1
|
Lialios P, Alimperti S. Role of E-cadherin in epithelial barrier dysfunction: implications for bacterial infection, inflammation, and disease pathogenesis. Front Cell Infect Microbiol 2025; 15:1506636. [PMID: 40007608 PMCID: PMC11850337 DOI: 10.3389/fcimb.2025.1506636] [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: 10/05/2024] [Accepted: 01/15/2025] [Indexed: 02/27/2025] Open
Abstract
Epithelial barriers serve as critical defense lines against microbial infiltration and maintain tissue homeostasis. E-cadherin, an essential component of adherens junctions, has emerged as a pivotal molecule that secures epithelial homeostasis. Lately, its pleiotropic role beyond barrier function, including its involvement in immune responses, has become more evident. Herein, we delve into the intricate relationship between (dys)regulation of epithelial homeostasis and the versatile functionality of E-cadherin, describing complex mechanisms that underlie barrier integrity and disruption in disease pathogenesis such as bacterial infection and inflammation, among others. Clinical implications of E-cadherin perturbations in host pathophysiology are emphasized; downregulation, proteolytic phenomena, abnormal localization/signaling and aberrant immune reactions are linked with a broad spectrum of pathology beyond infectious diseases. Finally, potential therapeutic interventions that may harness E-cadherin to mitigate barrier-associated tissue damage are explored. Overall, this review highlights the crucial role of E-cadherin in systemic health, offering insights that could pave the way for strategies to reinforce/restore barrier integrity and treat related diseases.
Collapse
Affiliation(s)
- Peter Lialios
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
- Center for Biological and Biomedical Engineering, Georgetown University, Washington, DC, United States
| | - Stella Alimperti
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
- Center for Biological and Biomedical Engineering, Georgetown University, Washington, DC, United States
| |
Collapse
|
2
|
Xie B, Xu S, Sivasankar S. Outside-in engineering of cadherin endocytosis using a conformation strengthening antibody. Nat Commun 2025; 16:1157. [PMID: 39881179 PMCID: PMC11779849 DOI: 10.1038/s41467-025-56478-6] [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: 08/15/2024] [Accepted: 01/15/2025] [Indexed: 01/31/2025] Open
Abstract
P-cadherin, a crucial cell-cell adhesion protein which is overexpressed in numerous malignant cancers, is a popular target for drug delivery antibodies. However, molecular guidelines for engineering antibodies that can be internalized upon binding to P-cadherin are unknown. Here, we use a combination of biophysical, biochemical, and cell biological methods to demonstrate that trapping the P-cadherin extracellular region in an X-dimer adhesive conformation triggers cadherin endocytosis via an outside-in signaling mechanism. We show that the anti-cancer drug delivery monoclonal antibody CQY684, traps P-cadherin in an X-dimer conformation and strengthens this adhesive structure. Formation of stable X-dimers results in the phosphorylation of p120-catenin, a suppressor of cadherin endocytosis. This triggers the dissociation of p120-catenin from the X-dimer cytoplasmic region, which increases P-cadherin turnover and targets the cadherin-antibody complex to the lysosome. Our results establish an outside-in signaling mechanism that provides fundamental insights into how cells regulate adhesion and that can be exploited by anti-cadherin antibodies for intracellular drug delivery.
Collapse
Affiliation(s)
- Bin Xie
- Biophysics Graduate Group, University of California, Davis, CA, USA
| | - Shipeng Xu
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Sanjeevi Sivasankar
- Biophysics Graduate Group, University of California, Davis, CA, USA.
- Department of Biomedical Engineering, University of California, Davis, CA, USA.
| |
Collapse
|
3
|
Quinn JM, Wang Y, Wood M, Flozak AS, Le PM, Yemelyanov A, Oakes PW, Gottardi CJ. α-catenin middle- and actin-binding domain unfolding mutants differentially impact epithelial strength and sheet migration. Mol Biol Cell 2024; 35:ar65. [PMID: 38507238 PMCID: PMC11151094 DOI: 10.1091/mbc.e23-01-0036] [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: 02/01/2023] [Revised: 02/23/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024] Open
Abstract
α-catenin (α-cat) displays force-dependent unfolding and binding to actin filaments through direct and indirect means, but features of adherens junction structure and function most vulnerable to loss of these allosteric mechanisms have not been directly compared. By reconstituting an α-cat F-actin-binding domain unfolding mutant known to exhibit enhanced binding to actin (α-cat-H0-FABD+) into α-cat knockout Madin Darby Canine Kidney (MDCK) cells, we show that partial loss of the α-cat catch bond mechanism (via an altered H0 α-helix) leads to stronger epithelial sheet integrity with greater colocalization between the α-cat-H0-FABD+ mutant and actin. α-cat-H0-FABD+ -expressing cells are less efficient at closing scratch-wounds, suggesting reduced capacity for more dynamic cell-cell coordination. Evidence that α-cat-H0-FABD+ is equally accessible to the conformationally sensitive α18 antibody epitope as WT α-cat and shows similar vinculin recruitment suggests this mutant engages lower tension cortical actin networks, as its M-domain is not persistently open. Conversely, α-cat-M-domain salt-bridge mutants with persistent recruitment of vinculin and phosphorylated myosin light chain show only intermediate monolayer adhesive strengths, but display less directionally coordinated and thereby slower migration speeds during wound-repair. These data show α-cat M- and FABD-unfolding mutants differentially impact cell-cell cohesion and migration properties, and suggest signals favoring α-cat-cortical actin interaction without persistent M-domain opening may improve epithelial monolayer strength through enhanced coupling to lower tension actin networks.
Collapse
Affiliation(s)
- Jeanne M. Quinn
- Department of Pulmonary Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
| | - Yuou Wang
- Department of Pulmonary Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
| | - Megan Wood
- Department of Pulmonary Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
| | - Annette S. Flozak
- Department of Pulmonary Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
| | - Phuong M. Le
- Department of Pulmonary Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
| | - Alex Yemelyanov
- Department of Pulmonary Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
| | - Patrick W. Oakes
- Department of Cell & Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153
| | - Cara J. Gottardi
- Department of Pulmonary Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
- Cell & Developmental Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611
| |
Collapse
|
4
|
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
|
5
|
Rudd JC, Maity S, Grunkemeyer JA, Snyder JC, Lovas S, Hansen LA. Membrane structure and internalization dynamics of human Flower isoforms hFWE3 and hFWE4 indicate a conserved endocytic role for hFWE4. J Biol Chem 2023; 299:104945. [PMID: 37348560 PMCID: PMC10366549 DOI: 10.1016/j.jbc.2023.104945] [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: 03/09/2023] [Revised: 06/09/2023] [Accepted: 06/12/2023] [Indexed: 06/24/2023] Open
Abstract
Human Flower (hFWE) isoforms hFWE1-4 are putative transmembrane (TM) proteins that reportedly mediate fitness comparisons during cell competition through extracellular display of their C-terminal tails. Isoform topology, subcellular localization, and duration of plasma membrane presentation are essential to this function. However, disagreement persists regarding the structure of orthologous fly and mouse FWEs, and experimental evidence for hFWE isoform subcellular localization or membrane structure is lacking. Here, we used AlphaFold2 and subsequent molecular dynamics-based structural predictions to construct epitope-tagged hFWE3 and hFWE4, the most abundant human isoforms, for experimental determination of their structure and internalization dynamics. We demonstrate that hFWE3 resides in the membrane of the endoplasmic reticulum (ER), while hFWE4 partially colocalizes with Rab4-, Rab5-, and Rab11-positive vesicles as well as with the plasma membrane. An array of imaging techniques revealed that hFWE4 positions both N- and C-terminal tails and a loop between second and third TM segments within the cytosol, while small (4-12aa) loops between the first and second and the third and fourth TM segments are either exposed to the extracellular space or within the lumen of cytoplasmic vesicles. Similarly, we found hFWE3 positions both N- and C-terminal tails in the cytosol, while a short loop between TM domains extends into the ER lumen. Finally, we demonstrate that hFWE4 exists only transiently at the cell surface and is rapidly internalized in an AP-2- and dynamin-1-dependent manner. Collectively, these data are consistent with a conserved role for hFWE4 in endocytic processes.
Collapse
Affiliation(s)
- Justin C Rudd
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, Nebraska, USA
| | - Sibaprasad Maity
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, Nebraska, USA
| | - James A Grunkemeyer
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, Nebraska, USA
| | - Joshua C Snyder
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Sándor Lovas
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, Nebraska, USA
| | - Laura A Hansen
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, Nebraska, USA.
| |
Collapse
|
6
|
Pulford CS, Uppalapati CK, Montgomery MR, Averitte RL, Hull EE, Leyva KJ. A Hybrid Epithelial to Mesenchymal Transition in Ex Vivo Cutaneous Squamous Cell Carcinoma Tissues. Int J Mol Sci 2022; 23:ijms23169183. [PMID: 36012449 PMCID: PMC9408944 DOI: 10.3390/ijms23169183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/09/2022] [Accepted: 08/14/2022] [Indexed: 11/16/2022] Open
Abstract
While most cases of cutaneous squamous cell carcinoma (cSCC) are benign, invasive cSCC is associated with higher mortality and is often more difficult to treat. As such, understanding the factors that influence the progression of cSCC are important. Aggressive cancers metastasize through a series of evolutionary changes, collectively called the epithelial-to-mesenchymal transition (EMT). During EMT, epithelial cells transition to a highly mobile mesenchymal cell type with metastatic capacities. While changes in expression of TGF-β, ZEB1, SNAI1, MMPs, vimentin, and E-cadherin are hallmarks of an EMT process occurring within cancer cells, including cSCC cells, EMT within tissues is not an “all or none” process. Using patient-derived cSCC and adjacent normal tissues, we show that cells within individual cSCC tumors are undergoing a hybrid EMT process, where there is variation in expression of EMT markers by cells within a tumor mass that may be facilitating invasion. Interestingly, cells along the outer edges of a tumor mass exhibit a more mesenchymal phenotype, with reduced E-cadherin, β-catenin, and cytokeratin expression and increased vimentin expression. Conversely, cells in the center of a tumor mass retain a higher expression of the epithelial markers E-cadherin and cytokeratin and little to no expression of vimentin, a mesenchymal marker. We also detected inverse expression changes in the miR-200 family and the EMT-associated transcription factors ZEB1 and SNAI1, suggesting that cSCC EMT dynamics are regulated in a miRNA-dependent manner. These novel findings in cSCC tumors provide evidence of phenotypic plasticity of the EMT process occurring within patient tissues, and extend the characterization of a hybrid EMT program occurring within a tumor mass. This hybrid EMT program may be promoting both survival and invasiveness of the tumors. A better understanding of this hybrid EMT process may influence therapeutic strategies in more invasive disease.
Collapse
Affiliation(s)
- Christopher S. Pulford
- Arizona College of Osteopathic Medicine, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA
| | - Chandana K. Uppalapati
- Department of Microbiology & Immunology, College of Graduate Studies, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA
| | | | - Richard L. Averitte
- Affiliated Dermatology & Affiliated Laboratories, 20401 N. 73rd Street #230, Scottsdale, AZ 85255, USA
| | - Elizabeth E. Hull
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA
| | - Kathryn J. Leyva
- Department of Microbiology & Immunology, College of Graduate Studies, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308, USA
- Correspondence: ; Tel.: 1-623-572-3294
| |
Collapse
|
7
|
Ramírez Moreno M, Bulgakova NA. The Cross-Talk Between EGFR and E-Cadherin. Front Cell Dev Biol 2022; 9:828673. [PMID: 35127732 PMCID: PMC8811214 DOI: 10.3389/fcell.2021.828673] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 12/31/2021] [Indexed: 12/18/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) and adhesion protein E-cadherin are major regulators of proliferation and differentiation in epithelial cells. Consistently, defects in both EGFR and E-cadherin-mediated intercellular adhesion are linked to various malignancies. These defects in either are further exacerbated by the reciprocal interactions between the two transmembrane proteins. On the one hand, EGFR can destabilize E-cadherin adhesion by increasing E-cadherin endocytosis, modifying its interactions with cytoskeleton and decreasing its expression, thus promoting tumorigenesis. On the other hand, E-cadherin regulates EGFR localization and tunes its activity. As a result, loss and mutations of E-cadherin promote cancer cell invasion due to uncontrolled activation of EGFR, which displays enhanced surface motility and changes in endocytosis. In this minireview, we discuss the molecular and cellular mechanisms of the cross-talk between E-cadherin and EGFR, highlighting emerging evidence for the role of endocytosis in this feedback, as well as its relevance to tissue morphogenesis, homeostasis and cancer progression.
Collapse
Affiliation(s)
| | - Natalia A. Bulgakova
- School of Biosciences and Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
8
|
Koshino A, Nagano A, Ota A, Hyodo T, Ueki A, Komura M, Sugimura-Nagata A, Ebi M, Ogasawara N, Kasai K, Hosokawa Y, Kasugai K, Takahashi S, Inaguma S. PBK Enhances Cellular Proliferation With Histone H3 Phosphorylation and Suppresses Migration and Invasion With CDH1 Stabilization in Colorectal Cancer. Front Pharmacol 2022; 12:772926. [PMID: 35115926 PMCID: PMC8804381 DOI: 10.3389/fphar.2021.772926] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/09/2021] [Indexed: 12/20/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most frequent gastrointestinal malignancies with high morbidity and mortality rates. Several biological markers for the prognostication of patient outcome of CRCs are available. Recently, our group identified two favorable factors for the survival of CRC patients: PDZ-binding kinase (PBK) and phospho-histone H3 (PHH3). Both showed a significant inverse association to pT stage. The aim of this study was to uncover the mechanism through which these cellular proliferation–associated protein expressions lead to favorable clinical outcome in CRC patients. We first confirmed co-expression of PBK and PHH3 in CRC cells. Further investigation showed that aberrantly expressed PBK up-regulated the cellular proliferation of CRC cells with accumulation of PHH3. The PBK inhibitor OTS514 suppressed cellular proliferation of CRC cells through down-regulation of PHH3 and induction of apoptosis. In vitro studies revealed that PBK suppressed the migration and invasion of CRC cells with suppression of Wnt/β-catenin signaling and CDH1 stabilization. Exogeneous PBK up-regulated the phosphorylated CDH1 at S840, S846, and S847 residues in cultured cells. Recombinant PBK directly phosphorylated HH3; however, it failed to phosphorylate CDH1 directly in vitro. The present study demonstrated the association of two markers PBK and PHH3 in CRC. We further identified one of the potential mechanisms by which higher expression of these cellular proliferation–associated proteins leads to the better survival of CRC patients, which likely involves PBK-mediated suppression of the migration and invasion of CRC cells. Our findings suggest that PBK-targeting therapeutics may be useful for the treatment of CRC patients with PBK-expressing tumors.
Collapse
Affiliation(s)
- Akira Koshino
- Division of Gastroenterology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Aya Nagano
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akinobu Ota
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Toshinori Hyodo
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Akane Ueki
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Masayuki Komura
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akane Sugimura-Nagata
- Division of Gastroenterology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Masahide Ebi
- Division of Gastroenterology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Naotaka Ogasawara
- Division of Gastroenterology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Kenji Kasai
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Yoshitaka Hosokawa
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Kunio Kasugai
- Division of Gastroenterology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Satoru Takahashi
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shingo Inaguma
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Department of Pathology, Aichi Medical University School of Medicine, Nagakute, Japan
- Department of Pathology, Nagoya City University East Medical Center, Nagoya, Japan
- *Correspondence: Shingo Inaguma,
| |
Collapse
|
9
|
How multisite phosphorylation impacts the conformations of intrinsically disordered proteins. PLoS Comput Biol 2021; 17:e1008939. [PMID: 33945530 PMCID: PMC8148376 DOI: 10.1371/journal.pcbi.1008939] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 05/25/2021] [Accepted: 04/06/2021] [Indexed: 12/15/2022] Open
Abstract
Phosphorylation of intrinsically disordered proteins (IDPs) can produce changes in structural and dynamical properties and thereby mediate critical biological functions. How phosphorylation effects intrinsically disordered proteins has been studied for an increasing number of IDPs, but a systematic understanding is still lacking. Here, we compare the collapse propensity of four disordered proteins, Ash1, the C-terminal domain of RNA polymerase (CTD2’), the cytosolic domain of E-Cadherin, and a fragment of the p130Cas, in unphosphorylated and phosphorylated forms using extensive all-atom molecular dynamics (MD) simulations. We find all proteins to show V-shape changes in their collapse propensity upon multi-site phosphorylation according to their initial net charge: phosphorylation expands neutral or overall negatively charged IDPs and shrinks positively charged IDPs. However, force fields including those tailored towards and commonly used for IDPs overestimate these changes. We find quantitative agreement of MD results with SAXS and NMR data for Ash1 and CTD2’ only when attenuating protein electrostatic interactions by using a higher salt concentration (e.g. 350 mM), highlighting the overstabilization of salt bridges in current force fields. We show that phosphorylation of IDPs also has a strong impact on the solvation of the protein, a factor that in addition to the actual collapse or expansion of the IDP should be considered when analyzing SAXS data. Compared to the overall mild change in global IDP dimension, the exposure of active sites can change significantly upon phosphorylation, underlining the large susceptibility of IDP ensembles to regulation through post-translational modifications. Intrinsically disordered proteins (IDPs) are a class of proteins that lack secondary and tertiary structures and instead explore a broad conformational ensemble. Their functions, from transcriptional regulation to signal transmission, are tightly regulated. IDPs are subject of extensive reversible post-translational modifications (PTMs), such as phosphorylation, methylation and glycosylation. Among these PTMs, phosphorylation is one of the most common and important PTMs. However, the mechanism of how phosphorylation affects the conformations and functions of IDPs remains unclear. To answer this question, we have performed extensive all-atom molecular dynamics simulations for four representative IDPs: Ash1, E-Cadherin, CTD2’ and p130Cas in their unphosphorylated and phosphorylated forms. Our results showed that all IDPs undergo a mild change upon multi-site phosphorylation, which is V-shaped: phosphorylation moderately expands neutral or overall negatively charged IDPs and shrinks positively charged IDPs. More importantly, in two of these IDPs, only two biologically relevant phosphorylation sites suffice to render the adjacent negatively charged active site significantly more exposed to the environment, which implies a higher probability to interact with other binding partners.
Collapse
|
10
|
Herrera A, Menendez A, Torroba B, Ochoa A, Pons S. Dbnl and β-catenin promote pro-N-cadherin processing to maintain apico-basal polarity. J Cell Biol 2021; 220:212044. [PMID: 33939796 PMCID: PMC8097490 DOI: 10.1083/jcb.202007055] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 02/15/2021] [Accepted: 03/15/2021] [Indexed: 12/30/2022] Open
Abstract
The neural tube forms when neural stem cells arrange into a pseudostratified, single-cell–layered epithelium, with a marked apico-basal polarity, and in which adherens junctions (AJs) concentrate in the subapical domain. We previously reported that sustained β-catenin expression promotes the formation of enlarged apical complexes (ACs), enhancing apico-basal polarity, although the mechanism through which this occurs remained unclear. Here, we show that β-catenin interacts with phosphorylated pro-N-cadherin early in its transit through the Golgi apparatus, promoting propeptide excision and the final maturation of N-cadherin. We describe a new β-catenin–dependent interaction of N-cadherin with Drebrin-like (Dbnl), an actin-binding protein that is involved in anterograde Golgi trafficking of proteins. Notably, Dbnl knockdown led to pro-N-cadherin accumulation and limited AJ formation. In brief, we demonstrate that Dbnl and β-catenin assist in the maturation of pro-N-cadherin, which is critical for AJ formation and for the recruitment AC components like aPKC and, consequently, for the maintenance of apico-basal polarity.
Collapse
Affiliation(s)
- Antonio Herrera
- Instituto de Biología Molecular de Barcelona, Parc Científic de Barcelona, Barcelona, Spain
| | - Anghara Menendez
- Instituto de Biología Molecular de Barcelona, Parc Científic de Barcelona, Barcelona, Spain
| | - Blanca Torroba
- Instituto de Biología Molecular de Barcelona, Parc Científic de Barcelona, Barcelona, Spain
| | - Andrea Ochoa
- Instituto de Biología Molecular de Barcelona, Parc Científic de Barcelona, Barcelona, Spain
| | - Sebastián Pons
- Instituto de Biología Molecular de Barcelona, Parc Científic de Barcelona, Barcelona, Spain
| |
Collapse
|
11
|
Erasmus JC, Smolarczyk K, Brezovjakova H, Mohd-Naim NF, Lozano E, Matter K, Braga VMM. Rac1-PAK1 regulation of Rab11 cycling promotes junction destabilization. J Cell Biol 2021; 220:212034. [PMID: 33914026 PMCID: PMC8091128 DOI: 10.1083/jcb.202002114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 09/21/2020] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
Rac1 GTPase is hyperactivated in tumors and contributes to malignancy. Rac1 disruption of junctions requires its effector PAK1, but the precise mechanisms are unknown. Here, we show that E-cadherin is internalized via micropinocytosis in a PAK1–dependent manner without catenin dissociation and degradation. In addition to internalization, PAK1 regulates E-cadherin transport by fine-tuning Rab small GTPase function. PAK1 phosphorylates a core Rab regulator, RabGDIβ, but not RabGDIα. Phosphorylated RabGDIβ preferentially associates with Rab5 and Rab11, which is predicted to promote Rab retrieval from membranes. Consistent with this hypothesis, Rab11 is activated by Rac1, and inhibition of Rab11 function partially rescues E-cadherin destabilization. Thus, Rac1 activation reduces surface cadherin levels as a net result of higher bulk flow of membrane uptake that counteracts Rab11-dependent E-cadherin delivery to junctions (recycling and/or exocytosis). This unique small GTPase crosstalk has an impact on Rac1 and PAK1 regulation of membrane remodeling during epithelial dedifferentiation, adhesion, and motility.
Collapse
Affiliation(s)
- Jennifer C Erasmus
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Kasia Smolarczyk
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Helena Brezovjakova
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Noor F Mohd-Naim
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Encarnación Lozano
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Karl Matter
- Institute of Ophthalmology, University College London, London, UK
| | - Vania M M Braga
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
12
|
Bastounis EE, Serrano-Alcalde F, Radhakrishnan P, Engström P, Gómez-Benito MJ, Oswald MS, Yeh YT, Smith JG, Welch MD, García-Aznar JM, Theriot JA. Mechanical competition triggered by innate immune signaling drives the collective extrusion of bacterially infected epithelial cells. Dev Cell 2021; 56:443-460.e11. [PMID: 33621492 PMCID: PMC7982222 DOI: 10.1016/j.devcel.2021.01.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 11/02/2020] [Accepted: 01/20/2021] [Indexed: 02/06/2023]
Abstract
Intracellular pathogens alter their host cells' mechanics to promote dissemination through tissues. Conversely, host cells may respond to the presence of pathogens by altering their mechanics to limit infection. Here, we monitored epithelial cell monolayers infected with intracellular bacterial pathogens, Listeria monocytogenes or Rickettsia parkeri, over days. Under conditions in which these pathogens trigger innate immune signaling through NF-κB and use actin-based motility to spread non-lytically intercellularly, we found that infected cell domains formed three-dimensional mounds. These mounds resulted from uninfected cells moving toward the infection site, collectively squeezing the softer and less contractile infected cells upward and ejecting them from the monolayer. Bacteria in mounds were less able to spread laterally in the monolayer, limiting the growth of the infection focus, while extruded infected cells underwent cell death. Thus, the coordinated forceful action of uninfected cells actively eliminates large domains of infected cells, consistent with this collective cell response representing an innate immunity-driven process.
Collapse
Affiliation(s)
- Effie E Bastounis
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | | | - Prathima Radhakrishnan
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA; Biophysics Program, Stanford University, Stanford, CA 94305, USA
| | - Patrik Engström
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - María J Gómez-Benito
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza 50009, Spain
| | - Mackenzi S Oswald
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Yi-Ting Yeh
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jason G Smith
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Matthew D Welch
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - José M García-Aznar
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza 50009, Spain
| | - Julie A Theriot
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
13
|
Gao L, Yang TT, Zhang JS, Liu HX, Cai DC, Wang LT, Wang J, Li XW, Gao K, Zhang SY, Cao YJ, Ji XX, Yang MM, Han B, Wang S, He L, Nie XY, Liu DM, Meng G, He CY. THBS1/CD47 Modulates the Interaction of γ-Catenin With E-Cadherin and Participates in Epithelial-Mesenchymal Transformation in Lipid Nephrotoxicity. Front Cell Dev Biol 2021; 8:601521. [PMID: 33681182 PMCID: PMC7930485 DOI: 10.3389/fcell.2020.601521] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/31/2020] [Indexed: 12/28/2022] Open
Abstract
Hyperlipidemia, an important risk factor for cardiovascular and end-stage renal diseases, often aggravates renal injury and compromises kidney function. Here, histological analysis of human kidney samples revealed that high lipid levels induced the development of renal fibrosis. To elucidate the mechanism underlying lipid nephrotoxicity, we used two types of mouse models (Apoe−/− and C57BL/6 mice fed a 45 and 60% high-fat diet, respectively). Histological analysis of kidney tissues revealed high-lipid-induced renal fibrosis and inflammation; this was confirmed by examining fibrotic and inflammatory marker expression using Western blotting and real-time polymerase chain reaction. Oxidized low-density lipoprotein (OX-LDL) significantly induced the fibrotic response in HK-2 tubular epithelial cells. RNA-sequencing and Gene Ontology analysis of differentially expressed mRNAs in OX-LDL-treated HK-2 tubular epithelial cells and real-time PCR validation in Apoe−/− mice showed that the expression of thrombospondin-1 (THBS1) in the high-fat group was significantly higher than that of the other top known genes, along with significant overexpression of its receptor CD47. THBS1 knockdown cells verified its relation to OX-LDL-induced fibrosis and inflammation. Liquid chromatography tandem mass spectrometry and STRING functional protein association network analyses predicted that THBS1/CD47 modulated the interaction between γ-catenin and E-cadherin and was involved in epithelial–mesenchymal transition, which was supported by immunoprecipitation and immunohistochemistry. CD47 downregulation following transfection with small-hairpin RNA in OX-LDL-treated tubular epithelial cells and treatment with anti-CD47 antibody restored the expression of E-cadherin and attenuated renal injury, fibrosis, and inflammatory response in OX-LDL-treated cells and in type 2 diabetes mellitus. These findings indicate that CD47 may serve as a potential therapeutic target in long-term lipid-induced kidney injury.
Collapse
Affiliation(s)
- Li Gao
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Ting-Ting Yang
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Jun-Sheng Zhang
- Pathophysiology Department, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hong-Xia Liu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Dong-Cheng Cai
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Lin-Tao Wang
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Jing Wang
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Xin-Wei Li
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Kun Gao
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Su-Ya Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Yu-Jia Cao
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Xiao-Xia Ji
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Miao-Miao Yang
- Pathophysiology Department, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Yizhiben Center for Judicial Expertise, Hefei, China
| | - Biao Han
- Pathophysiology Department, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Yizhiben Center for Judicial Expertise, Hefei, China
| | - Sheng Wang
- Center for Scientific Research of Anhui Medical University, Hefei, China
| | - Lu He
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Xiao-Yan Nie
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Dan-Mei Liu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Gang Meng
- Pathophysiology Department, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Yizhiben Center for Judicial Expertise, Hefei, China
| | - Chao-Yong He
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
14
|
Zeng C, Sun B, Cao X, Zhu H, Oluwadahunsi OM, Liu D, Zhu H, Zhang J, Zhang Q, Zhang G, Gibbons CA, Liu Y, Zhou J, Wang PG. Chemical Synthesis of Homogeneous Human E-Cadherin N-Linked Glycopeptides: Stereoselective Convergent Glycosylation and Chemoselective Solid-Phase Aspartylation. Org Lett 2020; 22:8349-8353. [PMID: 33045166 DOI: 10.1021/acs.orglett.0c02971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We report herein an efficient chemical synthesis of homogeneous human E-cadherin N-linked glycopeptides consisting of a heptapeptide sequence adjacent to the Asn-633 N-glycosylation site with representative N-glycan structures, including a conserved trisaccharide, a core-fucosylated tetrasaccharide, and a complex-type biantennary octasaccharide. The key steps are a chemoselective on-resin aspartylation using a pseudoproline-containing peptide and stereoselective glycosylation using glycosyl fluororide as a donor. This synthetic strategy demonstrates potential utility in accessing a wide range of homogeneous N-linked glycopeptides for the examination of their biological function.
Collapse
Affiliation(s)
- Chen Zeng
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Bin Sun
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xuefeng Cao
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | - Hailiang Zhu
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | | | - Ding Liu
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | - He Zhu
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | - Jiabin Zhang
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | - Qing Zhang
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | - Gaolan Zhang
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | | | - Yunpeng Liu
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| | - Jun Zhou
- Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States.,R&D Headquarters, WuXi AppTec, Shanghai 200131, China
| | - Peng George Wang
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China.,Department of Chemistry, Georgia State University, Atlanta, Georgia 30303, United States
| |
Collapse
|
15
|
Jansens RJJ, Marmiroli S, Favoreel HW. An Unbiased Approach to Mapping the Signaling Network of the Pseudorabies Virus US3 Protein. Pathogens 2020; 9:pathogens9110916. [PMID: 33167340 PMCID: PMC7694389 DOI: 10.3390/pathogens9110916] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 01/18/2023] Open
Abstract
The US3 serine/threonine protein kinase is conserved among the alphaherpesvirus family and represents an important virulence factor. US3 plays a role in viral nuclear egress, induces dramatic alterations of the cytoskeleton, represses apoptosis, enhances gene expression and modulates the immune response. Although several substrates of US3 have been identified, an unbiased screen to identify US3 phosphorylation targets has not yet been described. Here, we perform a shotgun and phosphoproteomics analysis of cells expressing the US3 protein of pseudorabies virus (PRV) to identify US3 phosphorylation targets in an unbiased way. We identified several cellular proteins that are differentially phosphorylated upon US3 expression and validated the phosphorylation of lamin A/C at serine 404, both in US3-transfected and PRV-infected cells. These results provide new insights into the signaling network of the US3 protein kinase and may serve as a basis for future research into the role of the US3 protein in the viral replication cycle.
Collapse
Affiliation(s)
- Robert J. J. Jansens
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium;
| | - Sandra Marmiroli
- Cellular Signaling Laboratory, Department of Surgery, Medicine, Dentistry, and Morphology, University of Modena & Reggio Emilia, 41121 Modena, Italy;
| | - Herman W. Favoreel
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium;
- Correspondence:
| |
Collapse
|
16
|
Piprek RP, Kloc M, Mizia P, Kubiak JZ. The Central Role of Cadherins in Gonad Development, Reproduction, and Fertility. Int J Mol Sci 2020; 21:E8264. [PMID: 33158211 PMCID: PMC7663743 DOI: 10.3390/ijms21218264] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/25/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022] Open
Abstract
Cadherins are a group of membrane proteins responsible for cell adhesion. They are crucial for cell sorting and recognition during the morphogenesis, but they also play many other roles such as assuring tissue integrity and resistance to stretching, mechanotransduction, cell signaling, regulation of cell proliferation, apoptosis, survival, carcinogenesis, etc. Within the cadherin superfamily, E- and N-cadherin have been especially well studied. They are involved in many aspects of sexual development and reproduction, such as germline development and gametogenesis, gonad development and functioning, and fertilization. E-cadherin is expressed in the primordial germ cells (PGCs) and also participates in PGC migration to the developing gonads where they become enclosed by the N-cadherin-expressing somatic cells. The differential expression of cadherins is also responsible for the establishment of the testis or ovary structure. In the adult testes, N-cadherin is responsible for the integrity of the seminiferous epithelium, regulation of sperm production, and the establishment of the blood-testis barrier. Sex hormones regulate the expression and turnover of N-cadherin influencing the course of spermatogenesis. In the adult ovaries, E- and N-cadherin assure the integrity of ovarian follicles and the formation of corpora lutea. Cadherins are expressed in the mature gametes and facilitate the capacitation of sperm in the female reproductive tract and gamete contact during fertilization. The germ cells and accompanying somatic cells express a series of different cadherins; however, their role in gonads and reproduction is still unknown. In this review, we show what is known and unknown about the role of cadherins in the germline and gonad development, and we suggest topics for future research.
Collapse
Affiliation(s)
- Rafał P. Piprek
- Department of Comparative Anatomy, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland;
| | - Malgorzata Kloc
- The Houston Methodist Research Institute, Houston, TX 77030, USA;
- Department of Surgery, The Houston Methodist Hospital, Houston, TX 77030, USA
- MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Paulina Mizia
- Department of Comparative Anatomy, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387 Krakow, Poland;
| | - Jacek Z. Kubiak
- Cycle Group, Institute of Genetics and Development of Rennes, Faculty of Medicine, UnivRennes, UMR 6290 CNRS/UR1, F-35000 Rennes, France
- Department of Regenerative Medicine and Cell Biology, Military Institute of Hygiene and Epidemiology (WIHE), 01-163 Warsaw, Poland
| |
Collapse
|
17
|
Amable G, Martínez-León E, Picco ME, Nemirovsky SI, Rozengurt E, Rey O. Metformin inhibition of colorectal cancer cell migration is associated with rebuilt adherens junctions and FAK downregulation. J Cell Physiol 2020; 235:8334-8344. [PMID: 32239671 PMCID: PMC7529638 DOI: 10.1002/jcp.29677] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 03/06/2020] [Indexed: 01/04/2023]
Abstract
E-cadherin, a central component of the adherens junction (AJ), is a single-pass transmembrane protein that mediates cell-cell adhesion. The loss of E-cadherin surface expression, and therefore cell-cell adhesion, leads to increased cell migration and invasion. Treatment of colorectal cancer (CRC)-derived cells (SW-480 and HT-29) with 2.0 mM metformin promoted a redistribution of cytosolic E-cadherin to de novo formed puncta along the length of the contacting membranes of these cells. Metformin also promoted translocation from the cytosol to the plasma membrane of p120-catenin, another core component of the AJs. Furthermore, E-cadherin and p120-catenin colocalized with β-catenin at cell-cell contacts. Western blot analysis of lysates of CRC-derived cells revealed a substantial metformin-induced increase in the level of p120-catenin as well as E-cadherin phosphorylation on Ser838/840 , a modification associated with β-catenin/E-cadherin interaction. These modifications in E-cadherin, p120-catenin and β-catenin localization suggest that metformin induces rebuilding of AJs in CRC-derived cells. Those modifications were accompanied by the inhibition of focal adhesion kinase (FAK), as revealed by a significant decrease in the phosphorylation of FAK at Tyr397 and paxillin at Tyr118 . These changes were associated with a reduction in the numbers, but an increase in the size, of focal adhesions and by the inhibition of cell migration. Overall, these observations indicate that metformin targets multiple pathways associated with CRC development and progression.
Collapse
Affiliation(s)
- Gastón Amable
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas “José de San Martín”, Ciudad Autónoma de Buenos Aires, 1120, Argentina
| | - Eduardo Martínez-León
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas “José de San Martín”, Ciudad Autónoma de Buenos Aires, 1120, Argentina
| | - María Elisa Picco
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas “José de San Martín”, Ciudad Autónoma de Buenos Aires, 1120, Argentina
| | - Sergio I. Nemirovsky
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, 1428EGA, Argentina
| | - Enrique Rozengurt
- Unit of Signal Transduction and Gastrointestinal Cancer, Division of Digestive Diseases, Department of Medicine, CURE: Digestive Diseases Research Center and Molecular Biology Institute, David Geffen School of Medicine, University of California at Los Angeles, CA, 90095-1768, USA
| | - Osvaldo Rey
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas “José de San Martín”, Ciudad Autónoma de Buenos Aires, 1120, Argentina
| |
Collapse
|
18
|
Han P, Weng W, Chen Y, Cai Y, Wang Y, Wang M, Zhan H, Yuan C, Yu X, Shao M, Sun H. Niclosamide ethanolamine attenuates systemic lupus erythematosus and lupus nephritis in MRL/lpr mice. Am J Transl Res 2020; 12:5015-5031. [PMID: 33042403 PMCID: PMC7540117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 08/01/2020] [Indexed: 06/11/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease with multiple organ involvement. Lupus nephritis (LN) is a severe manifestation of the disease and the most common cause of mortality in SLE patients. The etiology of LN is multifactorial and accumulating evidence suggests that mitochondrial dysfunction contributes to LN initiation and progression. Mild mitochondrial uncoupler niclosamide ethanolamine salt (NEN) has recently been shown to be efficacious in the treatment of both diabetic kidney disease and non-diabetic adriamycin nephropathy. However, its role in autoimmune kidney disease has not been explored. Here, we report for the first time that NEN attenuated SLE and lupus nephritis in MRL/lpr mice. NEN treatment reduced urinary protein excretion and attenuated glomerular lesions in this model. NEN treatment also decreased urinary excretion of tubular injury biomarkers NGAL and Kim-1, restored renal tubule phenotypic alterations, inhibited tubular proliferation, and suppressed renal interstitial inflammation and fibrosis. In addition, NEN diet supplementation restored redox imbalance, promoted mitochondrial biogenesis, and improved energy dysregulation in the kidney. Importantly, NEN prevented the enlargement of lymph nodes and the spleen, and decreased serum anti-dsDNA antibody levels in the MRL/lpr mice. Therefore, our data suggest that this mild mitochondrial uncoupling agent has great potential for translational application as a novel therapy for autoimmune disease.
Collapse
Affiliation(s)
- Pengxun Han
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen, China
| | - Wenci Weng
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen, China
| | - Yinghui Chen
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen, China
| | - Yuchun Cai
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen, China
| | - Yao Wang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen, China
| | - Menghua Wang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen, China
| | - Hongyue Zhan
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen, China
| | - Changjian Yuan
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen, China
| | - Xuewen Yu
- Department of Pathology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineGuangzhou, China
| | - Mumin Shao
- Department of Pathology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineGuangzhou, China
| | - Huili Sun
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese MedicineShenzhen, China
| |
Collapse
|
19
|
Kretschmer T, Turnwald EM, Janoschek R, Zentis P, Bae-Gartz I, Beers T, Handwerk M, Wohlfarth M, Ghilav M, Bloch W, Hucklenbruch-Rother E, Dötsch J, Appel S. Maternal high fat diet-induced obesity affects trophoblast differentiation and placental function in mice†. Biol Reprod 2020; 103:1260-1274. [PMID: 32915209 DOI: 10.1093/biolre/ioaa166] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 08/17/2020] [Accepted: 09/09/2020] [Indexed: 02/07/2023] Open
Abstract
Evidence suggests that maternal obesity (MO) can aggravate placental function causing severe pathologies during the perinatal window. However, molecular changes and mechanisms of placental dysfunction remain largely unknown. This work aimed to decipher structural and molecular alterations of the placental transfer zone associated with MO. To this end, mice were fed a high fat diet (HFD) to induce obesity before mating, and pregnant dams were sacrificed at E15.5 to receive placentas for molecular, histological, and ultrastructural analysis and to assess unidirectional materno-fetal transfer capacity. Laser-capture microdissection was used to collect specifically placental cells of the labyrinth zone for proteomics profiling. Using BeWo cells, fatty acid-mediated mechanisms of adherens junction stability, cell layer permeability, and lipid accumulation were deciphered. Proteomics profiling revealed downregulation of cell adhesion markers in the labyrinth zone of obese dams, and disturbed syncytial fusion and detachment of the basement membrane (BM) within this zone was observed, next to an increase in materno-fetal transfer in vivo across the placenta. We found that fetuses of obese dams develop a growth restriction and in those placentas, labyrinth zone volume-fraction was significantly reduced. Linoleic acid was shown to mediate beta-catenin level and increase cell layer permeability in vitro. Thus, MO causes fetal growth restriction, molecular and structural changes in the transfer zone leading to impaired trophoblast differentiation, BM disruption, and placental dysfunction despite increased materno-fetal transfer capacity. These adverse effects are probably mediated by fatty acids found in HFD demonstrating the need for obesity treatment to mitigate placental dysfunction and prevent offspring pathologies.
Collapse
Affiliation(s)
- Tobias Kretschmer
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Eva-Maria Turnwald
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ruth Janoschek
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Peter Zentis
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Core Facility Imaging, University of Cologne, Cologne, Germany
| | - Inga Bae-Gartz
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Tim Beers
- Department of Anatomy I, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Marion Handwerk
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Maria Wohlfarth
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Mojgan Ghilav
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sports Medicine, Institute of Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Eva Hucklenbruch-Rother
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jörg Dötsch
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Sarah Appel
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
20
|
Zhang X, Zou Y, Liu Y, Cao Y, Zhu J, Zhang J, Chen X, Zhang R, Li J. Inhibition of PIM1 kinase attenuates bleomycin-induced pulmonary fibrosis in mice by modulating the ZEB1/E-cadherin pathway in alveolar epithelial cells. Mol Immunol 2020; 125:15-22. [PMID: 32619930 DOI: 10.1016/j.molimm.2020.06.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/28/2020] [Accepted: 06/11/2020] [Indexed: 02/06/2023]
Abstract
PIM1 is serine/threonine protein kinase that is involved in numerous biological processes. Pulmonary fibrosis (PF) is a chronic pathological result of the dysfunctional repair of lung injury without effective therapeutic treatments. In the current study, we investigated whether PIM1 inhibition would improve bleomycin (BLM)-induced pulmonary fibrosis. In a BLM-induced pulmonary fibrosis model, PIM1 was persistently upregulated in fibrotic lung tissues. Furthermore, PIM1 inhibition by the PIM1-specific inhibitor SMI-4a showed protective effects against BLM-induced mortality. Furthermore, SMI-4a suppressed hydroxyproline deposition and reversed epithelial-mesenchymal transition (EMT) formation, which was characterized by E-cadherin and α-SMA expression in vivo. More importantly, the ZEB1/E-cadherin pathway was found to be closely associated with BLM-induced pulmonary fibrosis. After the in vitro treatment of A549 cells, PIM1 regulated E-cadherin expression by dependently modulating the activity of the transcription factor ZEB1. These findings were verified in vivo after SMI-4a administration. Finally, an shPIM1-expressing adeno-associated virus was delivered via intratracheal injection to induce a long-term PIM1 deficiency in the alveolar epithelium. AAV-mediated PIM1 knockdown in the lung tissues alleviated BLM-induced pulmonary fibrosis, as indicated by collagen accumulation reduction, pulmonary histopathological mitigation and EMT reversion. These findings enhance our understanding of the roles of PIM1 in BLM-induced pulmonary fibrosis and suggest PIM1 inhibition as a potential therapeutic strategy in chronic pulmonary injuries.
Collapse
Affiliation(s)
- Xinyi Zhang
- Department of Anesthesiology, Weifang Medical University, Weifang, China; Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yun Zou
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yuqi Liu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yumeng Cao
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jiali Zhu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jianhai Zhang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xia Chen
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Rui Zhang
- Department of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Jinbao Li
- Department of Anesthesiology, Weifang Medical University, Weifang, China; Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
21
|
Chen Y, Kotian N, Aranjuez G, Chen L, Messer CL, Burtscher A, Sawant K, Ramel D, Wang X, McDonald JA. Protein phosphatase 1 activity controls a balance between collective and single cell modes of migration. eLife 2020; 9:52979. [PMID: 32369438 PMCID: PMC7200163 DOI: 10.7554/elife.52979] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 04/23/2020] [Indexed: 12/12/2022] Open
Abstract
Collective cell migration is central to many developmental and pathological processes. However, the mechanisms that keep cell collectives together and coordinate movement of multiple cells are poorly understood. Using the Drosophila border cell migration model, we find that Protein phosphatase 1 (Pp1) activity controls collective cell cohesion and migration. Inhibition of Pp1 causes border cells to round up, dissociate, and move as single cells with altered motility. We present evidence that Pp1 promotes proper levels of cadherin-catenin complex proteins at cell-cell junctions within the cluster to keep border cells together. Pp1 further restricts actomyosin contractility to the cluster periphery rather than at individual internal border cell contacts. We show that the myosin phosphatase Pp1 complex, which inhibits non-muscle myosin-II (Myo-II) activity, coordinates border cell shape and cluster cohesion. Given the high conservation of Pp1 complexes, this study identifies Pp1 as a major regulator of collective versus single cell migration.
Collapse
Affiliation(s)
- Yujun Chen
- Division of Biology, Kansas State University, Manhattan, United States
| | - Nirupama Kotian
- Division of Biology, Kansas State University, Manhattan, United States
| | - George Aranjuez
- Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Lin Chen
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - C Luke Messer
- Division of Biology, Kansas State University, Manhattan, United States
| | - Ashley Burtscher
- Lerner Research Institute, Cleveland Clinic, Cleveland, United States
| | - Ketki Sawant
- Division of Biology, Kansas State University, Manhattan, United States
| | - Damien Ramel
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Xiaobo Wang
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | | |
Collapse
|
22
|
Huang C, Liu T, Wang Q, Hou W, Zhou C, Song Z, Shi YS, Gao X, Chen G, Yin Z, Hu Y. Loss of PP2A Disrupts the Retention of Radial Glial Progenitors in the Telencephalic Niche to Impair the Generation for Late-Born Neurons During Cortical Development†. Cereb Cortex 2020; 30:4183-4196. [PMID: 32186707 DOI: 10.1093/cercor/bhaa042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Telencephalic radial glial progenitors (RGPs) are retained in the ventricular zone (VZ), the niche for neural stem cells during cortical development. However, the underlying mechanism is not well understood. To study whether protein phosphatase 2A (PP2A) may regulate the above process, we generate Ppp2cα conditional knockout (cKO) mice, in which PP2A catalytic subunit α (PP2Acα) is inactivated in neural progenitor cells in the dorsal telencephalon. We show that RGPs are ectopically distributed in cortical areas outside of the VZ in Ppp2cα cKO embryos. Whereas deletion of PP2Acα does not affect the proliferation of RGPs, it significantly impairs the generation of late-born neurons. We find complete loss of apical adherens junctions (AJs) in the ventricular membrane in Ppp2cα cKO cortices. We observe abundant colocalization for N-cadherin and PP2Acα in control AJs. Moreover, in vitro analysis reveals direct interactions of N-cadherin to PP2Acα and to β-catenin. Overall, this study not only uncovers a novel function of PP2Acα in retaining RGPs into the VZ but also demonstrates the impact of PP2A-dependent retention of RGPs on the generation for late-born neurons.
Collapse
Affiliation(s)
- Chaoli Huang
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China
| | - Tingting Liu
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China
| | - Qihui Wang
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China
| | - Weikang Hou
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China
| | - Cuihua Zhou
- Department of Anesthesiology, The Second Affiliated Changzhou People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Zeyuan Song
- Department of Anesthesiology, The Second Affiliated Changzhou People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Yun Stone Shi
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China
| | - Xiang Gao
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China
| | - Guiquan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China
| | - Zhenyu Yin
- Department of Geriatric, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, China
| | - Yimin Hu
- Department of Anesthesiology, The Second Affiliated Changzhou People's Hospital of Nanjing Medical University, Changzhou 213000, China
| |
Collapse
|
23
|
Zaky DA, Abouelfadl DM, Nassar NN, Abdallah DM, Al-Shorbagy MY. The paradox of dipeptidyl peptidase IV inhibition in enterocytic differentiation and epithelial-mesenchymal transition in rat cholestatic sepsis. Toxicol Appl Pharmacol 2020; 394:114956. [PMID: 32171571 DOI: 10.1016/j.taap.2020.114956] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/19/2020] [Accepted: 03/10/2020] [Indexed: 12/16/2022]
Abstract
Proper enterocytic proliferation/differentiation, besides providing adequate adherens junctions (AJ) integrity, are responsible for strengthening of the gut barrier that acts as a first line defense against endotoxemia. However, the preferential role of the underlying PI3K/Akt (PKB) axis in triggering enterocytic proliferation/differentiation signaling and AJ assembly is still obscure in sepsis. Additionally, the potential involvement of dipeptidyl peptidase (DPP)-IV in cholestatic sepsis has not yet been reported. Common bile duct ligation (CBDL) insult was performed in adult male Sprague-Dawley rats except for sham operated animals; three doses of vildagliptin (VLD3, 10 and 30 mg/kg/d; p.o) were administered for 10 consecutive days post CBDL. VLD3/10/30 dose-dependently decreased DPP-IV and elevated GLP-1, IGF-1, PI3K, pS473-Akt (PKB), pS9-GSK-3β, pS133-CREB and cyclin-D1. VLD3/10 reduced fever, portal/aortic endotoxin and IgG, body weight loss as well as ileal NF-κB, TNF-α, MPO, TBARS, subepithelial/pericryptal and submucosal collagen deposition, vimentin immunoreactivity, N-cadherin, Zeb1 and pY654-β-catenin but increased E-cadherin, NPSH and colon/spleen indices - effects that were quite the opposite of VLD30. Accordingly, maintaining proper enterocytic proliferation/differentiation and phosphorylation inputs consequent to adequate DPP-IV inhibition is integral to AJ assembly in cholestatic sepsis; however, perturbed signals by excessive suppression of the enzyme activity induce toxic effects manifested as AJ disassembly and EMT, hence gut leakage and overt endotoxemia.
Collapse
Affiliation(s)
- Doaa A Zaky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini street, Cairo, P.O. Box 11562, Egypt.
| | | | - Noha N Nassar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini street, Cairo, P.O. Box 11562, Egypt
| | - Dalaal M Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini street, Cairo, P.O. Box 11562, Egypt
| | - Muhammad Y Al-Shorbagy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini street, Cairo, P.O. Box 11562, Egypt; School of Pharmacy, NewGiza University, Giza, Egypt
| |
Collapse
|
24
|
Manshouri R, Coyaud E, Kundu ST, Peng DH, Stratton SA, Alton K, Bajaj R, Fradette JJ, Minelli R, Peoples MD, Carugo A, Chen F, Bristow C, Kovacs JJ, Barton MC, Heffernan T, Creighton CJ, Raught B, Gibbons DL. ZEB1/NuRD complex suppresses TBC1D2b to stimulate E-cadherin internalization and promote metastasis in lung cancer. Nat Commun 2019; 10:5125. [PMID: 31719531 PMCID: PMC6851102 DOI: 10.1038/s41467-019-12832-z] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 09/29/2019] [Indexed: 02/08/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related death worldwide, due in part to the propensity of lung cancer to metastasize. Aberrant epithelial-to-mesenchymal transition (EMT) is a proposed model for the initiation of metastasis. During EMT cell-cell adhesion is reduced allowing cells to dissociate and invade. Of the EMT-associated transcription factors, ZEB1 uniquely promotes NSCLC disease progression. Here we apply two independent screens, BioID and an Epigenome shRNA dropout screen, to define ZEB1 interactors that are critical to metastatic NSCLC. We identify the NuRD complex as a ZEB1 co-repressor and the Rab22 GTPase-activating protein TBC1D2b as a ZEB1/NuRD complex target. We find that TBC1D2b suppresses E-cadherin internalization, thus hindering cancer cell invasion and metastasis. Non-small cell lung cancer (NSCLC) is often associated with metastasis to the lungs. Here, the authors perform independent screens and identify NuRD as a co-repressor of ZEB1, and demonstrate TBC1D2b as a downstream target of ZEB1/NuRD complex regulating NSCLC metastasis.
Collapse
Affiliation(s)
- Roxsan Manshouri
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Etienne Coyaud
- Department of Medical Biophysics, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, M5S 1A1, Canada
| | - Samrat T Kundu
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - David H Peng
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sabrina A Stratton
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kendra Alton
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rakhee Bajaj
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jared J Fradette
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Rosalba Minelli
- Department of Cancer Genomics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Michael D Peoples
- Department of Cancer Genomics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Alessandro Carugo
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Fengju Chen
- Department of Medicine and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Christopher Bristow
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jeffrey J Kovacs
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Michelle C Barton
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Tim Heffernan
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Chad J Creighton
- Department of Medicine and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Brian Raught
- Department of Medical Biophysics, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, M5S 1A1, Canada
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA. .,Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
25
|
Cdh4 Down-Regulation Impairs in Vivo Infiltration and Malignancy in Patients Derived Glioblastoma Cells. Int J Mol Sci 2019; 20:ijms20164028. [PMID: 31426573 PMCID: PMC6718984 DOI: 10.3390/ijms20164028] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/13/2019] [Accepted: 08/15/2019] [Indexed: 12/17/2022] Open
Abstract
The high invasive phenotype of glioblastoma is one of the main causes of therapy inefficacy and tumor relapse. Cell adhesion molecules of the cadherin family are involved in cell migration and are known as master regulators of epithelial tumor invasiveness, but their role in glioblastoma is less understood. In particular, we recently demonstrated, in the syngeneic murine model, the occurrence of a previously undescribed cadherin switch between Cdh2 and Cdh4 during gliomagenesis, which is necessary for the acquisition of the highly infiltrative and tumorigenic phenotype of these cells. In the present study, we tested the role of Cdh4 in human gliomas. Our results on patient-derived glioma cells demonstrate a positive correlation between Cdh4 expression levels and the loss of cell-cell contact inhibition of proliferation controls that allows cells to proliferate over confluence. Moreover, the silencing of Cdh4 by artificial microRNAs induced a decrease in the infiltrative ability of human glioma cells both in vitro and in vivo. More strikingly, Cdh4 silencing induced an impairment of the tumorigenic potential of these cells after orthotopic transplantation in immunodeficient mice. Overall, we conclude that in human glioblastoma, Cdh4 can also actively contribute in regulating cell invasiveness and malignancy.
Collapse
|
26
|
Song Y, Ye M, Zhou J, Wang Z, Zhu X. Targeting E-cadherin expression with small molecules for digestive cancer treatment. Am J Transl Res 2019; 11:3932-3944. [PMID: 31396310 PMCID: PMC6684918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 06/28/2019] [Indexed: 06/10/2023]
Abstract
Digestive system cancers, mainly including gastric cancer, hepatocellular carcinoma, pancreatic cancer, and colorectal cancer, are major public health problems and lead to serious cancer-related deaths worldwide. Clinically, treatment strategies of these cancers include surgery, chemotherapy, and immunotherapy. Although successful resection and chemotherapeutic drugs have improved the treatment level, the survival rate of patients with advanced digestive system cancers remains still low primarily due to tumor metastasis. E-cadherin, the prototypical member of the type-1 classical cadherins, has been characrized as an important molecule in epithelial-mesenchymal transition (EMT) process. Loss of E-cadherin is able to induce EMT process, which is associated with cancer stem cells and drug resistance in human cancer. Therefore, restoring E-cadherin could be a useful strategy for reversal of EMT and overcoming drug resistance. In this review, we describe pharmacological small molecules targeting E-cadherin expression for the treatment of digestive system cancers, which have emerged in the recent 5 years. We hope these compounds could be potentially used for treating cancer in the near future.
Collapse
Affiliation(s)
- Yizuo Song
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325027, Zhejiang, China
| | - Miaomiao Ye
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325027, Zhejiang, China
| | - Junhan Zhou
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325027, Zhejiang, China
| | - Zhiwei Wang
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325027, Zhejiang, China
- Center of Scientific Research, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325027, Zhejiang, China
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBoston, MA, USA
| | - Xueqiong Zhu
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325027, Zhejiang, China
| |
Collapse
|
27
|
Song Y, Ye M, Zhou J, Wang ZW, Zhu X. Restoring E-cadherin Expression by Natural Compounds for Anticancer Therapies in Genital and Urinary Cancers. MOLECULAR THERAPY-ONCOLYTICS 2019; 14:130-138. [PMID: 31194121 PMCID: PMC6551504 DOI: 10.1016/j.omto.2019.04.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
E-cadherin plays a pivotal role in cancer progression, including the epithelial-mesenchymal transition (EMT) process and tumor metastasis. Loss of E-cadherin contributes to enhanced invasion and metastasis in human cancers. Therefore, restoring E-cadherin could be a potential approach for cancer therapy. Multiple natural compounds have been shown to possess anti-tumor activities through the regulation of key molecules in signaling pathways, including E-cadherin. In this review, we describe the numerous compounds that restore the expression of E-cadherin in genital and urinary malignancies. We further discuss the potential anti-tumor molecular mechanisms of these agents as the activators of E-cadherin in genital and urinary cancers. Although these compounds exhibit their potential to inhibit the development and progression of cancers, there are several challenges to developing them as therapeutic drugs for cancer patients. Poor bioavailability in vivo is the main disadvantage of these compounds. Modification of compound structures has produced actual improvements in bioavailability. Nanoparticle-based delivery systems could be useful to deliver the agents to targeted organs. These compounds could be new promising therapeutic agents for the treatment of human genital and urinary cancers. Further investigations are required to determine the safety and side effects of natural compounds using animal models prior to clinical trials.
Collapse
Affiliation(s)
- Yizuo Song
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Miaomiao Ye
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Junhan Zhou
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zhi-Wei Wang
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.,Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Xueqiong Zhu
- Departmant of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| |
Collapse
|
28
|
Schuhmacher D, Sontag JM, Sontag E. Protein Phosphatase 2A: More Than a Passenger in the Regulation of Epithelial Cell-Cell Junctions. Front Cell Dev Biol 2019; 7:30. [PMID: 30895176 PMCID: PMC6414416 DOI: 10.3389/fcell.2019.00030] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/22/2019] [Indexed: 12/17/2022] Open
Abstract
Cell–cell adhesion plays a key role in the maintenance of the epithelial barrier and apicobasal cell polarity, which is crucial for homeostasis. Disruption of cell–cell adhesion is a hallmark of numerous pathological conditions, including invasive carcinomas. Adhesion between apposing cells is primarily regulated by three types of junctional structures: desmosomes, adherens junctions, and tight junctions. Cell junctional structures are highly regulated multiprotein complexes that also serve as signaling platforms to control epithelial cell function. The biogenesis, integrity, and stability of cell junctions is controlled by complex regulatory interactions with cytoskeletal and polarity proteins, as well as modulation of key component proteins by phosphorylation/dephosphorylation processes. Not surprisingly, many essential signaling molecules, including protein Ser/Thr phosphatase 2A (PP2A) are associated with intercellular junctions. Here, we examine how major PP2A enzymes regulate epithelial cell–cell junctions, either directly by associating with and dephosphorylating component proteins, or indirectly by affecting signaling pathways that control junctional integrity and cytoskeletal dynamics. PP2A deregulation has severe consequences on the stability and functionality of these structures, and disruption of cell–cell adhesion and cell polarity likely contribute to the link between PP2A dysfunction and human carcinomas.
Collapse
Affiliation(s)
- Diana Schuhmacher
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
| | - Jean-Marie Sontag
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Estelle Sontag
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
29
|
Tiwari P, Mrigwani A, Kaur H, Kaila P, Kumar R, Guptasarma P. Structural-Mechanical and Biochemical Functions of Classical Cadherins at Cellular Junctions: A Review and Some Hypotheses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1112:107-138. [DOI: 10.1007/978-981-13-3065-0_9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
30
|
Chae WJ, Bothwell ALM. Canonical and Non-Canonical Wnt Signaling in Immune Cells. Trends Immunol 2018; 39:830-847. [PMID: 30213499 DOI: 10.1016/j.it.2018.08.006] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 08/12/2018] [Accepted: 08/15/2018] [Indexed: 12/18/2022]
Abstract
Cell differentiation, proliferation, and death are vital for immune homeostasis. Wnt signaling plays essential roles in processes across species. The roles of Wnt signaling proteins and Wnt ligands have been studied in the past, but the context-dependent mechanisms and functions of these pathways in immune responses remain unclear. Recent findings regarding the role of Wnt ligands and Wnt signaling in immune cells and their immunomodulatory mechanisms suggest that Wnt ligands and signaling are significant in regulating immune responses. We introduce recent key findings and future perspectives on Wnt ligands and their signaling pathways in immune cells as well as the immunological roles and functions of Wnt antagonists.
Collapse
Affiliation(s)
- Wook-Jin Chae
- Department of Immunobiology, Yale University School of Medicine, 300 Cedar Street, New Haven, CT 06520, USA.
| | - Alfred L M Bothwell
- Department of Immunobiology, Yale University School of Medicine, 300 Cedar Street, New Haven, CT 06520, USA.
| |
Collapse
|
31
|
Guo S, Liang X, Guo M, Zhang X, Li Z. Migration inhibition of water stress proteins from Nostoc commune Vauch. via activation of autophagy in DLD-1 cells. Int J Biol Macromol 2018; 119:669-676. [PMID: 30071226 DOI: 10.1016/j.ijbiomac.2018.07.188] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/20/2018] [Accepted: 07/28/2018] [Indexed: 12/27/2022]
Abstract
Water stress proteins (WSP1) from Nostoc commune Vauch. had been proven to selectively induce colon cancer cells apoptosis. In this study, the effect of WSP1 on migration of human colon cancer cells was investigated. It showed that WSP1 inhibited DLD-1 cell migration, but with an insignificant effect on normal human colon epithelial cells. The data further indicated that WSP1 activated autophagy through down regulation of PI3K/AKT/mTOR pathway. Meanwhile, β‑catenin was degraded by autophagy, which then restrained epithelial-mesenchymal transition (EMT) of DLD-1 cell and its migration was subsequently suppressed significantly. The same changes occurred in xenografted nude mice according to the obtained immunohistochemical results. Consistently, the application of autophagy inhibitor largely reversed the inhibited migration by WSP1 treatment. Taken together, WSP1 could suppress migration of DLD-1 cells by autophagy inhibited EMT. The results suggested that WSP1 possessed the potential as a selective therapeutic agent against metastatic colon cancer.
Collapse
Affiliation(s)
- Songjia Guo
- Shanxi Provincial People's Hospital, Center for Precision Medicine, Taiyuan 030006, China; Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China
| | - Xinxin Liang
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China
| | - Min Guo
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China
| | - Xiaoli Zhang
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China
| | - Zhuoyu Li
- Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
32
|
Yamada M, Takahashi N, Matsuda Y, Sato K, Yokoji M, Sulijaya B, Maekawa T, Ushiki T, Mikami Y, Hayatsu M, Mizutani Y, Kishino S, Ogawa J, Arita M, Tabeta K, Maeda T, Yamazaki K. A bacterial metabolite ameliorates periodontal pathogen-induced gingival epithelial barrier disruption via GPR40 signaling. Sci Rep 2018; 8:9008. [PMID: 29899364 PMCID: PMC5998053 DOI: 10.1038/s41598-018-27408-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 06/04/2018] [Indexed: 01/15/2023] Open
Abstract
Several studies have demonstrated the remarkable properties of microbiota and their metabolites in the pathogenesis of several inflammatory diseases. 10-Hydroxy-cis-12-octadecenoic acid (HYA), a bioactive metabolite generated by probiotic microorganisms during the process of fatty acid metabolism, has been studied for its protective effects against epithelial barrier impairment in the intestines. Herein, we examined the effect of HYA on gingival epithelial barrier function and its possible application for the prevention and treatment of periodontal disease. We found that GPR40, a fatty acid receptor, was expressed on gingival epithelial cells; activation of GPR40 by HYA significantly inhibited barrier impairment induced by Porphyromonas gingivalis, a representative periodontopathic bacterium. The degradation of E-cadherin and beta-catenin, basic components of the epithelial barrier, was prevented in a GPR40-dependent manner in vitro. Oral inoculation of HYA in a mouse experimental periodontitis model suppressed the bacteria-induced degradation of E-cadherin and subsequent inflammatory cytokine production in the gingival tissue. Collectively, these results suggest that HYA exerts a protective function, through GPR40 signaling, against periodontopathic bacteria-induced gingival epithelial barrier impairment and contributes to the suppression of inflammatory responses in periodontal diseases.
Collapse
Affiliation(s)
- Miki Yamada
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Naoki Takahashi
- Research Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| | - Yumi Matsuda
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Keisuke Sato
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Mai Yokoji
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Benso Sulijaya
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tomoki Maekawa
- Research Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tatsuo Ushiki
- Division of Microscopic Anatomy and Bio-imaging, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yoshikazu Mikami
- Division of Microscopic Anatomy and Bio-imaging, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Manabu Hayatsu
- Division of Microscopic Anatomy and Bio-imaging, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yusuke Mizutani
- Division of Microscopic Anatomy and Bio-imaging, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Shigenobu Kishino
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Jun Ogawa
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Koichi Tabeta
- Division of Periodontology, Department of Oral Biological Science, Niigata University Faculty of Dentistry, Niigata, Japan
| | - Takeyasu Maeda
- Research Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kazuhisa Yamazaki
- Research Unit for Oral-Systemic Connection, Division of Oral Science for Health Promotion, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| |
Collapse
|
33
|
Chen CL, Wang SH, Chan PC, Shen MR, Chen HC. Phosphorylation of E-cadherin at threonine 790 by protein kinase Cδ reduces β-catenin binding and suppresses the function of E-cadherin. Oncotarget 2018; 7:37260-37276. [PMID: 27203386 PMCID: PMC5095074 DOI: 10.18632/oncotarget.9403] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 04/10/2016] [Indexed: 02/07/2023] Open
Abstract
Proper control of cell-cell adhesion is crucial for embryogenesis and tissue homeostasis. In this study, we show that protein kinase C (PKC)δ, a member of the novel PKC subfamily, localizes at cell-cell contacts of epithelial cells through its C2-like domain in an F-actin-dependent manner. Upon hepatocyte growth factor stimulation, PKCδ is phosphorylated and activated by Src, which then phosphorylates E-cadherin at Thr790. Phosphorylation of E-cadherin at Thr790 diminishes its interaction with β-catenin and impairs the homophilic interaction between the ectodomains of E-cadherin. The suppression of PKCδ by its dominant-negative mutants or specific short-hairpin RNA inhibits the disruption of cell-cell adhesions induced by hepatocyte growth factor. Elevated PKCδ expression in cancer cells is correlated with increased phosphorylation of E-cadherin at Thr790, reduced binding of E-cadherin to β-catenin, and poor homophilic interaction between E-cadherin. Analysis of surgical specimens confirmed that PKCδ is overexpressed in cervical cancer tissues, accompanied by increased phosphorylation of E-cadherin at Thr790. Together, our findings unveil a negative role for PKCδ in cell-cell adhesion through phosphorylation of E-cadherin.
Collapse
Affiliation(s)
- Chien-Lin Chen
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan
| | - Shu-Hui Wang
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan
| | - Po-Chao Chan
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan
| | - Meng-Ru Shen
- Department of Pharmacology, National Cheng Kung University, Tainan 704, Taiwan.,Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, Tainan 704, Taiwan
| | - Hong-Chen Chen
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan.,Graduate Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402, Taiwan.,Rong-Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
| |
Collapse
|
34
|
Ortega FE, Rengarajan M, Chavez N, Radhakrishnan P, Gloerich M, Bianchini J, Siemers K, Luckett WS, Lauer P, Nelson WJ, Theriot JA. Adhesion to the host cell surface is sufficient to mediate Listeria monocytogenes entry into epithelial cells. Mol Biol Cell 2017; 28:2945-2957. [PMID: 28877987 PMCID: PMC5662255 DOI: 10.1091/mbc.e16-12-0851] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 08/29/2017] [Accepted: 08/31/2017] [Indexed: 12/02/2022] Open
Abstract
Listeria monocytogenes invades epithelial cells by binding to the host cell receptor E-cadherin, a component of the adherens junction. E-cadherin serves primarily as an adhesive to mediate bacterial invasion; the canonical E-cadherin/catenin/F-actin complex is not required for this process. The intestinal epithelium is the first physiological barrier breached by the Gram-positive facultative pathogen Listeria monocytogenes during an in vivo infection. Listeria monocytogenes binds to the epithelial host cell receptor E-cadherin, which mediates a physical link between the bacterium and filamentous actin (F-actin). However, the importance of anchoring the bacterium to F-actin through E-cadherin for bacterial invasion has not been tested directly in epithelial cells. Here we demonstrate that depleting αE-catenin, which indirectly links E-cadherin to F-actin, did not decrease L. monocytogenes invasion of epithelial cells in tissue culture. Instead, invasion increased due to increased bacterial adhesion to epithelial monolayers with compromised cell–cell junctions. Furthermore, expression of a mutant E-cadherin lacking the intracellular domain was sufficient for efficient L. monocytogenes invasion of epithelial cells. Importantly, direct biotin-mediated binding of bacteria to surface lipids in the plasma membrane of host epithelial cells was sufficient for uptake. Our results indicate that the only requirement for L. monocytogenes invasion of epithelial cells is adhesion to the host cell surface, and that E-cadherin–mediated coupling of the bacterium to F-actin is not required.
Collapse
Affiliation(s)
- Fabian E Ortega
- Department of Biochemistry, Stanford University, Stanford, CA 94305
| | | | - Natalie Chavez
- Department of Biology, Stanford University, Stanford, CA 94305
| | | | | | - Julie Bianchini
- Department of Biology, Stanford University, Stanford, CA 94305
| | | | | | | | - W James Nelson
- Department of Biology, Stanford University, Stanford, CA 94305.,Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA 94305
| | - Julie A Theriot
- Department of Biochemistry, Stanford University, Stanford, CA 94305 .,Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305.,Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305
| |
Collapse
|
35
|
Shao X, Kang H, Loveless T, Lee GR, Seok C, Weis WI, Choi HJ, Hardin J. Cell-cell adhesion in metazoans relies on evolutionarily conserved features of the α-catenin·β-catenin-binding interface. J Biol Chem 2017; 292:16477-16490. [PMID: 28842483 DOI: 10.1074/jbc.m117.795567] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/31/2017] [Indexed: 01/26/2023] Open
Abstract
Stable tissue integrity during embryonic development relies on the function of the cadherin·catenin complex (CCC). The Caenorhabditis elegans CCC is a useful paradigm for analyzing in vivo requirements for specific interactions among the core components of the CCC, and it provides a unique opportunity to examine evolutionarily conserved mechanisms that govern the interaction between α- and β-catenin. HMP-1, unlike its mammalian homolog α-catenin, is constitutively monomeric, and its binding affinity for HMP-2/β-catenin is higher than that of α-catenin for β-catenin. A crystal structure shows that the HMP-1·HMP-2 complex forms a five-helical bundle structure distinct from the structure of the mammalian α-catenin·β-catenin complex. Deletion analysis based on the crystal structure shows that the first helix of HMP-1 is necessary for binding HMP-2 avidly in vitro and for efficient recruitment of HMP-1 to adherens junctions in embryos. HMP-2 Ser-47 and Tyr-69 flank its binding interface with HMP-1, and we show that phosphomimetic mutations at these two sites decrease binding affinity of HMP-1 to HMP-2 by 40-100-fold in vitro. In vivo experiments using HMP-2 S47E and Y69E mutants showed that they are unable to rescue hmp-2(zu364) mutants, suggesting that phosphorylation of HMP-2 on Ser-47 and Tyr-69 could be important for regulating CCC formation in C. elegans Our data provide novel insights into how cadherin-dependent cell-cell adhesion is modulated in metazoans by conserved elements as well as features unique to specific organisms.
Collapse
Affiliation(s)
| | | | - Timothy Loveless
- Department of Zoology, and.,Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Gyu Rie Lee
- Chemistry, Seoul National University, Seoul 08826, South Korea, and
| | - Chaok Seok
- Chemistry, Seoul National University, Seoul 08826, South Korea, and
| | - William I Weis
- the Departments of Structural Biology and Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305
| | | | - Jeff Hardin
- From the Program in Genetics, .,Department of Zoology, and.,Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin 53706
| |
Collapse
|
36
|
Liu Y, Li H, Ban Z, Nai M, Yang L, Chen Y, Xu Y. Annexin A2 inhibition suppresses ovarian cancer progression via regulating β-catenin/EMT. Oncol Rep 2017; 37:3643-3650. [PMID: 28440436 DOI: 10.3892/or.2017.5578] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 03/16/2017] [Indexed: 01/23/2023] Open
Abstract
Annexin A2 is a member of the Annexin family that acts as a Ca2+-dependent phospholipid and membrane binding protein, which is associated with the survival and spread of multiple neoplasms. However, the function of Annexin A2 in ovarian cancer progression remains unclear. In this study, we aimed to investigate the role and underlying molecular mechanism of Annexin A2 in cell proliferation and invasion in ovarian cancer. We found that the mRNA expression of Annexin A2 was upregulated in ovarian cancer tissues and cell lines. In the loss-of-function of Annexin A2, β-catenin was indicated to be significantly suppressed and EMT constrained. Moreover, cell proliferation and invasion were both markedly inhibited by the downregulation of Annexin A2. Additionally, the overexpression of β-catenin obviously reversed the effect of Annexin A2 on EMT, and cell proliferation and invasion, indicating that Annexin A2 suppression regulated EMT through controlling β-catenin. Taken together, this study showed that Annexin A2 inhibition suppresses proliferation and invasion in ovarian cancer via β-catenin/EMT, proposing the potential role of Annexin A2 in the prevention and treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yan Liu
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Hongyu Li
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Zhenying Ban
- Department of Pathology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Manman Nai
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Li Yang
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Yannan Chen
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Yiming Xu
- Department of Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|
37
|
Chen YT, Tai CY. μ2-Dependent endocytosis of N-cadherin is regulated by β-catenin to facilitate neurite outgrowth. Traffic 2017; 18:287-303. [DOI: 10.1111/tra.12473] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 02/10/2017] [Accepted: 02/17/2017] [Indexed: 01/07/2023]
Affiliation(s)
- Yi-ting Chen
- Taiwan International Graduate Program, Molecular and Cellular Biology Program; Academia Sinica; Taiwan Republic of China
- Institute of Molecular Biology; Academia Sinica; Taiwan Republic of China
- Graduate Institute of Life Sciences, National Defense Medical Center; Taiwan Republic of China
| | - Chin-Yin Tai
- Taiwan International Graduate Program, Molecular and Cellular Biology Program; Academia Sinica; Taiwan Republic of China
- Institute of Molecular Biology; Academia Sinica; Taiwan Republic of China
- Development Center for Biotechnology; Institute of Pharmaceutics; Taiwan Republic of China
| |
Collapse
|
38
|
Chen YJ, Huang J, Huang L, Austin E, Hong Y. Phosphorylation potential of Drosophila E-Cadherin intracellular domain is essential for development and adherens junction biosynthetic dynamics regulation. Development 2017; 144:1242-1248. [PMID: 28219947 DOI: 10.1242/dev.141598] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 02/07/2017] [Indexed: 02/06/2023]
Abstract
Phosphorylation of a highly conserved serine cluster in the intracellular domain of E-Cadherin is essential for binding to β-Catenin in vitro In cultured cells, phosphorylation of specific serine residues within the cluster is also required for regulation of adherens junction (AJ) stability and dynamics. However, much less is known about how such phosphorylation of E-Cadherin regulates AJ formation and dynamics in vivo In this report, we generated an extensive array of Drosophila E-Cadherin (DE-Cad) endogenous knock-in alleles that carry mutations targeting this highly conserved serine cluster. Analyses of these mutations suggest that the overall phosphorylation potential, rather than the potential site-specific phosphorylation, of the serine cluster enhances the recruitment of β-Catenin by DE-Cad in vivo Moreover, phosphorylation potential of the serine cluster only moderately increases the level of β-Catenin in AJs and is in fact dispensable for AJ formation in vivo Nonetheless, phosphorylation-dependent recruitment of β-Catenin is essential for development, probably by enhancing the interactions between DE-Cad and α-Catenin. In addition, several phospho-mutations dramatically reduced the biosynthetic turnover rate of DE-Cad during apical-basal polarization, and such biosynthetically stable DE-Cad mutants specifically rescued the polarity defects in embryonic epithelia lacking the polarity proteins Stardust and Crumbs.
Collapse
Affiliation(s)
- Yi-Jiun Chen
- Department of Cell Biology, University of Pittsburgh Medical School, Pittsburgh, PA 15261, USA
| | - Juan Huang
- Department of Cell Biology, University of Pittsburgh Medical School, Pittsburgh, PA 15261, USA.,Department of Biotechnology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, People's Republic of China
| | - Lynn Huang
- Department of Cell Biology, University of Pittsburgh Medical School, Pittsburgh, PA 15261, USA
| | - Erin Austin
- Department of Cell Biology, University of Pittsburgh Medical School, Pittsburgh, PA 15261, USA
| | - Yang Hong
- Department of Cell Biology, University of Pittsburgh Medical School, Pittsburgh, PA 15261, USA
| |
Collapse
|
39
|
Anti-androgen 2-hydroxyflutamide modulates cadherin, catenin and androgen receptor phosphorylation in androgen-sensitive LNCaP and androgen-independent PC3 prostate cancer cell lines acting via PI3K/Akt and MAPK/ERK1/2 pathways. Toxicol In Vitro 2017; 40:324-335. [PMID: 28163245 DOI: 10.1016/j.tiv.2017.01.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 12/22/2016] [Accepted: 01/25/2017] [Indexed: 01/04/2023]
Abstract
This study aimed to investigate rapid effect of anti-androgen 2-hydroxyflutamide (HF) on cadherin/catenin complex and androgen receptor (AR) phosphorylation in prostate cancer cell lines. In addition, a role of PI3K/Akt and MAPK/ERK1/2 pathways in mediating these effects was explored. We have demonstrated that in androgen-sensitive LNCaP cells HF induced rapid increase of E-cadherin phosphorylation at Ser 838/840 (p<0.05) in MAPK/ERK1/2-dependent manner, whereas phosphorylation of β-catenin at Tyr 654 was unchanged. Concomitantly, the reduction of the level of AR phosphorylated at Ser210/213 was found (p<0.01). In androgen-independent PC3 cells HF decreased Tyr 860 N-cadherin and Tyr 645 β-catenin phosphorylation (p<0.01), acting via both MAPK/ERK1/2 and PI3K/Akt pathways. Further, we evidenced that MAPK/ERK1/2 and PI3K/Akt pathways were differentially influenced by HF in LNCaP and PC3 cells. In LNCaP cells, both Akt (p<0.01) and ERK1/2 (p<0.001) phosphorylation were negatively regulated and this effect was mediated by Raf-1 (p<0.05). In contrast, in PC3 cells HF stimulated Akt (p<0.001) and ERK1/2 (p<0.001) activation, but had no effect on the crosstalk between PI3K/Akt and MEK/ERK1/2 pathways at the Raf-1 kinase level. Our findings expand the role of anti-androgen into non-genomic signaling, creating a link between anti-androgen action and phosphorylation of adherens junction proteins in prostate cancer cells.
Collapse
|
40
|
Schiffmacher AT, Xie V, Taneyhill LA. Cadherin-6B proteolysis promotes the neural crest cell epithelial-to-mesenchymal transition through transcriptional regulation. J Cell Biol 2016; 215:735-747. [PMID: 27856599 PMCID: PMC5146998 DOI: 10.1083/jcb.201604006] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 09/19/2016] [Accepted: 10/25/2016] [Indexed: 12/25/2022] Open
Abstract
Cadherin proteolysis reduces cell–cell adhesion and generates cleavage products that could possess independent functions. Here, Schiffmacher et al. reveal that the intracellular C-terminal fragment generated by Cadherin-6B proteolysis promotes chick cranial neural crest cell EMT through positive transcriptional feedback into the neural crest gene regulatory network. During epithelial-to-mesenchymal transitions (EMTs), cells disassemble cadherin-based junctions to segregate from the epithelia. Chick premigratory cranial neural crest cells reduce Cadherin-6B (Cad6B) levels through several mechanisms, including proteolysis, to permit their EMT and migration. Serial processing of Cad6B by a disintegrin and metalloproteinase (ADAM) proteins and γ-secretase generates intracellular C-terminal fragments (CTF2s) that could acquire additional functions. Here we report that Cad6B CTF2 possesses a novel pro-EMT role by up-regulating EMT effector genes in vivo. After proteolysis, CTF2 remains associated with β-catenin, which stabilizes and redistributes both proteins to the cytosol and nucleus, leading to up-regulation of β-catenin, CyclinD1, Snail2, and Snail2 promoter-based GFP expression in vivo. A CTF2 β-catenin–binding mutant, however, fails to alter gene expression, indicating that CTF2 modulates β-catenin–responsive EMT effector genes. Notably, CTF2 association with the endogenous Snail2 promoter in the neural crest is β-catenin dependent. Collectively, our data reveal how Cad6B proteolysis orchestrates multiple pro-EMT regulatory inputs, including CTF2-mediated up-regulation of the Cad6B repressor Snail2, to ensure proper cranial neural crest EMT.
Collapse
Affiliation(s)
- Andrew T Schiffmacher
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742
| | - Vivien Xie
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742
| | - Lisa A Taneyhill
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742
| |
Collapse
|
41
|
Cadwell CM, Su W, Kowalczyk AP. Cadherin tales: Regulation of cadherin function by endocytic membrane trafficking. Traffic 2016; 17:1262-1271. [PMID: 27624909 DOI: 10.1111/tra.12448] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/09/2016] [Accepted: 09/09/2016] [Indexed: 12/17/2022]
Abstract
Cadherins are the primary adhesion molecules in adherens junctions and desmosomes and play essential roles in embryonic development. Although significant progress has been made in understanding cadherin structure and function, we lack a clear vision of how cells confer plasticity upon adhesive junctions to allow for cellular rearrangements during development, wound healing and metastasis. Endocytic membrane trafficking has emerged as a fundamental mechanism by which cells confer a dynamic state to adhesive junctions. Recent studies indicate that the juxtamembrane domain of classical cadherins contains multiple endocytic motifs, or "switches," that can be used by cellular membrane trafficking machinery to regulate adhesion. The cadherin-binding protein p120-catenin (p120) appears to be the master regulator of access to these switches, thereby controlling cadherin endocytosis and turnover. This review focuses on p120 and other cadherin-binding proteins, ubiquitin ligases, and growth factors as key modulators of cadherin membrane trafficking.
Collapse
Affiliation(s)
- Chantel M Cadwell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia
| | - Wenji Su
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia.,Biochemistry, Cell, and Developmental Biology Graduate Training Program, Emory University, Atlanta, Georgia
| | - Andrew P Kowalczyk
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia.,Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia.,Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
42
|
Clarke DN, Miller PW, Lowe CJ, Weis WI, Nelson WJ. Characterization of the Cadherin-Catenin Complex of the Sea Anemone Nematostella vectensis and Implications for the Evolution of Metazoan Cell-Cell Adhesion. Mol Biol Evol 2016; 33:2016-29. [PMID: 27189570 PMCID: PMC4948710 DOI: 10.1093/molbev/msw084] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The cadherin-catenin complex (CCC) mediates cell-cell adhesion in bilaterian animals by linking extracellular cadherin-based adhesions to the actin cytoskeleton. However, it is unknown whether the basic organization of the complex is conserved across all metazoans. We tested whether protein interactions and actin-binding properties of the CCC are conserved in a nonbilaterian animal, the sea anemone Nematostella vectensis We demonstrated that N. vectensis has a complete repertoire of cadherin-catenin proteins, including two classical cadherins, one α-catenin, and one β-catenin. Using size-exclusion chromatography and multi-angle light scattering, we showed that α-catenin and β-catenin formed a heterodimer that bound N. vectensis Cadherin-1 and -2. Nematostella vectensis α-catenin bound F-actin with equivalent affinity as either a monomer or an α/β-catenin heterodimer, and its affinity for F-actin was, in part, regulated by a novel insert between the N- and C-terminal domains. Nematostella vectensis α-catenin inhibited Arp2/3 complex-mediated nucleation of actin filaments, a regulatory property previously thought to be unique to mammalian αE-catenin. Thus, despite significant differences in sequence, the key interactions of the CCC are conserved between bilaterians and cnidarians, indicating that the core function of the CCC as a link between cell adhesions and the actin cytoskeleton is ancestral in the eumetazoans.
Collapse
Affiliation(s)
| | - Phillip W Miller
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine
| | | | - William I Weis
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine Department of Structural Biology, Stanford University School of Medicine
| | - William James Nelson
- Department of Biology, Stanford University Department of Molecular and Cellular Physiology, Stanford University School of Medicine
| |
Collapse
|
43
|
Research that is fit to print. Nat Biotechnol 2016; 34:587-8. [PMID: 27281410 DOI: 10.1038/nbt.3577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
44
|
Abstract
The classic cadherin-catenin complex (CCC) mediates cell-cell adhesion in metazoans. Although substantial insights have been gained by studying the CCC in vertebrate tissue culture, analyzing requirements for and regulation of the CCC in vertebrates remains challenging.
Caenorhabditis elegans is a powerful system for connecting the molecular details of CCC function with functional requirements in a living organism. Recent data, using an “angstroms to embryos” approach, have elucidated functions for key residues, conserved across all metazoans, that mediate cadherin/β-catenin binding. Other recent work reveals a novel, potentially ancestral, role for the
C. elegans p120ctn homologue in regulating polarization of blastomeres in the early embryo via Cdc42 and the partitioning-defective (PAR)/atypical protein kinase C (aPKC) complex. Finally, recent work suggests that the CCC is trafficked to the cell surface via the clathrin adaptor protein complex 1 (AP-1) in surprising ways. These studies continue to underscore the value of
C. elegans as a model system for identifying conserved molecular mechanisms involving the CCC.
Collapse
Affiliation(s)
- Jeff Hardin
- Department of Zoology, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
45
|
Phosphoregulation of the C. elegans cadherin-catenin complex. Biochem J 2015; 472:339-52. [PMID: 26443865 DOI: 10.1042/bj20150410] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 10/06/2015] [Indexed: 12/20/2022]
Abstract
Adherens junctions play key roles in mediating cell-cell contacts during tissue development. In Caenorhabditis elegans embryos, the cadherin-catenin complex (CCC), composed of the classical cadherin HMR-1 and members of three catenin families, HMP-1, HMP-2 and JAC-1, is necessary for normal blastomere adhesion, gastrulation, ventral enclosure of the epidermis and embryo elongation. Disruption of CCC assembly or function results in embryonic lethality. Previous work suggests that components of the CCC are subject to phosphorylation. However, the identity of phosphorylated residues in CCC components and their contributions to CCC stability and function in a living organism remain speculative. Using mass spectrometry, we systematically identify phosphorylated residues in the essential CCC subunits HMR-1, HMP-1 and HMP-2 in vivo. We demonstrate that HMR-1/cadherin phosphorylation occurs on three sites within its β-catenin binding domain that each contributes to CCC assembly on lipid bilayers. In contrast, phosphorylation of HMP-2/β-catenin inhibits its association with HMR-1/cadherin in vitro, suggesting a role in CCC disassembly. Although HMP-1/α-catenin is also phosphorylated in vivo, phosphomimetic mutations do not affect its ability to associate with other CCC components or interact with actin in vitro. Collectively, our findings support a model in which distinct phosphorylation events contribute to rapid CCC assembly and disassembly, both of which are essential for morphogenetic rearrangements during development.
Collapse
|
46
|
Choi HJ, Loveless T, Lynch AM, Bang I, Hardin J, Weis WI. A conserved phosphorylation switch controls the interaction between cadherin and β-catenin in vitro and in vivo. Dev Cell 2015; 33:82-93. [PMID: 25850673 DOI: 10.1016/j.devcel.2015.02.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 12/20/2014] [Accepted: 02/05/2015] [Indexed: 10/23/2022]
Abstract
In metazoan adherens junctions, β-catenin links the cytoplasmic tail of classical cadherins to the F-actin-binding protein α-catenin. Phosphorylation of a Ser/Thr-rich region in the cadherin tail dramatically enhances affinity for β-catenin and promotes cell-cell adhesion in cell culture systems, but its importance has not been demonstrated in vivo. Here, we identify a critical phosphorylated serine in the C. elegans cadherin HMR-1 required for strong binding to the β-catenin homolog HMP-2. Ablation of this phosphoserine interaction produces developmental defects that resemble full loss-of-function (Hammerhead and Humpback) phenotypes. Most metazoans possess a single gene for β-catenin, which is also a transcriptional coactivator in Wnt signaling. Nematodes and planaria, however, have a set of paralogous β-catenins; for example, C. elegans HMP-2 functions only in cell-cell adhesion, whereas SYS-1 mediates transcriptional activation through interactions with POP-1/Tcf. Our structural data define critical sequence differences responsible for the unique ligand specificities of these two proteins.
Collapse
Affiliation(s)
- Hee-Jung Choi
- School of Biological Sciences, Seoul National University, Seoul 151-747, South Korea.
| | - Timothy Loveless
- Program in Cellular and Molecular Biology, University of Wisconsin, Madison, WI 53706, USA
| | - Allison M Lynch
- Department of Zoology, University of Wisconsin, Madison, WI 53706, USA
| | - Injin Bang
- School of Biological Sciences, Seoul National University, Seoul 151-747, South Korea
| | - Jeff Hardin
- Program in Cellular and Molecular Biology, University of Wisconsin, Madison, WI 53706, USA; Department of Zoology, University of Wisconsin, Madison, WI 53706, USA
| | - William I Weis
- Departments of Structural Biology and of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
47
|
Escobar DJ, Desai R, Ishiyama N, Folmsbee SS, Novak MN, Flozak AS, Daugherty RL, Mo R, Nanavati D, Sarpal R, Leckband D, Ikura M, Tepass U, Gottardi CJ. α-Catenin phosphorylation promotes intercellular adhesion through a dual-kinase mechanism. J Cell Sci 2015; 128:1150-65. [PMID: 25653389 DOI: 10.1242/jcs.163824] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The cadherin-catenin adhesion complex is a key contributor to epithelial tissue stability and dynamic cell movements during development and tissue renewal. How this complex is regulated to accomplish these functions is not fully understood. We identified several phosphorylation sites in mammalian αE-catenin (also known as catenin α-1) and Drosophila α-Catenin within a flexible linker located between the middle (M)-region and the carboxy-terminal actin-binding domain. We show that this phospho-linker (P-linker) is the main phosphorylated region of α-catenin in cells and is sequentially modified at casein kinase 2 and 1 consensus sites. In Drosophila, the P-linker is required for normal α-catenin function during development and collective cell migration, although no obvious defects were found in cadherin-catenin complex assembly or adherens junction formation. In mammalian cells, non-phosphorylatable forms of α-catenin showed defects in intercellular adhesion using a mechanical dispersion assay. Epithelial sheets expressing phosphomimetic forms of α-catenin showed faster and more coordinated migrations after scratch wounding. These findings suggest that phosphorylation and dephosphorylation of the α-catenin P-linker are required for normal cadherin-catenin complex function in Drosophila and mammalian cells.
Collapse
Affiliation(s)
- David J Escobar
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA The Driskill Graduate Training Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ridhdhi Desai
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5 Canada
| | - Noboru Ishiyama
- University Health Network, Princess Margaret Cancer Center, University of Toronto, Toronto, ON M5T 2M9, Canada
| | - Stephen S Folmsbee
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA The Driskill Graduate Training Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Megan N Novak
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA The Driskill Graduate Training Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Annette S Flozak
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Rebecca L Daugherty
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA The Driskill Graduate Training Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Rigen Mo
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Dhaval Nanavati
- Department of Chemistry of Life Processes, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ritu Sarpal
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5 Canada
| | - Deborah Leckband
- Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana, IL 61801, USA
| | - Mitsu Ikura
- University Health Network, Princess Margaret Cancer Center, University of Toronto, Toronto, ON M5T 2M9, Canada
| | - Ulrich Tepass
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 3G5 Canada
| | - Cara J Gottardi
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA Department of Cellular and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
48
|
Abstract
E-cadherin belongs to the classic cadherin subfamily of calcium-dependent cell adhesion molecules and is crucial for the formation and function of epithelial adherens junctions. In this study, we demonstrate that Vangl2, a vertebrate regulator of planar cell polarity (PCP), controls E-cadherin in epithelial cells. E-cadherin co-immunoprecipitates with Vangl2 from embryonic kidney extracts, and this association is also observed in transfected fibroblasts. Vangl2 enhances the internalization of E-cadherin when overexpressed. Conversely, the quantitative ratio of E-cadherin exposed to the cell surface is increased in cultured renal epithelial cells derived from Vangl2(Lpt/+) mutant mice. Interestingly, Vangl2 is also internalized through protein traffic involving Rab5- and Dynamin-dependent endocytosis. Taken together with recent reports regarding the transport of Frizzled3, MMP14 and nephrin, these results suggest that one of the molecular functions of Vangl2 is to enhance the internalization of specific plasma membrane proteins with broad selectivity. This function may be involved in the control of intercellular PCP signalling or in the PCP-related rearrangement of cell adhesions.
Collapse
|