-molecule data. The extension of single-molecule fluorescence measurements to cis and trans (adhesive) cadherin interactions at membrane junctions identified unexpected cooperativity between cis and trans binding that appears to enhance intercellular binding kinetics. Comparisons of intercellular binding kinetics, kinetic Monte Carlo simulations, and single-molecule fluorescence data suggest a strategy to bridge protein binding kinetics across length scales. Although cadherin is the focus of these studies, the approaches can be extended to other intercellular adhesion proteins.
Collapse
Affiliation(s)
- Deborah Leckband
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois; Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois; Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois.
| | - Daniel K Schwartz
- Chemical and Biomolecular Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Yinghao Wu
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
22
|
Xie B, Xu S, Schecterson L, Gumbiner BM, Sivasankar S. Strengthening E-cadherin adhesion via antibody-mediated binding stabilization. Structure 2024; 32:217-227.e3. [PMID: 38052206 PMCID: PMC10872345 DOI: 10.1016/j.str.2023.11.002] [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: 07/05/2023] [Revised: 09/10/2023] [Accepted: 11/08/2023] [Indexed: 12/07/2023]
Abstract
E-cadherins (Ecads) are a crucial cell-cell adhesion protein with tumor suppression properties. Ecad adhesion can be enhanced by the monoclonal antibody 66E8, which has potential applications in inhibiting cancer metastasis. However, the biophysical mechanisms underlying 66E8-mediated adhesion strengthening are unknown. Here, we use molecular dynamics simulations, site-directed mutagenesis, and single-molecule atomic force microscopy experiments to demonstrate that 66E8 strengthens Ecad binding by stabilizing the primary Ecad adhesive conformation: the strand-swap dimer. By forming electrostatic interactions with Ecad, 66E8 stabilizes the swapped β-strand and its hydrophobic pocket and impedes Ecad conformational changes, which are necessary for rupture of the strand-swap dimer. Our findings identify fundamental mechanistic principles for strengthening of Ecad binding using monoclonal antibodies.
Collapse
Affiliation(s)
- Bin Xie
- Biophysics Graduate Group, University of California, Davis, Davis, CA, USA
| | - Shipeng Xu
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Leslayann Schecterson
- Seattle Children's Research Institute, Center for Developmental Biology and Regenerative Medicine, Seattle, WA, USA
| | - Barry M Gumbiner
- Seattle Children's Research Institute, Center for Developmental Biology and Regenerative Medicine, Seattle, WA, USA
| | - Sanjeevi Sivasankar
- Biophysics Graduate Group, University of California, Davis, Davis, CA, USA; Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
23
|
Su Z, Almo SC, Wu Y. Computational simulations of bispecific T cell engagers by a multiscale model. Biophys J 2024; 123:235-247. [PMID: 38102828 PMCID: PMC10808035 DOI: 10.1016/j.bpj.2023.12.012] [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: 06/08/2023] [Revised: 11/04/2023] [Accepted: 12/12/2023] [Indexed: 12/17/2023] Open
Abstract
The use of bispecific antibodies as T cell engagers can bypass the normal T cell receptor-major histocompatibility class interaction, redirect the cytotoxic activity of T cells, and lead to highly efficient tumor cell killing. However, this immunotherapy also causes significant on-target off-tumor toxicologic effects, especially when it is used to treat solid tumors. To avoid these adverse events, it is necessary to understand the fundamental mechanisms involved in the physical process of T cell engagement. We developed a multiscale computational framework to reach this goal. The framework combines simulations on the intercellular and multicellular levels. On the intercellular level, we simulated the spatial-temporal dynamics of three-body interactions among bispecific antibodies, CD3 and tumor-associated antigens (TAAs). The derived number of intercellular bonds formed between CD3 and TAAs was further transferred to the multicellular simulations as the input parameter of adhesive density between cells. Through the simulations under various molecular and cellular conditions, we were able to gain new insights into how to adopt the most appropriate strategy to maximize the drug efficacy and avoid the off-target effect. For instance, we discovered that the low antibody-binding affinity resulted in the formation of large clusters at the cell-cell interface, which could be important to control the downstream signaling pathways. We also tested different molecular architectures of the bispecific antibody and suggested the existence of an optimal length in regulating the T cell engagement. Overall, the current multiscale simulations serve as a proof-of-concept study to help in the future design of new biological therapeutics.
Collapse
Affiliation(s)
- Zhaoqian Su
- Data Science Institute, Vanderbilt University, Nashville, Tennessee
| | - Steven C Almo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York; Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, New York
| | - Yinghao Wu
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
24
|
Mukherjee S, Goswami S, Dash S, Samanta D. Structural basis of molecular recognition among classical cadherins mediating cell adhesion. Biochem Soc Trans 2023; 51:2103-2115. [PMID: 37970977 DOI: 10.1042/bst20230356] [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: 09/01/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023]
Abstract
Cadherins are type-I membrane glycoproteins that primarily participate in calcium-dependent cell adhesion and homotypic cell sorting in various stages of embryonic development. Besides their crucial role in cellular and physiological processes, increasing studies highlight their involvement in pathophysiological functions ranging from cancer progression and metastasis to being entry receptors for pathogens. Cadherins mediate these cellular processes through homophilic, as well as heterophilic interactions (within and outside the superfamily) by their membrane distal ectodomains. This review provides an in-depth structural perspective of molecular recognition among type-I and type-II classical cadherins. Furthermore, this review offers structural insights into different dimeric assemblies like the 'strand-swap dimer' and 'X-dimer' as well as mechanisms relating these dimer forms like 'two-step adhesion' and 'encounter complex'. Alongside providing structural details, this review connects structural studies to bond mechanics merging crystallographic and single-molecule force spectroscopic findings. Finally, the review discusses the recent discoveries on dimeric intermediates that uncover prospects of further research beyond two-step adhesion.
Collapse
Affiliation(s)
- Sarbartha Mukherjee
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| | - Saumyadeep Goswami
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| | - Sagarika Dash
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| | - Dibyendu Samanta
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| |
Collapse
|
25
|
Zhang N, Häring M, Wolf F, Großhans J, Kong D. Dynamics and functions of E-cadherin complexes in epithelial cell and tissue morphogenesis. MARINE LIFE SCIENCE & TECHNOLOGY 2023; 5:585-601. [PMID: 38045551 PMCID: PMC10689684 DOI: 10.1007/s42995-023-00206-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 10/31/2023] [Indexed: 12/05/2023]
Abstract
Cell-cell adhesion is at the center of structure and dynamics of epithelial tissue. E-cadherin-catenin complexes mediate Ca2+-dependent trans-homodimerization and constitute the kernel of adherens junctions. Beyond the basic function of cell-cell adhesion, recent progress sheds light the dynamics and interwind interactions of individual E-cadherin-catenin complex with E-cadherin superclusters, contractile actomyosin and mechanics of the cortex and adhesion. The nanoscale architecture of E-cadherin complexes together with cis-interactions and interactions with cortical actomyosin adjust to junctional tension and mechano-transduction by reinforcement or weakening of specific features of the interactions. Although post-translational modifications such as phosphorylation and glycosylation have been implicated, their role for specific aspects of in E-cadherin function has remained unclear. Here, we provide an overview of the E-cadherin complex in epithelial cell and tissue morphogenesis focusing on nanoscale architectures by super-resolution approaches and post-translational modifications from recent, in particular in vivo, studies. Furthermore, we review the computational modelling in E-cadherin complexes and highlight how computational modelling has contributed to a deeper understanding of the E-cadherin complexes.
Collapse
Affiliation(s)
- Na Zhang
- Department of Biology, Philipps University, 35043 Marburg, Germany
| | - Matthias Häring
- Göttingen Campus Institute for Dynamics of Biological Networks (CIDBN), Georg August University Göttingen, 37073 Göttingen, Germany
| | - Fred Wolf
- Göttingen Campus Institute for Dynamics of Biological Networks (CIDBN), Georg August University Göttingen, 37073 Göttingen, Germany
| | - Jörg Großhans
- Department of Biology, Philipps University, 35043 Marburg, Germany
- Göttingen Campus Institute for Dynamics of Biological Networks (CIDBN), Georg August University Göttingen, 37073 Göttingen, Germany
| | - Deqing Kong
- Department of Biology, Philipps University, 35043 Marburg, Germany
- Göttingen Campus Institute for Dynamics of Biological Networks (CIDBN), Georg August University Göttingen, 37073 Göttingen, Germany
| |
Collapse
|
26
|
Abstract
Multicellular organisms generate tissues of diverse shapes and functions from cells and extracellular matrices. Their adhesion molecules mediate cell-cell and cell-matrix interactions, which not only play crucial roles in maintaining tissue integrity but also serve as key regulators of tissue morphogenesis. Cells constantly probe their environment to make decisions: They integrate chemical and mechanical information from the environment via diffusible ligand- or adhesion-based signaling to decide whether to release specific signaling molecules or enzymes, to divide or differentiate, to move away or stay, or even whether to live or die. These decisions in turn modify their environment, including the chemical nature and mechanical properties of the extracellular matrix. Tissue morphology is the physical manifestation of the remodeling of cells and matrices by their historical biochemical and biophysical landscapes. We review our understanding of matrix and adhesion molecules in tissue morphogenesis, with an emphasis on key physical interactions that drive morphogenesis.
Collapse
Affiliation(s)
- Di Wu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA;
| | - Kenneth M Yamada
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA;
| | - Shaohe Wang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA;
| |
Collapse
|
27
|
Naturale VF, Pickett MA, Feldman JL. Persistent cell contacts enable E-cadherin/HMR-1- and PAR-3-based symmetry breaking within a developing C. elegans epithelium. Dev Cell 2023; 58:1830-1846.e12. [PMID: 37552986 PMCID: PMC10592304 DOI: 10.1016/j.devcel.2023.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 05/10/2023] [Accepted: 07/17/2023] [Indexed: 08/10/2023]
Abstract
Tissue-wide patterning is essential to multicellular development, requiring cells to individually generate polarity axes and coordinate them in space and time with neighbors. Using the C. elegans intestinal epithelium, we identified a patterning mechanism that is informed by cell contact lifetime asymmetry and executed via the scaffolding protein PAR-3 and the transmembrane protein E-cadherin/HMR-1. Intestinal cells break symmetry as PAR-3 and HMR-1 recruit apical determinants into punctate "local polarity complexes" (LPCs) at homotypic contacts. LPCs undergo an HMR-1-based migration to a common midline, thereby establishing tissue-wide polarity. Thus, symmetry breaking results from PAR-3-dependent intracellular polarization coupled to HMR-1-based tissue-level communication, which occurs through a non-adhesive signaling role for HMR-1. Differential lifetimes between homotypic and heterotypic cell contacts are created by neighbor exchanges and oriented divisions, patterning where LPCs perdure and thereby breaking symmetry. These cues offer a logical and likely conserved framework for how epithelia without obvious molecular asymmetries can polarize.
Collapse
Affiliation(s)
| | - Melissa A Pickett
- Department of Biology, Stanford University, Stanford, CA 94305, USA; Department of Biological Sciences, San José State University, San José, CA 95192, USA
| | - Jessica L Feldman
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
28
|
Lloyd CJ, Guo S, Kinrade B, Zahiri H, Eves R, Ali SK, Yildiz F, Voets IK, Davies PL, Klose KE. A peptide-binding domain shared with an Antarctic bacterium facilitates Vibrio cholerae human cell binding and intestinal colonization. Proc Natl Acad Sci U S A 2023; 120:e2308238120. [PMID: 37729203 PMCID: PMC10523503 DOI: 10.1073/pnas.2308238120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/31/2023] [Indexed: 09/22/2023] Open
Abstract
Vibrio cholerae, the causative agent of the disease cholera, is responsible for multiple pandemics. V. cholerae binds to and colonizes the gastrointestinal tract within the human host, as well as various surfaces in the marine environment (e.g., zooplankton) during interepidemic periods. A large adhesin, the Flagellar Regulated Hemagglutinin A (FrhA), enhances binding to erythrocytes and epithelial cells and enhances intestinal colonization. We identified a peptide-binding domain (PBD) within FrhA that mediates hemagglutination, binding to epithelial cells, intestinal colonization, and facilitates biofilm formation. Intriguingly, this domain is also found in the ice-binding protein of the Antarctic bacterium Marinomonas primoryensis, where it mediates binding to diatoms. Peptide inhibitors of the M. primoryensis PBD inhibit V. cholerae binding to human cells as well as to diatoms and inhibit biofilm formation. Moreover, the M. primoryensis PBD inserted into FrhA allows V. cholerae to bind human cells and colonize the intestine and also enhances biofilm formation, demonstrating the interchangeability of the PBD from these bacteria. Importantly, peptide inhibitors of PBD reduce V. cholerae intestinal colonization in infant mice. These studies demonstrate how V. cholerae uses a PBD shared with a diatom-binding Antarctic bacterium to facilitate intestinal colonization in humans and biofilm formation in the environment.
Collapse
Affiliation(s)
- Cameron J. Lloyd
- South Texas Center for Emerging Infectious Diseases, University of Texas, San Antonio, TX78249
- Department of Molecular Microbiology and Immunology, University of Texas, San Antonio, TX78249
| | - Shuaiqi Guo
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ONK7L 3N6, Canada
| | - Brett Kinrade
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ONK7L 3N6, Canada
| | - Hossein Zahiri
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ONK7L 3N6, Canada
| | - Robert Eves
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ONK7L 3N6, Canada
| | - Syed Khalid Ali
- South Texas Center for Emerging Infectious Diseases, University of Texas, San Antonio, TX78249
- Department of Molecular Microbiology and Immunology, University of Texas, San Antonio, TX78249
| | - Fitnat Yildiz
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, CA95064
| | - Ilja K. Voets
- Laboratory of Macromolecular and Organic Chemistry, Eindhoven University of Technology, Eindhoven5612, the Netherlands
| | - Peter L. Davies
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ONK7L 3N6, Canada
| | - Karl E. Klose
- South Texas Center for Emerging Infectious Diseases, University of Texas, San Antonio, TX78249
- Department of Molecular Microbiology and Immunology, University of Texas, San Antonio, TX78249
| |
Collapse
|
29
|
Nagendra K, Izzet A, Judd NB, Zakine R, Friedman L, Harrison OJ, Pontani LL, Shapiro L, Honig B, Brujic J. Push-pull mechanics of E-cadherin ectodomains in biomimetic adhesions. Biophys J 2023; 122:3506-3515. [PMID: 37528581 PMCID: PMC10502478 DOI: 10.1016/j.bpj.2023.07.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/12/2023] [Accepted: 07/27/2023] [Indexed: 08/03/2023] Open
Abstract
E-cadherin plays a central role in cell-cell adhesion. The ectodomains of wild-type cadherins form a crystalline-like two-dimensional lattice in cell-cell interfaces mediated by both trans (apposed cell) and cis (same cell) interactions. In addition to these extracellular forces, adhesive strength is further regulated by cytosolic phenomena involving α and β catenin-mediated interactions between cadherin and the actin cytoskeleton. Cell-cell adhesion can be further strengthened under tension through mechanisms that have not been definitively characterized in molecular detail. Here we quantitatively determine the role of the cadherin ectodomain in mechanosensing. To this end, we devise an E-cadherin-coated emulsion system, in which droplet surface tension is balanced by protein binding strength to give rise to stable areas of adhesion. To reach the honeycomb/cohesive limit, an initial emulsion compression by centrifugation facilitates E-cadherin trans binding, whereas a high protein surface concentration enables the cis-enhanced stabilization of the interface. We observe an abrupt concentration dependence on recruitment into adhesions of constant crystalline density, reminiscent of a first-order phase transition. Removing the lateral cis interaction with a "cis mutant" shifts this transition to higher surface densities leading to denser, yet weaker adhesions. In both proteins, the stabilization of progressively larger areas of deformation is consistent with single-molecule experiments that show a force-dependent lifetime enhancement in the cadherin ectodomain, which may be attributed to the "X-dimer" bond.
Collapse
Affiliation(s)
- Kartikeya Nagendra
- Center for Soft Matter Research, Department of Physics, New York University, New York, New York; Molecular Biophysics and Biochemistry Training Program, NYU Grossman School of Medicine, New York, New York
| | - Adrien Izzet
- Center for Soft Matter Research, Department of Physics, New York University, New York, New York
| | - Nicolas B Judd
- Center for Soft Matter Research, Department of Physics, New York University, New York, New York
| | - Ruben Zakine
- Center for Soft Matter Research, Department of Physics, New York University, New York, New York
| | - Leah Friedman
- Center for Soft Matter Research, Department of Physics, New York University, New York, New York; Département de Physique, École Normale Supérieure, PSL University, Paris, France
| | - Oliver J Harrison
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York
| | - Léa-Laetitia Pontani
- Laboratoire Jean Perrin, Institut de Biologie Paris-Seine, Sorbonne Université, CNRS, Paris, France
| | - Lawrence Shapiro
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York
| | - Barry Honig
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York; Department of Medicine, Division of Nephrology, Columbia University, New York, New York; Department of Systems Biology, Columbia University, New York, New York
| | - Jasna Brujic
- Center for Soft Matter Research, Department of Physics, New York University, New York, New York; Laboratoire de Physique et Mécanique de Milieux Hétérogènes, UMR 7636, CNRS, ESPCI Paris-PSL, Sorbonne Université, Université Paris Cité, Paris, France.
| |
Collapse
|
30
|
Wu Y, Sun SX. Mechanics of cell-cell junctions. Biophys J 2023; 122:3354-3368. [PMID: 37475215 PMCID: PMC10465726 DOI: 10.1016/j.bpj.2023.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/01/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023] Open
Abstract
Tissue cells in epithelial or endothelial monolayers are connected through cell-cell junctions, which are stabilized by transmembrane E-cadherin bonds and intracellular actin filaments. These bonds and junctions play a crucial role in maintaining the barrier function of epithelia and endothelia and are believed to transmit forces between cells. Additionally, E-cadherin bonds can impact the shape of cell-cell junctions. In this study, we develop a continuum mechanical model of the cell-cell junction by explicitly incorporating the cell membrane, distributions of E-cadherin bonds, cytoplasmic fluid pressure, and F-actin dynamics. The static force-balanced version of the model is able to analyze the influences of cell cortical tension, actin dynamics, and cytoplasmic pressure on the junction shape and E-cadherin bonds. Furthermore, an extended model that incorporates fluid flow, across the cell boundary as well as around the cell, is also examined. This model can couple cell-shape changes with cell cortical tension and fluid flow, and predicts the additional effect of fluid motion on cell-cell junction mechanics. Taken together, our models serve as an intermediate link between molecular-scale models of cell-junction molecules and cell-scale models of tissue and epithelia.
Collapse
Affiliation(s)
- Yufei Wu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland; Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland
| | - Sean X Sun
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland; Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland; Center for Cell Dynamics, Johns Hopkins School of Medicine, Baltimore, Maryland.
| |
Collapse
|
31
|
Shome S, Jia K, Sivasankar S, Jernigan RL. Characterizing interactions in E-cadherin assemblages. Biophys J 2023; 122:3069-3077. [PMID: 37345249 PMCID: PMC10432173 DOI: 10.1016/j.bpj.2023.06.009] [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: 04/21/2022] [Revised: 09/26/2022] [Accepted: 06/14/2023] [Indexed: 06/23/2023] Open
Abstract
Cadherin intermolecular interactions are critical for cell-cell adhesion and play essential roles in tissue formation and the maintenance of tissue structures. In this study, we focus on E-cadherin, a classical cadherin that connects epithelial cells, to understand how they interact in cis and trans conformations when attached to the same cell or opposing cells. We employ coevolutionary sequence analysis and molecular dynamics simulations to confirm previously known interaction sites as well as to identify new interaction sites. The sequence coevolutionary results yield a surprising result indicating that there are no strongly favored intermolecular interaction sites, which is unusual and suggests that many interaction sites may be possible, with none being strongly preferred over others. By using molecular dynamics, we test the persistence of these interactions and how they facilitate adhesion. We build several types of cadherin assemblages, with different numbers and combinations of cis and trans interfaces to understand how these conformations act to facilitate adhesion. Our results suggest that, in addition to the established interaction sites on the EC1 and EC2 domains, an additional plausible cis interface at the EC3-EC5 domain exists. Furthermore, we identify specific mutations at cis/trans binding sites that impair adhesion within E-cadherin assemblages.
Collapse
Affiliation(s)
- Sayane Shome
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, Iowa
| | - Kejue Jia
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, Iowa
| | - Sanjeevi Sivasankar
- Department of Biomedical Engineering, University of California, Davis, Davis, California
| | - Robert L Jernigan
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, Iowa.
| |
Collapse
|
32
|
Xie B, Xu S, Schecterson L, Gumbiner BM, Sivasankar S. Strengthening E-cadherin adhesion via antibody mediated binding stabilization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.04.547716. [PMID: 37461464 PMCID: PMC10350017 DOI: 10.1101/2023.07.04.547716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
E-cadherins (Ecads) are a crucial cell-cell adhesion protein with tumor suppression properties. Ecad adhesion can be enhanced by the monoclonal antibody 66E8, which has potential applications in inhibiting cancer metastasis. However, the biophysical mechanisms underlying 66E8 mediated adhesion strengthening are unknown. Here, we use molecular dynamics simulations, site directed mutagenesis and single molecule atomic force microscopy experiments to demonstrate that 66E8 strengthens Ecad binding by stabilizing the primary Ecad adhesive conformation: the strand-swap dimer. By forming electrostatic interactions with Ecad, 66E8 stabilizes the swapped β-strand and its hydrophobic pocket and impedes Ecad conformational changes, which are necessary for rupture of the strand-swap dimer. Our findings identify fundamental mechanistic principles for strengthening of Ecad binding using monoclonal antibodies.
Collapse
|
33
|
Nielsen MS, van Opbergen CJM, van Veen TAB, Delmar M. The intercalated disc: a unique organelle for electromechanical synchrony in cardiomyocytes. Physiol Rev 2023; 103:2271-2319. [PMID: 36731030 PMCID: PMC10191137 DOI: 10.1152/physrev.00021.2022] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 01/24/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
The intercalated disc (ID) is a highly specialized structure that connects cardiomyocytes via mechanical and electrical junctions. Although described in some detail by light microscopy in the 19th century, it was in 1966 that electron microscopy images showed that the ID represented apposing cell borders and provided detailed insight into the complex ID nanostructure. Since then, much has been learned about the ID and its molecular composition, and it has become evident that a large number of proteins, not all of them involved in direct cell-to-cell coupling via mechanical or gap junctions, reside at the ID. Furthermore, an increasing number of functional interactions between ID components are emerging, leading to the concept that the ID is not the sum of isolated molecular silos but an interacting molecular complex, an "organelle" where components work in concert to bring about electrical and mechanical synchrony. The aim of the present review is to give a short historical account of the ID's discovery and an updated overview of its composition and organization, followed by a discussion of the physiological implications of the ID architecture and the local intermolecular interactions. The latter will focus on both the importance of normal conduction of cardiac action potentials as well as the impact on the pathophysiology of arrhythmias.
Collapse
Affiliation(s)
- Morten S Nielsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Chantal J M van Opbergen
- The Leon Charney Division of Cardiology, New York University Grossmann School of Medicine, New York, New York, United States
| | - Toon A B van Veen
- Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mario Delmar
- The Leon Charney Division of Cardiology, New York University Grossmann School of Medicine, New York, New York, United States
| |
Collapse
|
34
|
Cui L, Shi X, Li H, Wang S, Guo L, Lan Z, Dai Y, Zhang Q, Wu Y, Liu W. Crystal structures and solution conformations of HtrA from Helicobacter pylori reveal pH-dependent oligomeric conversion and conformational rearrangements. Int J Biol Macromol 2023:125274. [PMID: 37301353 DOI: 10.1016/j.ijbiomac.2023.125274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/06/2023] [Accepted: 06/07/2023] [Indexed: 06/12/2023]
Abstract
Helicobacter pylori is a Gram-negative microaerophilic bacterium that infects over 50 % of the world's population, making it a major risk factor for chronic gastritis, ulcer diseases of the stomach and duodenum, MALT lymphoma, and gastric cancer. The clinical consequences of H. pylori infection are closely linked with the expression of virulence factors secreted by the bacterium. One such virulence factor is high temperature requirement A (HtrA), which possesses chaperone and serine protease activity. In the host stomach, HtrA secreted from H. pylori (HpHtrA) disrupts intercellular adhesions by cleaving epithelial adhesion proteins including E-cadherin and desmoglein-2. This disruption causes intercellular junctions to open, allowing the bacterium to pass through the epithelial barrier, access the intercellular space, and colonize the gastric mucosa. HtrA proteases are well known for their structural complexity, reflected in their diverse oligomer forms and multi-tasking activities in both prokaryotes and eukaryotes. In this study, we determined crystal structures and solution conformations of HpHtrA monomer and trimer, which revealed large domain rearrangements between them. Notably, this is the first report of a monomeric structure in the HtrA family. We further found a pH-dependent dynamic trimer-to-monomer conversion and concurrent conformational changes that seem closely linked with a pH-sensing ability through the protonation of certain Asp residues. These results advance our understanding of the functional roles and the related mechanisms of this protease in bacterial infection, which may shed light on the development of HtrA-targeted therapies for H. pylori-associated diseases.
Collapse
Affiliation(s)
- Liwei Cui
- Institute of Immunology, PLA, Army Medical University, Chongqing 400038, China; Department of Tropical Medicine and Infectious Diseases, Hainan Hospital of Chinese PLA General Hospital, Sanya, Hainan 572000, China
| | - Xiangrui Shi
- Department of Obstetrics and Gynecology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Huiling Li
- Department of Tropical Medicine and Infectious Diseases, Hainan Hospital of Chinese PLA General Hospital, Sanya, Hainan 572000, China
| | - Sheng Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Avenue, Wuhan, Hubei 430074, China
| | - Ling Guo
- Institute of Immunology, PLA, Army Medical University, Chongqing 400038, China; Center for drug evaluation and inspection of Chongqing Medical Products Administration, Chongqing 401120, China
| | - Zhu Lan
- Institute of Immunology, PLA, Army Medical University, Chongqing 400038, China
| | - Yujie Dai
- Department of Obstetrics and Gynecology, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Qinghua Zhang
- Department of Obstetrics and Gynecology, Daping Hospital, Army Medical University, Chongqing 400042, China.
| | - Yuzhang Wu
- Institute of Immunology, PLA, Army Medical University, Chongqing 400038, China.
| | - Wei Liu
- Institute of Immunology, PLA, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
35
|
Su Z, Almo SC, Wu Y. Understanding the General Principles of T Cell Engagement by Multiscale Computational Simulations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.07.544116. [PMID: 37333150 PMCID: PMC10274768 DOI: 10.1101/2023.06.07.544116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The use of bispecific antibodies as T cell engagers can bypass the normal TCR-MHC interaction, redirect the cytotoxic activity of T-cells, and lead to highly efficient tumor cell killing. However, this immunotherapy also causes significant on-target off-tumor toxicologic effects, especially when they were used to treat solid tumors. In order to avoid these adverse events, it is necessary to understand the fundamental mechanisms during the physical process of T cell engagement. We developed a multiscale computational framework to reach this goal. The framework combines simulations on the intercellular and multicellular levels. On the intercellular level, we simulated the spatial-temporal dynamics of three-body interactions among bispecific antibodies, CD3 and TAA. The derived number of intercellular bonds formed between CD3 and TAA were further transferred into the multicellular simulations as the input parameter of adhesive density between cells. Through the simulations under various molecular and cellular conditions, we were able to gain new insights of how to adopt the most appropriate strategy to maximize the drug efficacy and avoid the off-target effect. For instance, we discovered that the low antibody binding affinity resulted in the formation of large clusters at the cell-cell interface, which could be important to control the downstream signaling pathways. We also tested different molecular architectures of the bispecific antibody and suggested the existence of an optimal length in regulating the T cell engagement. Overall, the current multiscale simulations serve as a prove-of-concept study to help the future design of new biological therapeutics. SIGNIFICANCE T-cell engagers are a class of anti-cancer drugs that can directly kill tumor cells by bringing T cells next to them. However, current treatments using T-cell engagers can cause serious side-effects. In order to reduce these effects, it is necessary to understand how T cells and tumor cells interact together through the connection of T-cell engagers. Unfortunately, this process is not well studied due to the limitations in current experimental techniques. We developed computational models on two different scales to simulate the physical process of T cell engagement. Our simulation results provide new insights into the general properties of T cell engagers. The new simulation methods can therefore serve as a useful tool to design novel antibodies for cancer immunotherapy.
Collapse
|
36
|
Larsen ISB, Povolo L, Zhou L, Tian W, Mygind KJ, Hintze J, Jiang C, Hartill V, Prescott K, Johnson CA, Mullegama SV, McConkie-Rosell A, McDonald M, Hansen L, Vakhrushev SY, Schjoldager KT, Clausen H, Worzfeld T, Joshi HJ, Halim A. The SHDRA syndrome-associated gene TMEM260 encodes a protein-specific O-mannosyltransferase. Proc Natl Acad Sci U S A 2023; 120:e2302584120. [PMID: 37186866 PMCID: PMC10214176 DOI: 10.1073/pnas.2302584120] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/21/2023] [Indexed: 05/17/2023] Open
Abstract
Mutations in the TMEM260 gene cause structural heart defects and renal anomalies syndrome, but the function of the encoded protein remains unknown. We previously reported wide occurrence of O-mannose glycans on extracellular immunoglobulin, plexin, transcription factor (IPT) domains found in the hepatocyte growth factor receptor (cMET), macrophage-stimulating protein receptor (RON), and plexin receptors, and further demonstrated that two known protein O-mannosylation systems orchestrated by the POMT1/2 and transmembrane and tetratricopeptide repeat-containing proteins 1-4 gene families were not required for glycosylation of these IPT domains. Here, we report that the TMEM260 gene encodes an ER-located protein O-mannosyltransferase that selectively glycosylates IPT domains. We demonstrate that disease-causing TMEM260 mutations impair O-mannosylation of IPT domains and that TMEM260 knockout in cells results in receptor maturation defects and abnormal growth of 3D cell models. Thus, our study identifies the third protein-specific O-mannosylation pathway in mammals and demonstrates that O-mannosylation of IPT domains serves critical functions during epithelial morphogenesis. Our findings add a new glycosylation pathway and gene to a growing group of congenital disorders of glycosylation.
Collapse
Affiliation(s)
- Ida Signe Bohse Larsen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200Copenhagen N, Denmark
| | - Lorenzo Povolo
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200Copenhagen N, Denmark
| | - Luping Zhou
- Faculty of Medicine, Institute of Pharmacology, University of Marburg, 35043Marburg, Germany
| | - Weihua Tian
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200Copenhagen N, Denmark
| | - Kasper Johansen Mygind
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200Copenhagen N, Denmark
| | - John Hintze
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200Copenhagen N, Denmark
| | - Chen Jiang
- Faculty of Medicine, Institute of Pharmacology, University of Marburg, 35043Marburg, Germany
| | - Verity Hartill
- Leeds Institute of Medical Research, University of Leeds, St James’ University Hospital, LeedsLS2 9JT, United Kingdom
- Yorkshire Regional Genetics Service, Chapel Allerton Hospital, LeedsLS7 4SA, United Kingdom
| | - Katrina Prescott
- Yorkshire Regional Genetics Service, Chapel Allerton Hospital, LeedsLS7 4SA, United Kingdom
| | - Colin A. Johnson
- Leeds Institute of Medical Research, University of Leeds, St James’ University Hospital, LeedsLS2 9JT, United Kingdom
| | | | - Allyn McConkie-Rosell
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC27710
| | - Marie McDonald
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC27710
| | - Lars Hansen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200Copenhagen N, Denmark
| | - Sergey Y. Vakhrushev
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200Copenhagen N, Denmark
| | - Katrine T. Schjoldager
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200Copenhagen N, Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200Copenhagen N, Denmark
| | - Thomas Worzfeld
- Faculty of Medicine, Institute of Pharmacology, University of Marburg, 35043Marburg, Germany
- Max-Planck-Institute for Heart and Lung Research, 61231Bad Nauheim, Germany
| | - Hiren J. Joshi
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200Copenhagen N, Denmark
| | - Adnan Halim
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200Copenhagen N, Denmark
| |
Collapse
|
37
|
Lin WH, Cooper LM, Anastasiadis PZ. Cadherins and catenins in cancer: connecting cancer pathways and tumor microenvironment. Front Cell Dev Biol 2023; 11:1137013. [PMID: 37255594 PMCID: PMC10225604 DOI: 10.3389/fcell.2023.1137013] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/03/2023] [Indexed: 06/01/2023] Open
Abstract
Cadherin-catenin complexes are integral components of the adherens junctions crucial for cell-cell adhesion and tissue homeostasis. Dysregulation of these complexes is linked to cancer development via alteration of cell-autonomous oncogenic signaling pathways and extrinsic tumor microenvironment. Advances in multiomics have uncovered key signaling events in multiple cancer types, creating a need for a better understanding of the crosstalk between cadherin-catenin complexes and oncogenic pathways. In this review, we focus on the biological functions of classical cadherins and associated catenins, describe how their dysregulation influences major cancer pathways, and discuss feedback regulation mechanisms between cadherin complexes and cellular signaling. We discuss evidence of cross regulation in the following contexts: Hippo-Yap/Taz and receptor tyrosine kinase signaling, key pathways involved in cell proliferation and growth; Wnt, Notch, and hedgehog signaling, key developmental pathways involved in human cancer; as well as TGFβ and the epithelial-to-mesenchymal transition program, an important process for cancer cell plasticity. Moreover, we briefly explore the role of cadherins and catenins in mechanotransduction and the immune tumor microenvironment.
Collapse
|
38
|
Nishiguchi S, Kasai RS, Uchihashi T. Antiparallel dimer structure of CELSR cadherin in solution revealed by high-speed atomic force microscopy. Proc Natl Acad Sci U S A 2023; 120:e2302047120. [PMID: 37094146 PMCID: PMC10160967 DOI: 10.1073/pnas.2302047120] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/03/2023] [Indexed: 04/26/2023] Open
Abstract
Cadherin EGF LAG seven-pass G-type receptors (CELSR) cadherins, members of the cadherin superfamily, and adhesion G-protein-coupled receptors, play a vital role in cell-cell adhesion. The mutual binding of the extracellular domains (ectodomains) of CELSR cadherins between cells is crucial for tissue formation, including the establishment of planar cell polarity, which directs the proper patterning of cells. CELSR cadherins possess nine cadherin ectodomains (EC1-EC9) and noncadherin ectodomains. However, the structural and functional mechanisms of the binding mode of CELSR cadherins have not been determined. In this study, we investigated the binding mode of CELSR cadherins using single-molecule fluorescence microscopy, high-speed atomic force microscopy (HS-AFM), and bead aggregation assay. The fluorescence microscopy analysis results indicated that the trans-dimer of the CELSR cadherin constitutes the essential adhesive unit between cells. HS-AFM analysis and bead aggregation assay results demonstrated that EC1-EC8 entirely overlap and twist to form antiparallel dimer conformations and that the binding of EC1-EC4 is sufficient to sustain bead aggregation. The interaction mechanism of CELSR cadherin may elucidate the variation of the binding mechanism within the cadherin superfamily and physiological role of CELSR cadherins in relation to planar cell polarity.
Collapse
Affiliation(s)
- Shigetaka Nishiguchi
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki444-8787, Japan
| | - Rinshi S. Kasai
- Institute for Life and Medical Sciences, Kyoto University, Kyoto606-8507, Japan
- Institute for Glyco-core Research, Gifu University, Gifu501-1193, Japan
| | - Takayuki Uchihashi
- Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki444-8787, Japan
- Department of Physics, Nagoya University, Nagoya464-8602, Japan
- Institute for Glyco-core Research, Nagoya University, Nagoya464-8602, Japan
| |
Collapse
|
39
|
Troyanovsky SM. Adherens junction: the ensemble of specialized cadherin clusters. Trends Cell Biol 2023; 33:374-387. [PMID: 36127186 PMCID: PMC10020127 DOI: 10.1016/j.tcb.2022.08.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022]
Abstract
The cell-cell connections in adherens junctions (AJs) are mediated by transmembrane receptors, type I cadherins (referred to here as cadherins). These cadherin-based connections (or trans bonds) are weak. To upregulate their strength, cadherins exploit avidity, the increased affinity of binding between cadherin clusters compared with isolated monomers. Formation of such clusters is a unique molecular process that is driven by a synergy of direct and indirect cis interactions between cadherins located at the same cell. In addition to their role in adhesion, cadherin clusters provide structural scaffolds for cytosolic proteins, which implicate cadherin into different cellular activities and signaling pathways. The cluster lifetime, which depends on the actin cytoskeleton, and on the mechanical forces it generates, determines the strength of AJs and their plasticity. The key aspects of cadherin adhesion, therefore, cannot be understood at the level of isolated cadherin molecules, but should be discussed in the context of cadherin clusters.
Collapse
Affiliation(s)
- Sergey M Troyanovsky
- Department of Dermatology, Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Cell and Molecular Biology, Northwestern University, The Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
40
|
Srinivas CS, Singaraju GS, Kaur V, Das S, Ghosh SK, Sagar A, Kumar A, Bhatia T, Rakshit S. Transient interactions drive the lateral clustering of cadherin-23 on membrane. Commun Biol 2023; 6:293. [PMID: 36934176 PMCID: PMC10024700 DOI: 10.1038/s42003-023-04677-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 03/07/2023] [Indexed: 03/20/2023] Open
Abstract
Cis and trans-interactions among cadherins secure multicellularity. While the molecular structure of trans-interactions of cadherins is well understood, work to identify the molecular cues that spread the cis-interactions two-dimensionally is still ongoing. Here, we report that transient, weak, yet multivalent, and spatially distributed hydrophobic interactions that are involved in liquid-liquid phase separations of biomolecules in solution, alone can drive the lateral-clustering of cadherin-23 on a membrane. No specific cis-dimer interactions are required for the lateral clustering. In cells, the cis-clustering accelerates cell-cell adhesion and, thus, contributes to cell-adhesion kinetics along with strengthening the junction. Although the physiological connection of cis-clustering with rapid adhesion is yet to be explored, we speculate that the over-expression of cadherin-23 in M2-macrophages may facilitate faster attachments to circulatory tumor cells during metastasis.
Collapse
Affiliation(s)
- Cheerneni S Srinivas
- Department of Chemical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Gayathri S Singaraju
- Department of Chemical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Veerpal Kaur
- Department of Chemical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Sayan Das
- Department of Chemical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Sanat K Ghosh
- Department of Physical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Amin Sagar
- Department of Chemical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Anuj Kumar
- Department of Physical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
- Centre for Protein Science Design and Engineering, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Tripta Bhatia
- Department of Physical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Sabyasachi Rakshit
- Department of Chemical Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India.
- Centre for Protein Science Design and Engineering, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India.
| |
Collapse
|
41
|
Totoki Y, Saito-Adachi M, Shiraishi Y, Komura D, Nakamura H, Suzuki A, Tatsuno K, Rokutan H, Hama N, Yamamoto S, Ono H, Arai Y, Hosoda F, Katoh H, Chiba K, Iida N, Nagae G, Ueda H, Shihang C, Sekine S, Abe H, Nomura S, Matsuura T, Sakai E, Ohshima T, Rino Y, Yeoh KG, So J, Sanghvi K, Soong R, Fukagawa A, Yachida S, Kato M, Seto Y, Ushiku T, Nakajima A, Katai H, Tan P, Ishikawa S, Aburatani H, Shibata T. Multiancestry genomic and transcriptomic analysis of gastric cancer. Nat Genet 2023; 55:581-594. [PMID: 36914835 DOI: 10.1038/s41588-023-01333-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/06/2023] [Indexed: 03/16/2023]
Abstract
Gastric cancer is among the most common malignancies worldwide, characterized by geographical, epidemiological and histological heterogeneity. Here, we report an extensive, multiancestral landscape of driver events in gastric cancer, involving 1,335 cases. Seventy-seven significantly mutated genes (SMGs) were identified, including ARHGAP5 and TRIM49C. We also identified subtype-specific drivers, including PIGR and SOX9, which were enriched in the diffuse subtype of the disease. SMGs also varied according to Epstein-Barr virus infection status and ancestry. Non-protein-truncating CDH1 mutations, which are characterized by in-frame splicing alterations, targeted localized extracellular domains and uniquely occurred in sporadic diffuse-type cases. In patients with gastric cancer with East Asian ancestry, our data suggested a link between alcohol consumption or metabolism and the development of RHOA mutations. Moreover, mutations with potential roles in immune evasion were identified. Overall, these data provide comprehensive insights into the molecular landscape of gastric cancer across various subtypes and ancestries.
Collapse
Affiliation(s)
- Yasushi Totoki
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Mihoko Saito-Adachi
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Yuichi Shiraishi
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Daisuke Komura
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiromi Nakamura
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Akihiro Suzuki
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Kanagawa, Japan.,Genome Science and Medicine Laboratory, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Kenji Tatsuno
- Genome Science and Medicine Laboratory, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Hirofumi Rokutan
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Natsuko Hama
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Shogo Yamamoto
- Genome Science and Medicine Laboratory, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Hanako Ono
- Division of Bioinformatics, National Cancer Center Research Institute, Tokyo, Japan
| | - Yasuhito Arai
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Fumie Hosoda
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Hiroto Katoh
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kenichi Chiba
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Naoko Iida
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Genta Nagae
- Genome Science and Medicine Laboratory, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Hiroki Ueda
- Biological Data Science, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Chen Shihang
- Genome Science and Medicine Laboratory, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Shigeki Sekine
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroyuki Abe
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sachiyo Nomura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tetsuya Matsuura
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Eiji Sakai
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Takashi Ohshima
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Kanagawa, Japan
| | - Yasushi Rino
- Department of Surgery, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Khay Guan Yeoh
- Dept of Medicine, National University of Singapore, Singapore, Singapore
| | - Jimmy So
- Dept of Surgery, National University of Singapore, Singapore, Singapore
| | - Kaushal Sanghvi
- Dept of Surgery, Tan Tock Seng Hospital, Singapore, Singapore
| | - Richie Soong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Akihiko Fukagawa
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Shinichi Yachida
- Department of Cancer Genome Informatics, Graduate School of Medicine, Osaka University, Osaka, Japan.,Division of Genomic Medicine, National Cancer Center Research Institute, Tokyo, Japan
| | - Mamoru Kato
- Division of Bioinformatics, National Cancer Center Research Institute, Tokyo, Japan
| | - Yasuyuki Seto
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Hitoshi Katai
- Department of Gastric Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Patrick Tan
- Cancer and Stem Cell Biology, Duke-NUS Medical School Singapore, Singapore, Singapore.,Epigenomic and Epitranscriptomic Regulation, Genome Institute of Singapore, Singapore, Singapore
| | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Aburatani
- Genome Science and Medicine Laboratory, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Tatsuhiro Shibata
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan. .,Laboratory of Molecular Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
42
|
Kasiah J, McNeill H. Fat and Dachsous cadherins in mammalian development. Curr Top Dev Biol 2023; 154:223-244. [PMID: 37100519 DOI: 10.1016/bs.ctdb.2023.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Cell growth and patterning are critical for tissue development. Here we discuss the evolutionarily conserved cadherins, Fat and Dachsous, and the roles they play during mammalian tissue development and disease. In Drosophila, Fat and Dachsous regulate tissue growth via the Hippo pathway and planar cell polarity (PCP). The Drosophila wing has been an ideal tissue to observe how mutations in these cadherins affect tissue development. In mammals, there are multiple Fat and Dachsous cadherins, which are expressed in many tissues, but mutations in these cadherins that affect growth and tissue organization are context dependent. Here we examine how mutations in the Fat and Dachsous mammalian genes affect development in mammals and contribute to human disease.
Collapse
Affiliation(s)
- Jennysue Kasiah
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO, United States
| | - Helen McNeill
- Department of Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO, United States.
| |
Collapse
|
43
|
Kwak M, Southard KM, Kim WR, Lin A, Kim NH, Gopalappa R, Lee HJ, An M, Choi SH, Jung Y, Noh K, Farlow J, Georgakopoulos A, Robakis NK, Kang MK, Kutys ML, Seo D, Kim HH, Kim YH, Cheon J, Gartner ZJ, Jun YW. Adherens junctions organize size-selective proteolytic hotspots critical for Notch signalling. Nat Cell Biol 2022; 24:1739-1753. [PMID: 36456828 PMCID: PMC10665132 DOI: 10.1038/s41556-022-01031-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 10/19/2022] [Indexed: 12/02/2022]
Abstract
Adherens junctions (AJs) create spatially, chemically and mechanically discrete microdomains at cellular interfaces. Here, using a mechanogenetic platform that generates artificial AJs with controlled protein localization, clustering and mechanical loading, we find that AJs also organize proteolytic hotspots for γ-secretase with a spatially regulated substrate selectivity that is critical in the processing of Notch and other transmembrane proteins. Membrane microdomains outside of AJs exclusively organize Notch ligand-receptor engagement (LRE microdomains) to initiate receptor activation. Conversely, membrane microdomains within AJs exclusively serve to coordinate regulated intramembrane proteolysis (RIP microdomains). They do so by concentrating γ-secretase and primed receptors while excluding full-length Notch. AJs induce these functionally distinct microdomains by means of lipid-dependent γ-secretase recruitment and size-dependent protein segregation. By excluding full-length Notch from RIP microdomains, AJs prevent inappropriate enzyme-substrate interactions and suppress spurious Notch activation. Ligand-induced ectodomain shedding eliminates size-dependent segregation, releasing Notch to translocate into AJs for processing by γ-secretase. This mechanism directs radial differentiation of ventricular zone-neural progenitor cells in vivo and more broadly regulates the proteolysis of other large cell-surface receptors such as amyloid precursor protein. These findings suggest an unprecedented role of AJs in creating size-selective spatial switches that choreograph γ-secretase processing of multiple transmembrane proteins regulating development, homeostasis and disease.
Collapse
Affiliation(s)
- Minsuk Kwak
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, Republic of Korea
| | - Kaden M Southard
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Woon Ryoung Kim
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
| | - Annie Lin
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
| | - Nam Hyeong Kim
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, Republic of Korea
- Department of Nano Engineering, Sungkyunkwan University, Suwon, Republic of Korea
- Imnewrun Inc., Suwon, Republic of Korea
| | - Ramu Gopalappa
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun Jung Lee
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
| | - Minji An
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Seo Hyun Choi
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Yunmin Jung
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Kunwoo Noh
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Justin Farlow
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Anastasios Georgakopoulos
- Department of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nikolaos K Robakis
- Department of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Min K Kang
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Matthew L Kutys
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA
| | - Daeha Seo
- Department of Physics and Chemistry, DGIST, Daegu, Republic of Korea
| | - Hyongbum Henry Kim
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Plus Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong Ho Kim
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, Republic of Korea
- Department of Nano Engineering, Sungkyunkwan University, Suwon, Republic of Korea
- Imnewrun Inc., Suwon, Republic of Korea
| | - Jinwoo Cheon
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul, Republic of Korea
| | - Zev J Gartner
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| | - Young-Wook Jun
- Department of Otolaryngology, University of California, San Francisco, CA, USA.
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA.
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA.
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea.
- Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
44
|
Huebner RJ, Wallingford JB. Dishevelled controls bulk cadherin dynamics and the stability of individual cadherin clusters during convergent extension. Mol Biol Cell 2022; 33:br26. [PMID: 36222834 PMCID: PMC9727802 DOI: 10.1091/mbc.e22-06-0194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Animals are shaped through the movement of large cellular collectives. Such morphogenetic processes require cadherin-based cell adhesion to maintain tissue cohesion and planar cell polarity to coordinate movement. Despite a vast literature surrounding cadherin-based adhesion and planar cell polarity, it is unclear how these molecular networks interface. Here we investigate the relationship between cadherins and planar cell polarity during gastrulation cell movements in Xenopus laevis. We first assessed bulk cadherin localization and found that cadherins were enriched at a specific subset of morphogenetically active cell-cell junctions. We then found that cadherin and actin had coupled temporal dynamics and that disruption of planar cell polarity uncoupled these dynamics. Next, using superresolution time-lapse microscopy and quantitative image analysis, we were able to measure the lifespan and size of individual cadherin clusters. Finally, we show that planar cell polarity not only controls the size of cadherin clusters but, more interestingly, regulates cluster stability. These results reveal an intriguing link between two essential cellular properties, adhesion and planar polarity, and provide insight into the molecular control of morphogenetic cell movements.
Collapse
Affiliation(s)
- Robert J. Huebner
- Department of Molecular Biosciences, University of Texas, Austin, TX 78712
| | - John B. Wallingford
- Department of Molecular Biosciences, University of Texas, Austin, TX 78712,*Address correspondence to: John B. Wallingford ()
| |
Collapse
|
45
|
Ibata N, Terentjev EM. Nucleation of cadherin clusters on cell-cell interfaces. Sci Rep 2022; 12:18485. [PMID: 36323859 PMCID: PMC9630535 DOI: 10.1038/s41598-022-23220-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 10/27/2022] [Indexed: 12/05/2022] Open
Abstract
Cadherins mediate cell-cell adhesion and help the cell determine its shape and function. Here we study collective cadherin organization and interactions within cell-cell contact areas, and find the cadherin density at which a 'gas-liquid' phase transition occurs, when cadherin monomers begin to aggregate into dense clusters. We use a 2D lattice model of a cell-cell contact area, and coarse-grain to the continuous number density of cadherin to map the model onto the Cahn-Hilliard coarsening theory. This predicts the density required for nucleation, the characteristic length scale of the process, and the number density of clusters. The analytical predictions of the model are in good agreement with experimental observations of cadherin clustering in epithelial tissues.
Collapse
Affiliation(s)
- Neil Ibata
- grid.5335.00000000121885934Cavendish Laboratory, University of Cambridge, JJ Thomson Avenue, Cambridge, CB3 0HE UK
| | - Eugene M. Terentjev
- grid.5335.00000000121885934Cavendish Laboratory, University of Cambridge, JJ Thomson Avenue, Cambridge, CB3 0HE UK
| |
Collapse
|
46
|
Kanadome T, Hayashi K, Seto Y, Eiraku M, Nakajima K, Nagai T, Matsuda T. Development of intensiometric indicators for visualizing N-cadherin interaction across cells. Commun Biol 2022; 5:1065. [PMID: 36207396 PMCID: PMC9546846 DOI: 10.1038/s42003-022-04023-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 09/22/2022] [Indexed: 11/09/2022] Open
Abstract
N-cadherin (NCad) is a classical cadherin that mediates cell–cell interactions in a Ca2+-dependent manner. NCad participates in various biological processes, from ontogenesis to higher brain functions, though the visualization of NCad interactions in living cells remains limited. Here, we present intensiometric NCad interaction indicators, named INCIDERs, that utilize dimerization-dependent fluorescent proteins. INCIDERs successfully visualize reversible NCad interactions across cells. Compared to FRET-based indicators, INCIDERs have a ~70-fold higher signal contrast, enabling clear identification of NCad interactions. In primary neuronal cells, NCad interactions are visualized between closely apposed processes. Furthermore, visualization of NCad interaction at cell adhesion sites in dense cell populations is achieved by two-photon microscopy. INCIDERs are useful tools in the spatiotemporal investigation of NCad interactions across cells; future research should evaluate the potential of INCIDERs in mapping complex three-dimensional architectures in multi-cellular systems. Intensiometric N-cadherin (NCad) interaction indicators, named INCIDERs, visualize reversible NCad-mediated cell-cell interactions.
Collapse
Affiliation(s)
- Takashi Kanadome
- Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), Kawaguchi, Saitama, 332-0012, Japan.,Department of Biomolecular Science and Engineering, SANKEN (The Institute of Scientific and Industrial Research), Osaka University, 8-1 Mihogaoka, Ibaraki, 567-0047, Japan
| | - Kanehiro Hayashi
- Department of Anatomy, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yusuke Seto
- Laboratory of Developmental Systems, Institute for Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Mototsugu Eiraku
- Laboratory of Developmental Systems, Institute for Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan.,Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, 606-8507, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Takeharu Nagai
- Department of Biomolecular Science and Engineering, SANKEN (The Institute of Scientific and Industrial Research), Osaka University, 8-1 Mihogaoka, Ibaraki, 567-0047, Japan
| | - Tomoki Matsuda
- Department of Biomolecular Science and Engineering, SANKEN (The Institute of Scientific and Industrial Research), Osaka University, 8-1 Mihogaoka, Ibaraki, 567-0047, Japan.
| |
Collapse
|
47
|
Brittle A, Warrington SJ, Strutt H, Manning E, Tan SE, Strutt D. Distinct mechanisms of planar polarization by the core and Fat-Dachsous planar polarity pathways in the Drosophila wing. Cell Rep 2022; 40:111419. [PMID: 36170824 PMCID: PMC9631118 DOI: 10.1016/j.celrep.2022.111419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
Planar polarity describes the coordinated polarization of cells within a tissue plane, and in animals can be determined by the “core” or Fat-Dachsous pathways. Current models for planar polarity establishment involve two components: tissue-level “global” cues that determine the overall axis of polarity and cell-level feedback-mediated cellular polarity amplification. Here, we investigate the contributions of global cues versus cellular feedback amplification in the core and Fat-Dachsous pathways during Drosophila pupal wing development. We present evidence that these pathways generate planar polarity via distinct mechanisms. Core pathway function is consistent with strong feedback capable of self-organizing cell polarity, which can then be aligned with the tissue axis via weak or transient global cues. Conversely, generation of cell polarity by the Ft-Ds pathway depends on strong global cues in the form of graded patterns of gene expression, which can then be amplified by weak feedback mechanisms. The core and Fat-Dachsous planar polarity pathways function via distinct mechanisms The core can self-organize planar polarity and be oriented by weak upstream cues Fat-Dachsous are oriented by strong gradient cues but show poor self-organization
Collapse
Affiliation(s)
- Amy Brittle
- School of Biosciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | | | - Helen Strutt
- School of Biosciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Elizabeth Manning
- School of Biosciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Su Ee Tan
- School of Biosciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - David Strutt
- School of Biosciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK.
| |
Collapse
|
48
|
Boni N, Shapiro L, Honig B, Wu Y, Rubinstein R. On the formation of ordered protein assemblies in cell-cell interfaces. Proc Natl Acad Sci U S A 2022; 119:e2206175119. [PMID: 35969779 PMCID: PMC9407605 DOI: 10.1073/pnas.2206175119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/20/2022] [Indexed: 11/18/2022] Open
Abstract
Crystal structures of many cell-cell adhesion receptors reveal the formation of linear "molecular zippers" comprising an ordered one-dimensional array of proteins that form both intercellular (trans) and intracellular (cis) interactions. The clustered protocadherins (cPcdhs) provide an exemplar of this phenomenon and use it as a basis of barcoding of vertebrate neurons. Here, we report both Metropolis and kinetic Monte Carlo simulations of cPcdh zipper formation using simplified models of cPcdhs that nevertheless capture essential features of their three-dimensional structure. The simulations reveal that the formation of long zippers is an implicit feature of cPcdh structure and is driven by their cis and trans interactions that have been quantitatively characterized in previous work. Moreover, in agreement with cryo-electron tomography studies, the zippers are found to organize into two-dimensional arrays even in the absence of attractive interactions between individual zippers. Our results suggest that the formation of ordered two-dimensional arrays of linear zippers of adhesion proteins is a common feature of cell-cell interfaces. From the perspective of simulations, they demonstrate the importance of a realistic depiction of adhesion protein structure and interactions if important biological phenomena are to be properly captured.
Collapse
Affiliation(s)
- Nadir Boni
- School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Lawrence Shapiro
- Zuckerman Mind, Brain and Behavior Institute, Columbia University, New York, NY 10027
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032
| | - Barry Honig
- Zuckerman Mind, Brain and Behavior Institute, Columbia University, New York, NY 10027
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032
- Department of Systems Biology, Columbia University, New York, NY 10032
- Department of Medicine, Division of Nephrology, Columbia University, New York, NY 10032
| | - Yinghao Wu
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Rotem Rubinstein
- School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv-Yafo, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv-Yafo, Israel
| |
Collapse
|
49
|
Vasile F, Lavore F, Gazzola S, Vettraino C, Parisini E, Piarulli U, Belvisi L, Civera M. A combined fragment-based virtual screening and STD-NMR approach for the identification of E-cadherin ligands. Front Chem 2022; 10:946087. [PMID: 36059878 PMCID: PMC9437437 DOI: 10.3389/fchem.2022.946087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/22/2022] [Indexed: 12/02/2022] Open
Abstract
Cadherins promote cell-cell adhesion by forming homophilic interactions via their N-terminal extracellular domains. Hence, they have broad-ranging physiological effects on tissue organization and homeostasis. When dysregulated, cadherins contribute to different aspects of cancer progression and metastasis; therefore, targeting the cadherin adhesive interface with small-molecule antagonists is expected to have potential therapeutic and diagnostic value. Here, we used molecular docking simulations to evaluate the propensity of three different libraries of commercially available drug-like fragments (nearly 18,000 compounds) to accommodate into the Trp2 binding pocket of E-cadherin, a crucial site for the orchestration of the protein’s dimerization mechanism. Top-ranked fragments featuring five different aromatic chemotypes were expanded by means of a similarity search on the PubChem database (Tanimoto index >90%). Of this set, seven fragments containing an aromatic scaffold linked to an aliphatic chain bearing at least one amine group were finally selected for further analysis. Ligand-based NMR data (Saturation Transfer Difference, STD) and molecular dynamics simulations suggest that these fragments can bind E-cadherin mostly through their aromatic moiety, while their aliphatic portions may also diversely engage with the mobile regions of the binding site. A tetrahydro-β-carboline scaffold functionalized with an ethylamine emerged as the most promising fragment.
Collapse
Affiliation(s)
- Francesca Vasile
- Department of Chemistry, Università Degli Studi di Milano, Milan, Italy
| | - Francesca Lavore
- Department of Chemistry, Università Degli Studi di Milano, Milan, Italy
| | - Silvia Gazzola
- Department of Science and High Technology, Università Degli Studi Dell’Insubria, Como, Italy
| | - Chiara Vettraino
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia @Polimi, Milan, Italy
| | - Emilio Parisini
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia @Polimi, Milan, Italy
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Department of Chemistry “Giacomo Ciamician”, Università Degli Studi di Bologna, Bologna, Italy
| | - Umberto Piarulli
- Department of Science and High Technology, Università Degli Studi Dell’Insubria, Como, Italy
| | - Laura Belvisi
- Department of Chemistry, Università Degli Studi di Milano, Milan, Italy
| | - Monica Civera
- Department of Chemistry, Università Degli Studi di Milano, Milan, Italy
- *Correspondence: Monica Civera,
| |
Collapse
|
50
|
Maker A, Bolejack M, Schecterson L, Hammerson B, Abendroth J, Edwards TE, Staker B, Myler PJ, Gumbiner BM. Regulation of multiple dimeric states of E-cadherin by adhesion activating antibodies revealed through Cryo-EM and X-ray crystallography. PNAS NEXUS 2022; 1:pgac163. [PMID: 36157596 PMCID: PMC9491697 DOI: 10.1093/pnasnexus/pgac163] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/15/2022] [Indexed: 01/29/2023]
Abstract
E-cadherin adhesion is regulated at the cell surface, a process that can be replicated by activating antibodies. We use cryo-electron microscopy (EM) and X-ray crystallography to examine functional states of the cadherin adhesive dimer. This dimer is mediated by N-terminal beta strand-swapping involving Trp2, and forms via a different transient X-dimer intermediate. X-dimers are observed in cryo-EM along with monomers and strand-swap dimers, indicating that X-dimers form stable interactions. A novel EC4-mediated dimer was also observed. Activating Fab binding caused no gross structural changes in E-cadherin monomers, but can facilitate strand swapping. Moreover, activating Fab binding is incompatible with the formation of the X-dimer. Both cryo-EM and X-ray crystallography reveal a distinctive twisted strand-swap dimer conformation caused by an outward shift in the N-terminal beta strand that may represent a strengthened state. Thus, regulation of adhesion involves changes in cadherin dimer configurations.
Collapse
Affiliation(s)
- Allison Maker
- Department of Biochemistry, University of Washington, USA,Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, USA
| | - Madison Bolejack
- UCB Pharma, Bainbridge, WA, USA,Seattle Structural Genomics Center for Infectious Disease, USA
| | - Leslayann Schecterson
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, USA
| | - Brad Hammerson
- Seattle Structural Genomics Center for Infectious Disease, USA,Center for Global Infectious Disease Research, Seattle Children's Research Institute, USA
| | - Jan Abendroth
- UCB Pharma, Bainbridge, WA, USA,Seattle Structural Genomics Center for Infectious Disease, USA
| | - Thomas E Edwards
- UCB Pharma, Bainbridge, WA, USA,Seattle Structural Genomics Center for Infectious Disease, USA
| | - Bart Staker
- Seattle Structural Genomics Center for Infectious Disease, USA,Center for Global Infectious Disease Research, Seattle Children's Research Institute, USA
| | - Peter J Myler
- Seattle Structural Genomics Center for Infectious Disease, USA,Center for Global Infectious Disease Research, Seattle Children's Research Institute, USA,Department of Pediatrics, University of Washington, USA,Department of Biomedical Informatics and Medical Education, University of Washington, USA
| | | |
Collapse
|