1
|
Abstract
Evidence implicating Eph receptor tyrosine kinases and their ephrin ligands (that together make up the 'Eph system') in cancer development and progression has been accumulating since the discovery of the first Eph receptor approximately 35 years ago. Advances in the past decade and a half have considerably increased the understanding of Eph receptor-ephrin signalling mechanisms in cancer and have uncovered intriguing new roles in cancer progression and drug resistance. This Review focuses mainly on these more recent developments. I provide an update on the different mechanisms of Eph receptor-ephrin-mediated cell-cell communication and cell autonomous signalling, as well as on the interplay of the Eph system with other signalling systems. I further discuss recent advances in elucidating how the Eph system controls tumour expansion, invasiveness and metastasis, supports cancer stem cells, and drives therapy resistance. In addition to functioning within cancer cells, the Eph system also mediates the reciprocal communication between cancer cells and cells of the tumour microenvironment. The involvement of the Eph system in tumour angiogenesis is well established, but recent findings also demonstrate roles in immune cells, cancer-associated fibroblasts and the extracellular matrix. Lastly, I discuss strategies under evaluation for therapeutic targeting of Eph receptors-ephrins in cancer and conclude with an outlook on promising future research directions.
Collapse
Affiliation(s)
- Elena B Pasquale
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
2
|
Pospíšil J, Hrabovský M, Bohačiaková D, Hovádková Z, Jurásek M, Mlčoušková J, Paruch K, Nevolová Š, Damborsky J, Hampl A, Jaros J. Geometric Control of Cell Behavior by Biomolecule Nanodistribution. ACS Biomater Sci Eng 2022; 8:4789-4806. [PMID: 36202388 PMCID: PMC9667466 DOI: 10.1021/acsbiomaterials.2c00650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Many dynamic interactions within the cell microenvironment
modulate
cell behavior and cell fate. However, the pathways and mechanisms
behind cell–cell or cell–extracellular matrix interactions
remain understudied, as they occur at a nanoscale level. Recent progress
in nanotechnology allows for mimicking of the microenvironment at
nanoscale in vitro; electron-beam lithography (EBL)
is currently the most promising technique. Although this nanopatterning
technique can generate nanostructures of good quality and resolution,
it has resulted, thus far, in the production of only simple shapes
(e.g., rectangles) over a relatively small area (100 × 100 μm),
leaving its potential in biological applications unfulfilled. Here,
we used EBL for cell-interaction studies by coating cell-culture-relevant
material with electron-conductive indium tin oxide, which formed nanopatterns
of complex nanohexagonal structures over a large area (500 ×
500 μm). We confirmed the potential of EBL for use in cell-interaction
studies by analyzing specific cell responses toward differentially
distributed nanohexagons spaced at 1000, 500, and 250 nm. We found
that our optimized technique of EBL with HaloTags enabled the investigation
of broad changes to a cell-culture-relevant surface and can provide
an understanding of cellular signaling mechanisms at a single-molecule
level.
Collapse
Affiliation(s)
- Jakub Pospíšil
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic.,Core Facility Cellular Imaging, CEITEC, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Miloš Hrabovský
- TESCAN Orsay Holding a.s., Libušina tř. 863, Brno 623 00, Czech Republic
| | - Dáša Bohačiaková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic.,International Clinical Research Center (ICRC), St. Anne's University Hospital, Pekařská 53, Brno 656 91, Czech Republic
| | | | | | - Jarmila Mlčoušková
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Kamil Paruch
- International Clinical Research Center (ICRC), St. Anne's University Hospital, Pekařská 53, Brno 656 91, Czech Republic.,Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Šárka Nevolová
- International Clinical Research Center (ICRC), St. Anne's University Hospital, Pekařská 53, Brno 656 91, Czech Republic.,Loschmidt Laboratories, Department of Experimental Biology and Research Centre for Toxic Compounds in the Environment (RECETOX), Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Jiri Damborsky
- International Clinical Research Center (ICRC), St. Anne's University Hospital, Pekařská 53, Brno 656 91, Czech Republic.,Loschmidt Laboratories, Department of Experimental Biology and Research Centre for Toxic Compounds in the Environment (RECETOX), Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Aleš Hampl
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic.,International Clinical Research Center (ICRC), St. Anne's University Hospital, Pekařská 53, Brno 656 91, Czech Republic
| | - Josef Jaros
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic.,International Clinical Research Center (ICRC), St. Anne's University Hospital, Pekařská 53, Brno 656 91, Czech Republic
| |
Collapse
|
3
|
Gomez-Soler M, Gehring MP, Lechtenberg BC, Zapata-Mercado E, Ruelos A, Matsumoto MW, Hristova K, Pasquale EB. Ligands with different dimeric configurations potently activate the EphA2 receptor and reveal its potential for biased signaling. iScience 2022; 25:103870. [PMID: 35243233 PMCID: PMC8858996 DOI: 10.1016/j.isci.2022.103870] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/13/2021] [Accepted: 02/01/2022] [Indexed: 12/03/2022] Open
Abstract
The EphA2 receptor tyrosine kinase activates signaling pathways with different, and sometimes opposite, effects in cancer and other pathologies. Thus, highly specific and potent biased ligands that differentially control EphA2 signaling responses could be therapeutically valuable. Here, we use EphA2-specific monomeric peptides to engineer dimeric ligands with three different geometric configurations to combine a potential ability to differentially modulate EphA2 signaling responses with the high potency and prolonged receptor residence time characteristic of dimeric ligands. The different dimeric peptides readily induce EphA2 clustering, autophosphorylation and signaling, the best with sub-nanomolar potency. Yet, there are differences in two EphA2 signaling responses induced by peptides with different configurations, which exhibit distinct potency and efficacy. The peptides bias signaling when compared with the ephrinA1-Fc ligand and do so via different mechanisms. These findings provide insights into Eph receptor signaling, and proof-of-principle that different Eph signaling responses can be distinctly modulated.
Collapse
Affiliation(s)
- Maricel Gomez-Soler
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Marina P. Gehring
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Bernhard C. Lechtenberg
- Ubiquitin Signalling Division, The Walter and Eliza Hall Institute of Medical Research, Parkville Victoria 3052, Australia and Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Elmer Zapata-Mercado
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Alyssa Ruelos
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Mike W. Matsumoto
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Kalina Hristova
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Elena B. Pasquale
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| |
Collapse
|
4
|
A biophysical perspective of the regulatory mechanisms of ezrin/radixin/moesin proteins. Biophys Rev 2022; 14:199-208. [PMID: 35340609 PMCID: PMC8921360 DOI: 10.1007/s12551-021-00928-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/14/2021] [Indexed: 02/08/2023] Open
Abstract
Many signal transductions resulting from ligand-receptor interactions occur at the cell surface. These signaling pathways play essential roles in cell polarization, membrane morphogenesis, and the modulation of membrane tension at the cell surface. However, due to the large number of membrane-binding proteins, including actin-membrane linkers, and transmembrane proteins present at the cell surface, the molecular mechanisms underlying the regulation at the cell surface are yet unclear. Here, we describe the molecular functions of one of the key players at the cell surface, ezrin/radixin/moesin (ERM) proteins from a biophysical point of view. We focus our discussion on biophysical properties of ERM proteins revealed by using biophysical tools in live cells and in vitro reconstitution systems. We first describe the structural properties of ERM proteins and then discuss the interactions of ERM proteins with PI(4,5)P2 and the actin cytoskeleton. These properties of ERM proteins revealed by using biophysical approaches have led to a better understanding of their physiological functions in cells and tissues. Supplementary Information The online version contains supplementary material available at 10.1007/s12551-021-00928-0.
Collapse
|
5
|
Penchev VR, Chang YT, Begum A, Ewachiw T, Gocke C, Li J, McMillan RH, Wang Q, Anders R, Marchionni L, Maitra A, Uren A, Rasheed Z, Matsui W. Ezrin Promotes Stem Cell Properties in Pancreatic Ductal Adenocarcinoma. Mol Cancer Res 2019; 17:929-936. [PMID: 30655325 DOI: 10.1158/1541-7786.mcr-18-0367] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 10/09/2018] [Accepted: 01/08/2019] [Indexed: 01/25/2023]
Abstract
Self-renewal maintains the long-term clonogenic growth that is required for cancer relapse and progression, but the cellular processes regulating this property are not fully understood. In many diseases, self-renewal is enhanced in cancer stem cells (CSC), and in pancreatic ductal adenocarcinoma (PDAC), CSCs are characterized by the surface expression of CD44. In addition to cell adhesion, CD44 impacts cell shape and morphology by modulating the actin cytoskeleton via Ezrin, a member of the Ezrin/Radixin/Moesin (ERM) family of linker proteins. We examined the expression of Ezrin in PDAC cells and found higher levels of both total and activated Ezrin in CSCs compared with bulk tumor cells. We also found that the knockdown of Ezrin in PDAC cells decreased clonogenic growth, self-renewal, cell migration, and CSC frequency in vitro as well as tumor initiation in vivo. These effects were associated with cytoskeletal changes that are similar to those occurring during the differentiation of normal stem cells, and the inhibition of actin remodeling reversed the impact of Ezrin loss. Finally, targeting Ezrin using a small-molecule inhibitor limited the self-renewal of clinically derived low-passage PDAC xenografts. Our findings demonstrate that Ezrin modulates CSCs properties and may represent a novel target for the treatment of PDAC. IMPLICATIONS: Our findings demonstrate that Ezrin modulates CSCs' properties and may represent a novel target for the treatment of PDAC.
Collapse
Affiliation(s)
- Vesselin R Penchev
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yu-Tai Chang
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Asma Begum
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Theodore Ewachiw
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christian Gocke
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joey Li
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ross H McMillan
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Qiuju Wang
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert Anders
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Luigi Marchionni
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anirban Maitra
- Department of Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Aykut Uren
- Department of Oncology, Lombardy Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C
| | - Zeshaan Rasheed
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - William Matsui
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
6
|
Hadavi E, Leijten J, Brinkmann J, Jonkheijm P, Karperien M, van Apeldoorn A. Fibronectin and Collagen IV Microcontact Printing Improves Insulin Secretion by INS1E Cells. Tissue Eng Part C Methods 2018; 24:628-636. [PMID: 30306836 DOI: 10.1089/ten.tec.2018.0151] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
IMPACT STATEMENT This research deals with finding a proper bioengineering strategy for the creation of improved β-cell replacement therapy in type 1 diabetes. It specifically deals with the microenvironment of β-cells and its relationship to their endocrine function.
Collapse
Affiliation(s)
- Elahe Hadavi
- 1 Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente , Enschede, The Netherlands
| | - Jeroen Leijten
- 1 Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente , Enschede, The Netherlands
| | - Jenny Brinkmann
- 2 MESA+ Institute for Nanotechnology, Molecular Nanofabrication Group, University of Twente , Enschede, The Netherlands
| | - Pascal Jonkheijm
- 2 MESA+ Institute for Nanotechnology, Molecular Nanofabrication Group, University of Twente , Enschede, The Netherlands
| | - Marcel Karperien
- 1 Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente , Enschede, The Netherlands
| | - Aart van Apeldoorn
- 1 Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente , Enschede, The Netherlands .,3 Complex Tissue Regeneration Department, MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University , Maastricht, The Netherlands
| |
Collapse
|
7
|
Meng Y, Eirin A, Zhu XY, Tang H, Chanana P, Lerman A, Van Wijnen AJ, Lerman LO. The metabolic syndrome alters the miRNA signature of porcine adipose tissue-derived mesenchymal stem cells. Cytometry A 2017; 93:93-103. [PMID: 28678424 DOI: 10.1002/cyto.a.23165] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 05/24/2017] [Accepted: 06/15/2017] [Indexed: 01/08/2023]
Abstract
Autologous transplantation of mesenchymal stem cells (MSCs) is a viable option for the treatment of several diseases. Evidence indicates that MSCs release extracellular vesicles (EVs) and that EVs shuttle miRNAs to damaged parenchymal cells to activate an endogenous repair program. We hypothesize that comorbidities may interfere with the packaging of cargo in MSC-derived EVs. Therefore, we examined whether metabolic syndrome (MetS) modulates the miRNA content packed within MSC-derived EVs. MSCs were collected from swine abdominal adipose tissue after 16 weeks of lean or obese diet (n = 7 each). Next-generation RNA sequencing of miRNAs (miRNA-seq) was performed to identify miRNAs enriched in MSC-derived EVs and their predicted target genes. Functional pathway analysis of the top 50 target genes of the top 4 miRNAs enriched in each group was performed using gene ontology analysis. Lean- and MetS-EVs were enriched in, respectively, 14 and 8 distinct miRNAs. Target genes of miRNAs enriched in MetS-EVs were implicated in the development of MetS and its complications, including diabetes-related pathways, validated transcriptional targets of AP1 family members Fra1 and Fra2, Class A/1 (Rhodopsin-like receptors), and Peptide ligand-binding receptors. In contrast, miRNAs enriched in Lean EVs target primarily EphrinA-EPHA and the Rho family of GTPases. MetS alters the miRNA content of EVs derived from porcine adipose tissue MSCs. These alterations could impair the efficacy and limit the therapeutic use of autologous MSCs in subjects with MetS. Our findings may assist in developing adequate regenerative strategies to preserve the reparative potency of MSCs in individuals with MetS. © 2017 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Yu Meng
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota.,Department of Nephrology, the First Hospital Affiliated to Jinan University, Guangzhou, 510630, China
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Pritha Chanana
- Division of Health Sciences Research & Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Amir Lerman
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | | | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota.,Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
8
|
Bulanova DR, Akimov YA, Rokka A, Laajala TD, Aittokallio T, Kouvonen P, Pellinen T, Kuznetsov SG. Orphan G protein-coupled receptor GPRC5A modulates integrin β1-mediated epithelial cell adhesion. Cell Adh Migr 2017; 11:434-446. [PMID: 27715394 PMCID: PMC5810789 DOI: 10.1080/19336918.2016.1245264] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
G-Protein Coupled Receptor (GPCR), Class C, Group 5, Member A (GPRC5A) has been implicated in several malignancies. The underlying mechanisms, however, remain poorly understood. Using a panel of human cell lines, we demonstrate that CRISPR/Cas9-mediated knockout and RNAi-mediated depletion of GPRC5A impairs cell adhesion to integrin substrates: collagens I and IV, fibronectin, as well as to extracellular matrix proteins derived from the Engelbreth-Holm-Swarm (EHS) mouse sarcoma (Matrigel). Consistent with the phenotype, knock-out of GPRC5A correlated with a reduced integrin β1 (ITGB1) protein expression, impaired phosphorylation of the focal adhesion kinase (FAK), and lower activity of small GTPases RhoA and Rac1. Furthermore, we provide the first evidence for a direct interaction between GPRC5A and a receptor tyrosine kinase EphA2, an upstream regulator of FAK, although its contribution to the observed adhesion phenotype is unclear. Our findings reveal an unprecedented role for GPRC5A in regulation of the ITGB1-mediated cell adhesion and it's downstream signaling, thus indicating a potential novel role for GPRC5A in human epithelial cancers.
Collapse
Affiliation(s)
- Daria R Bulanova
- a Institute for Molecular Medicine Finland (FIMM), University of Helsinki , Helsinki , Finland
| | - Yevhen A Akimov
- a Institute for Molecular Medicine Finland (FIMM), University of Helsinki , Helsinki , Finland
| | - Anne Rokka
- c Turku Centre for Biotechnology , University of Turku and Abo Academy , Turku , Finland
| | - Teemu D Laajala
- a Institute for Molecular Medicine Finland (FIMM), University of Helsinki , Helsinki , Finland.,b Department of Mathematics and Statistics , University of Turku , Turku , Finland
| | - Tero Aittokallio
- a Institute for Molecular Medicine Finland (FIMM), University of Helsinki , Helsinki , Finland.,b Department of Mathematics and Statistics , University of Turku , Turku , Finland
| | - Petri Kouvonen
- c Turku Centre for Biotechnology , University of Turku and Abo Academy , Turku , Finland
| | - Teijo Pellinen
- a Institute for Molecular Medicine Finland (FIMM), University of Helsinki , Helsinki , Finland
| | - Sergey G Kuznetsov
- a Institute for Molecular Medicine Finland (FIMM), University of Helsinki , Helsinki , Finland
| |
Collapse
|
9
|
Singh DR, Ahmed F, Paul MD, Gedam M, Pasquale EB, Hristova K. The SAM domain inhibits EphA2 interactions in the plasma membrane. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:31-38. [PMID: 27776928 DOI: 10.1016/j.bbamcr.2016.10.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 09/26/2016] [Accepted: 10/18/2016] [Indexed: 11/18/2022]
Abstract
All members of the Eph receptor family of tyrosine kinases contain a SAM domain near the C terminus, which has been proposed to play a role in receptor homotypic interactions and/or interactions with binding partners. The SAM domain of EphA2 is known to be important for receptor function, but its contribution to EphA2 lateral interactions in the plasma membrane has not been determined. Here we use a FRET-based approach to directly measure the effect of the SAM domain on the stability of EphA2 dimers on the cell surface in the absence of ligand binding. We also investigate the functional consequences of EphA2 SAM domain deletion. Surprisingly, we find that the EphA2 SAM domain inhibits receptor dimerization and decreases EphA2 tyrosine phosphorylation. This role is dramatically different from the role of the SAM domain of the related EphA3 receptor, which we previously found to stabilize EphA3 dimers and increase EphA3 tyrosine phosphorylation in cells in the absence of ligand. Thus, the EphA2 SAM domain likely contributes to a unique mode of EphA2 interaction that leads to distinct signaling outputs.
Collapse
Affiliation(s)
- Deo R Singh
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 Charles Street, Baltimore, MD 21218, United States
| | - Fozia Ahmed
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 Charles Street, Baltimore, MD 21218, United States
| | - Michael D Paul
- Program in Molecular Biophysics, Johns Hopkins University, 3400 Charles street, Baltimore, MD 21218, United States
| | - Manasee Gedam
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 Charles Street, Baltimore, MD 21218, United States
| | - Elena B Pasquale
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Road, La Jolla, San Diego, CA 92037, United States
| | - Kalina Hristova
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 Charles Street, Baltimore, MD 21218, United States; Program in Molecular Biophysics, Johns Hopkins University, 3400 Charles street, Baltimore, MD 21218, United States.
| |
Collapse
|
10
|
Luján P, Varsano G, Rubio T, Hennrich ML, Sachsenheimer T, Gálvez-Santisteban M, Martín-Belmonte F, Gavin AC, Brügger B, Köhn M. PRL-3 disrupts epithelial architecture by altering the post-mitotic midbody position. J Cell Sci 2016; 129:4130-4142. [PMID: 27656108 PMCID: PMC5117205 DOI: 10.1242/jcs.190215] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 09/16/2016] [Indexed: 12/20/2022] Open
Abstract
Disruption of epithelial architecture is a fundamental event during epithelial tumorigenesis. We show that the expression of the cancer-promoting phosphatase PRL-3 (PTP4A3), which is overexpressed in several epithelial cancers, in polarized epithelial MDCK and Caco2 cells leads to invasion and the formation of multiple ectopic, fully polarized lumens in cysts. Both processes disrupt epithelial architecture and are hallmarks of cancer. The pathological relevance of these findings is supported by the knockdown of endogenous PRL-3 in MCF-7 breast cancer cells grown in three-dimensional branched structures, showing the rescue from multiple-lumen- to single-lumen-containing branch ends. Mechanistically, it has been previously shown that ectopic lumens can arise from midbodies that have been mislocalized through the loss of mitotic spindle orientation or through the loss of asymmetric abscission. Here, we show that PRL-3 triggers ectopic lumen formation through midbody mispositioning without altering the spindle orientation or asymmetric abscission, instead, PRL-3 accelerates cytokinesis, suggesting that this process is an alternative new mechanism for ectopic lumen formation in MDCK cysts. The disruption of epithelial architecture by PRL-3 revealed here is a newly recognized mechanism for PRL-3-promoted cancer progression.
Collapse
Affiliation(s)
- Pablo Luján
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg 69117, Germany
| | - Giulia Varsano
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg 69117, Germany
| | - Teresa Rubio
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg 69117, Germany
| | - Marco L Hennrich
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Heidelberg 69117, Germany
| | - Timo Sachsenheimer
- Heidelberg University Biochemistry Center, University of Heidelberg, Heidelberg 69120, Germany
| | - Manuel Gálvez-Santisteban
- Department of Development and Differentiation, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28049, Spain
| | - Fernando Martín-Belmonte
- Department of Development and Differentiation, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28049, Spain
| | - Anne-Claude Gavin
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Heidelberg 69117, Germany
| | - Britta Brügger
- Heidelberg University Biochemistry Center, University of Heidelberg, Heidelberg 69120, Germany
| | - Maja Köhn
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg 69117, Germany
| |
Collapse
|
11
|
Johnson C, Segovia B, Kandpal RP. EPHA7 and EPHA10 Physically Interact and Differentially Co-localize in Normal Breast and Breast Carcinoma Cell Lines, and the Co-localization Pattern Is Altered in EPHB6-expressing MDA-MB-231 Cells. Cancer Genomics Proteomics 2016; 13:359-368. [PMID: 27566654 PMCID: PMC5070625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 06/27/2016] [Indexed: 06/06/2023] Open
Abstract
Erythropoietin-producing hepatocellular carcinoma cell (EPH) receptors comprise the most abundant receptor tyrosine kinase family characterized to date in mammals including humans. These proteins are involved in axon guidance, tissue organization, vascular development and the intricate process of various diseases including cancer. These diverse functions of EPH receptors are attributed, in part, to their abilities for heterodimerization. While the interacting partners of kinase-deficient EPHB6 receptor have been characterized, the interaction of the kinase-dead EPHA10 with any other receptor has not been identified. By using co-immunoprecipitation, we demonstrated physical interaction between kinase-deficient EPHA10 with kinase-sufficient EPHA7 receptor. Immunocytochemical analyses have revealed that these two receptors co-localize on the cell surface, and soluble portions of the receptors exist as a complex in the cytoplasm as well as the nuclei. While EPHA7 and EPHA10 co-localize similarly on the membrane in MCF10A and MCF7 cells, they were differentially co-localized in MDA-MB-231 cells stably transfected with empty pcDNA vector (MDA-MB-231-PC) or an expression construct of EPHB6 (MDA-MB-231-B6). The full-length isoforms of these receptors were co-localized on the cell surface, and the soluble forms were present as a complex in the cytoplasm as well as the nucleus in MDA-MB-231-PC cells. MDA-MB-231-B6 cells, on the other hand, were distinguished by the absence of any signal in the nuclei. Our results represent the first demonstration of physical interaction between EPHA10 and EPHA7 and their cellular co-localization. Furthermore, these observations also suggest gene-regulatory functions of the complex of the soluble forms of these receptors in breast carcinoma cells of differential invasiveness.
Collapse
Affiliation(s)
- Candace Johnson
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, U.S.A
| | - Briana Segovia
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, U.S.A
| | - Raj P Kandpal
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, U.S.A.
| |
Collapse
|
12
|
Singh DR, Pasquale EB, Hristova K. A small peptide promotes EphA2 kinase-dependent signaling by stabilizing EphA2 dimers. Biochim Biophys Acta Gen Subj 2016; 1860:1922-8. [PMID: 27281300 DOI: 10.1016/j.bbagen.2016.06.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 05/31/2016] [Accepted: 06/03/2016] [Indexed: 12/01/2022]
Abstract
BACKGROUND The EphA2 receptor tyrosine kinase is known to promote cancer cell malignancy in the absence of activation by ephrin ligands. This behavior depends on high EphA2 phosphorylation on Ser897 and low tyrosine phosphorylation, resulting in increased cell migration and invasiveness. We have previously shown that EphA2 forms dimers in the absence of ephrin ligand binding, and that dimerization of unliganded EphA2 can decrease EphA2 Ser897 phosphorylation. We have also identified a small peptide called YSA, which binds EphA2 and competes with the naturally occurring ephrin ligands. METHODS Here, we investigate the effect of YSA on EphA2 dimer stability and EphA2 function using quantitative FRET techniques, Western blotting, and cell motility assays. RESULTS We find that the YSA peptide stabilizes the EphA2 dimer, increases EphA2 Tyr phosphorylation, and decreases both Ser897 phosphorylation and cell migration. CONCLUSIONS The experiments demonstrate that the small peptide ligand YSA reduces EphA2 Ser897 pro-tumorigenic signaling by stabilizing the EphA2 dimer. GENERAL SIGNIFICANCE This work is a proof-of-principle demonstration that EphA2 homointeractions in the plasma membrane can be pharmacologically modulated to decrease the pro-tumorigenic signaling of the receptor.
Collapse
Affiliation(s)
- Deo R Singh
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 Charles Street, Baltimore, MD 21218, United States
| | - Elena B Pasquale
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Road, La Jolla, San Diego, CA 92037, United States
| | - Kalina Hristova
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 Charles Street, Baltimore, MD 21218, United States.
| |
Collapse
|
13
|
Eph/ephrin signaling in the kidney and lower urinary tract. Pediatr Nephrol 2016; 31:359-71. [PMID: 25903642 DOI: 10.1007/s00467-015-3112-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 03/30/2015] [Accepted: 03/31/2015] [Indexed: 02/06/2023]
Abstract
Development and homeostasis of the highly specialized cell types and tissues that constitute the organs of the urinary system, the kidneys and ureters, the bladder, and the urethra, require the tightly regulated exchange of signals in and between these tissues. Eph/ephrin signaling is a bidirectional signaling pathway that has been functionally implicated in many developmental and homeostatic contexts, most prominently in the vascular and neural system. Expression and knockout analyses have now provided evidence that Eph/ephrin signaling is of crucial relevance for cell and tissue interactions in the urinary system as well. A clear requirement has emerged in the formation of the vesicoureteric junction, in urorectal septation and glomerulogenesis during embryonic development, in maintenance of medullary tubular cells and podocytes in homeostasis, and in podocyte and glomerular injury responses. Deregulation of Eph/ephrin signaling may also contribute to the formation and progression of tumors in the urinary system, most prominently bladder and renal cell carcinoma. While in the embryonic contexts Eph/ephrin signaling regulates adhesion of epithelial cells, in the adult setting, cell-shape changes and cell survival seem to be the primary cellular processes mediated by this signaling module. With progression of the genetic analyses of mice conditionally mutant for compound alleles of Eph receptor and ephrin ligand genes, additional essential functions are likely to arise in the urinary system.
Collapse
|
14
|
Dave A, Martin S, Kumar R, Craig JE, Burdon KP, Sharma S. EPHA2 MUTATIONS CONTRIBUTE TO CONGENITAL CATARACT THROUGH DIVERSE MECHANISMS. Mol Vis 2016; 22:18-30. [PMID: 26900323 PMCID: PMC4734147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 01/12/2016] [Indexed: 10/29/2022] Open
Abstract
PURPOSE Congenital cataract is a leading cause of childhood blindness. Mutations in the EPHA2 gene are one of the causes of inherited congenital cataract. The EPHA2 gene encodes a membrane-bound tyrosine kinase receptor and is highly expressed in epithelial cells, including in the ocular lens. Signaling through the EPHA2 receptor plays a pivotal role in epithelial cell homeostasis. The aim of this study was to determine the effect of congenital cataract causing mutations in the EPHA2 gene on the encoded protein in epithelial cells. METHODS The effect of five disease-causing mutations, p.P584L (c.1751C>T), p.T940I (c.2819C>T), p.D942fsXC71 (c.2826-9G>A), p.A959T (c.2875G>A), and p.V972GfsX39 (c.2915_2916delTG), on localization of the protein was examined in two in vitro epithelial cell culture systems: Madin-Darby Canine Kidney (MDCK) and human colorectal adenocarcinoma (Caco-2) epithelial cells. Myc-tagged mutant constructs were generated by polymerase chain reaction (PCR)-based mutagenesis. The Myc-tagged wild-type construct was used as a control. The Myc-tagged wild-type and mutant proteins were ectopically expressed and detected by immunofluorescence labeling. RESULTS Two of the mutations, p.T940I and p.D942fsXC71, located within the cytoplasmic sterile-α-motif (SAM) domain of EPHA2, led to mis-localization of the protein to the perinuclear space and co-localization with the cis-golgi apparatus, indicating sub-organellar/cellular retention of the mutant proteins. The mutant proteins carrying the remaining three mutations, similar to the wild-type EPHA2, localized to the cell membrane. CONCLUSIONS Mis-localization of two of the mutant proteins in epithelial cells suggests that some disease-causing mutations in EPHA2 likely affect lens epithelial cell homeostasis and contribute to cataract. This study suggests that mutations in EPHA2 contribute to congenital cataract through diverse mechanisms.
Collapse
Affiliation(s)
- Alpana Dave
- Department of Ophthalmology, Flinders University, Bedford Park, South Australia, Australia
| | - Sarah Martin
- Department of Ophthalmology, Flinders University, Bedford Park, South Australia, Australia
| | - Raman Kumar
- Robinson Research Institute, The University of Adelaide, Australia,School of Medicine, The University of Adelaide, Australia
| | - Jamie E. Craig
- Department of Ophthalmology, Flinders University, Bedford Park, South Australia, Australia
| | - Kathryn P. Burdon
- Department of Ophthalmology, Flinders University, Bedford Park, South Australia, Australia
| | - Shiwani Sharma
- Department of Ophthalmology, Flinders University, Bedford Park, South Australia, Australia
| |
Collapse
|
15
|
Singh DR, Ahmed F, King C, Gupta N, Salotto M, Pasquale EB, Hristova K. EphA2 Receptor Unliganded Dimers Suppress EphA2 Pro-tumorigenic Signaling. J Biol Chem 2015; 290:27271-27279. [PMID: 26363067 DOI: 10.1074/jbc.m115.676866] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Indexed: 01/08/2023] Open
Abstract
The EphA2 receptor tyrosine kinase promotes cell migration and cancer malignancy through a ligand- and kinase-independent distinctive mechanism that has been linked to high Ser-897 phosphorylation and low tyrosine phosphorylation. Here, we demonstrate that EphA2 forms dimers in the plasma membrane of HEK293T cells in the absence of ephrin ligand binding, suggesting that the current seeding mechanism model of EphA2 activation is incomplete. We also characterize a dimerization-deficient EphA2 mutant that shows enhanced ability to promote cell migration, concomitant with increased Ser-897 phosphorylation and decreased tyrosine phosphorylation compared with EphA2 wild type. Our data reveal a correlation between unliganded dimerization and tumorigenic signaling and suggest that EphA2 pro-tumorigenic activity is mediated by the EphA2 monomer. Thus, a therapeutic strategy that aims at the stabilization of EphA2 dimers may be beneficial for the treatment of cancers linked to EphA2 overexpression.
Collapse
Affiliation(s)
- Deo R Singh
- Department of Materials Science and Engineering, The Johns Hopkins University, Baltimore, Maryland 21218
| | - Fozia Ahmed
- Department of Materials Science and Engineering, The Johns Hopkins University, Baltimore, Maryland 21218
| | - Christopher King
- Department of Program in Molecular Biophysics, The Johns Hopkins University, Baltimore, Maryland 21218 and
| | - Nisha Gupta
- Department of Materials Science and Engineering, The Johns Hopkins University, Baltimore, Maryland 21218
| | - Matt Salotto
- Department of Materials Science and Engineering, The Johns Hopkins University, Baltimore, Maryland 21218
| | - Elena B Pasquale
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, San Diego, California 92037
| | - Kalina Hristova
- Department of Materials Science and Engineering, The Johns Hopkins University, Baltimore, Maryland 21218; Department of Program in Molecular Biophysics, The Johns Hopkins University, Baltimore, Maryland 21218 and.
| |
Collapse
|
16
|
Harada K, Negishi M, Katoh H. HGF-induced serine 897 phosphorylation of EphA2 regulates epithelial morphogenesis of MDCK cells in 3D culture. J Cell Sci 2015; 128:1912-21. [PMID: 25908849 DOI: 10.1242/jcs.163790] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 03/20/2015] [Indexed: 02/02/2023] Open
Abstract
Expression of EphA2 is upregulated in various cancers that are derived from epithelial cells and correlates with the ability of a cancer cell to undergo migration and invasion. Here we have investigated the role of EphA2 in the epithelial morphogenesis of Madin-Darby canine kidney (MDCK) cells in three-dimensional culture. We show that EphA2 is phosphorylated on serine residue 897 through hepatocyte growth factor (HGF) stimulation using a phosphatidylinositol 3-kinase (PI3K)-Akt-dependent mechanism and that this phosphorylation is required for the formation of extensions, the first step of tubulogenesis, in MDCK cysts. By contrast, stimulation using the ligand ephrinA1 dephosphorylates EphA2 on serine residue 897 and suppresses the HGF-induced morphological change. Furthermore, activation of the small GTPase RhoG is involved in the HGF-induced formation of extensions downstream of EphA2. These observations suggest that a ligand-independent activity of EphA2 contributes to epithelial morphogenesis.
Collapse
Affiliation(s)
- Kohei Harada
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Manabu Negishi
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hironori Katoh
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
17
|
Xiao Y, Sun M, Zhan Z, Ye Y, Huang M, Zou Y, Liang L, Yang X, Xu H. Increased phosphorylation of ezrin is associated with the migration and invasion of fibroblast-like synoviocytes from patients with rheumatoid arthritis. Rheumatology (Oxford) 2014; 53:1291-300. [PMID: 24599913 DOI: 10.1093/rheumatology/keu013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE Increasing evidence indicates that the cytoskeletal protein ezrin may play a critical role in cell motility. This study aims to investigate the role of ezrin in regulating the migration and invasion of fibroblast-like synoviocytes (FLSs) from patients with RA. METHODS Synovial tissues were obtained from 12 patients with RA and 6 with OA, and then FLSs were separated from synovial tissues. The expression of ezrin and phosphorylated ezrin (p-ezrin) was examined by Western blotting or IF staining. A specific inhibitor of ezrin phosphorylation and small interference RNA-mediated ezrin knockdown were used to inhibit the phosphorylation of ezrin. Migration and invasion of FLSs in vitro were measured by the Boyden chamber assay. RESULTS Increased expression of p-ezrin protein was found in synovial tissue and FLSs in patients with RA compared with patients with OA. Stimulation with TNF-α and IL-1β increased ezrin phosphorylation in RA FLSs. Inhibition of p-ezrin protein by a specific inhibitor of phosphorylation of ezrin and small interfering RNA-mediated knockdown reduced in vitro migration and invasion, as well as actin stress fibre formation in RA FLS. Furthermore, rho kinase and p38 mitogen-activated protein kinase (MAPK) signal pathways were involved in the phosphorylation of ezrin and invasion of RA FLSs. CONCLUSION Increased expression of p-ezrin may contribute to aberrant aggressive behaviours of RA FLSs, which are mediated by rho kinase and the p38 MAPK pathway. This suggests a novel strategy targeting phosphorylation of ezrin to prevent synovial invasiveness and joint destruction in RA.
Collapse
Affiliation(s)
- Youjun Xiao
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Mengying Sun
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhongping Zhan
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yujin Ye
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Mingcheng Huang
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yaoyao Zou
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liuqin Liang
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiuyan Yang
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hanshi Xu
- Department of Rheumatology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
18
|
Lázaro-Diéguez F, Cohen D, Fernandez D, Hodgson L, van Ijzendoorn SCD, Müsch A. Par1b links lumen polarity with LGN-NuMA positioning for distinct epithelial cell division phenotypes. ACTA ACUST UNITED AC 2013; 203:251-64. [PMID: 24165937 PMCID: PMC3812971 DOI: 10.1083/jcb.201303013] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Columnar epithelia establish their luminal domains and their mitotic spindles parallel to the basal surface and undergo symmetric cell divisions in which the cleavage furrow bisects the apical domain. Hepatocyte lumina interrupt the lateral domain of neighboring cells perpendicular to two basal domains and their cleavage furrow rarely bifurcates the luminal domains. We determine that the serine/threonine kinase Par1b defines lumen position in concert with the position of the astral microtubule anchoring complex LGN-NuMA to yield the distinct epithelial division phenotypes. Par1b signaling via the extracellular matrix (ECM) in polarizing cells determined RhoA/Rho-kinase activity at cell-cell contact sites. Columnar MDCK and Par1b-depleted hepatocytic HepG2 cells featured high RhoA activity that correlated with robust LGN-NuMA recruitment to the metaphase cortex, spindle alignment with the substratum, and columnar organization. Reduced RhoA activity at the metaphase cortex in HepG2 cells and Par1b-overexpressing MDCK cells correlated with a single or no LGN-NuMA crescent, tilted spindles, and the development of lateral lumen polarity.
Collapse
Affiliation(s)
- Francisco Lázaro-Diéguez
- Department of Developmental and Molecular Biology and 2 Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461
| | | | | | | | | | | |
Collapse
|
19
|
Lisabeth EM, Falivelli G, Pasquale EB. Eph receptor signaling and ephrins. Cold Spring Harb Perspect Biol 2013; 5:5/9/a009159. [PMID: 24003208 DOI: 10.1101/cshperspect.a009159] [Citation(s) in RCA: 301] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The Eph receptors are the largest of the RTK families. Like other RTKs, they transduce signals from the cell exterior to the interior through ligand-induced activation of their kinase domain. However, the Eph receptors also have distinctive features. Instead of binding soluble ligands, they generally mediate contact-dependent cell-cell communication by interacting with surface-associated ligands-the ephrins-on neighboring cells. Eph receptor-ephrin complexes emanate bidirectional signals that affect both receptor- and ephrin-expressing cells. Intriguingly, ephrins can also attenuate signaling by Eph receptors coexpressed in the same cell. Additionally, Eph receptors can modulate cell behavior independently of ephrin binding and kinase activity. The Eph/ephrin system regulates many developmental processes and adult tissue homeostasis. Its abnormal function has been implicated in various diseases, including cancer. Thus, Eph receptors represent promising therapeutic targets. However, more research is needed to better understand the many aspects of their complex biology that remain mysterious.
Collapse
Affiliation(s)
- Erika M Lisabeth
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA
| | | | | |
Collapse
|
20
|
Park JE, Son AI, Zhou R. Roles of EphA2 in Development and Disease. Genes (Basel) 2013; 4:334-57. [PMID: 24705208 PMCID: PMC3924825 DOI: 10.3390/genes4030334] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 05/22/2013] [Accepted: 05/23/2013] [Indexed: 01/12/2023] Open
Abstract
The Eph family of receptor tyrosine kinases (RTKs) has been implicated in the regulation of many aspects of mammalian development. Recent analyses have revealed that the EphA2 receptor is a key modulator for a wide variety of cellular functions. This review focuses on the roles of EphA2 in both development and disease.
Collapse
Affiliation(s)
- Jeong Eun Park
- Susan Lehman-Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA.
| | - Alexander I Son
- Susan Lehman-Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA.
| | - Renping Zhou
- Susan Lehman-Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
21
|
Wang S, Noberini R, Stebbins JL, Das S, Zhang Z, Wu B, Mitra S, Billet S, Fernandez A, Bhowmick NA, Kitada S, Pasquale EB, Fisher PB, Pellecchia M. Targeted delivery of paclitaxel to EphA2-expressing cancer cells. Clin Cancer Res 2012; 19:128-37. [PMID: 23155185 DOI: 10.1158/1078-0432.ccr-12-2654] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE YSA is an EphA2-targeting peptide that effectively delivers anticancer agents to prostate cancer tumors. Here, we report on how we increased the drug-like properties of this delivery system. EXPERIMENTAL DESIGN By introducing non-natural amino acids, we have designed two new EphA2 targeting peptides: YNH, where norleucine and homoserine replace the two methionine residues of YSA, and dYNH, where a D-tyrosine replaces the L-tyrosine at the first position of the YNH peptide. We describe the details of the synthesis of YNH and dYNH paclitaxel conjugates (YNH-PTX and dYNH-PTX) and their characterization in cells and in vivo. RESULTS dYNH-PTX showed improved stability in mouse serum and significantly reduced tumor size in a prostate cancer xenograft model and also reduced tumor vasculature in a syngeneic orthotopic allograft mouse model of renal cancer compared with vehicle or paclitaxel treatments. CONCLUSION This study reveals that targeting EphA2 with dYNH drug conjugates could represent an effective way to deliver anticancer agents to a variety of tumor types.
Collapse
Affiliation(s)
- Si Wang
- Cancer Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 90237, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
McRobert EA, Young AN, Bach LA. Advanced glycation end-products induce calpain-mediated degradation of ezrin. FEBS J 2012; 279:3240-50. [PMID: 22805611 DOI: 10.1111/j.1742-4658.2012.08710.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Advanced glycation end-products (AGEs) are important mediators of diabetic complications via incompletely understood pathways. AGEs bind to intracellular ERM proteins (ezrin, radixin and moesin) that modulate cell shape, motility, adhesion and signal transduction. AGEs bind to the N-terminal domain of ezrin but not full-length ezrin. The AGE binding site may be made accessible either by proteolysis releasing an N-terminal fragment or ezrin activation by phosphorylation. Increased intracellular calcium is a primary event in cell activation by high glucose or AGEs. Calpain activity is increased concomitantly, and ezrin is a calpain substrate. The present study assessed whether glycated proteins affect ezrin cleavage and activation in renal tubule epithelial cells. After 7 days, AGE-BSA decreased ezrin levels in MDCK renal tubular cells to 66 ± 4% of control. AGE-RNAse, ribosylated fetal bovine serum and methylglyoxal-BSA all had similar effects. The AGE-BSA-induced decrease in ezrin was abolished by calpastatin peptide, a specific calpain inhibitor, and 1,2-bis-aminophenoxyethane-tetraacetic acid acetoxymethyl ester (BAPTA-AM), a calcium chelator. Ezrin breakdown products were increased in AGE-BSA-treated cells, with a main fragment of ∼ 43 kDa. In vitro, calpain 1 cleaved recombinant human ezrin, generating breakdown fragments including an N-terminal fragment of ∼ 43 kDa. Studies with ezrin mutants showed that non-phosphorylated ezrin was more susceptible to calpain cleavage. AGE-BSA decreased phosphorylated ERM levels to 31 ± 12% in MDCK cells. Thus, AGE-BSA promotes calpain-mediated proteolysis of ezrin in MDCK cells by both increasing calpain activity and reducing phosphorylation. Therapies targeting both glycated proteins and calpain may provide protection against diabetic complications.
Collapse
|
23
|
Wang S, Placzek WJ, Stebbins JL, Mitra S, Noberini R, Koolpe M, Zhang Z, Dahl R, Pasquale EB, Pellecchia M. Novel targeted system to deliver chemotherapeutic drugs to EphA2-expressing cancer cells. J Med Chem 2012; 55:2427-36. [PMID: 22329578 DOI: 10.1021/jm201743s] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The efficacy of anticancer drugs is often limited by their systemic toxicities and adverse side effects. We report that the EphA2 receptor is overexpressed preferentially in several human cancer cell lines compared to normal tissues and that an EphA2 targeting peptide (YSAYPDSVPMMS) can be effective in delivering anticancer agents to such tumors. Hence, we report on the synthesis and characterizations of a novel EphA2-targeting agent conjugated with the chemotherapeutic drug paclitaxel. We found that the peptide-drug conjugate is dramatically more effective than paclitaxel alone at inhibiting tumor growth in a prostate cancer xenograft model, delivering significantly higher levels of drug to the tumor site. We believe these studies open the way to the development of a new class of therapeutic compounds that exploit the EphA2 receptor for drug delivery to cancer cells.
Collapse
Affiliation(s)
- Si Wang
- Cancer Research Center, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Road, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|