1
|
Lyu C, Yuan L, Yang Y, Zhang D, Hu W, Zhao K, Ding Y, Chen W, Xiao K, Chen Y, Liu W. Ligand preference of EphB2 receptor is selectively regulated by N-glycosylation. iScience 2025; 28:112386. [PMID: 40330885 PMCID: PMC12052844 DOI: 10.1016/j.isci.2025.112386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/25/2024] [Accepted: 04/04/2025] [Indexed: 05/08/2025] Open
Abstract
The Eph receptors and their ephrin ligands play important roles in cell communication and neuron development. Eph interacts with ephrin in a complex manner. Here, we found ephrin-B2 instead of well-recorded ephrin-A5 specifically recognize and activate EphB2 receptor in primary cortical neurons. Domain-swapping and N/Q mutagenesis results show that the ectodomain of EphB2 and its N-glycosylation sites are critical for the ephrin binding selectivity. The N265, N336, N428, and N482Q mutant EphB2 cannot distinguish ephrin-B2 from ephrin-A5. Furthermore, the N-glycosylation sites in EphB2 are evolutionarily conserved and the N-glycan-directed binding strategy is commonly used in other Eph family members. A gain-of-function EphB6 mutant restores its ephrin-B2 binding ability. Finally, EphB2 is robustly glycosylated in the mouse brain and N-glycosylation is required for EphB2 signaling-induced cell rounding and dendritic spine formation. Collectively, our findings provide a molecular basis to understand the exquisite Eph/ephrin interaction preferences.
Collapse
Affiliation(s)
- Chunyu Lyu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Lin Yuan
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, Guangdong 518057, China
| | - Yang Yang
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Dongsheng Zhang
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Wei Hu
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China
| | - Keli Zhao
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Yuzhen Ding
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Wei Chen
- Department of Cell Biology, Zhejiang University School of Medicine, and Liangzhu Laboratory, Zhejiang University, Hangzhou 310000, Zhejiang, China
| | - Kang Xiao
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
- HKUST Shenzhen-Hong Kong Collaborative Innovation Research Institute, Futian, Shenzhen 518045, China
| | - Yu Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen–Hong Kong Institute of Brain Science–Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong 518055, China
- SIAT-HKUST Joint Laboratory for Brain Science, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wei Liu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| |
Collapse
|
2
|
Liu M, Charek JG, Vicetti Miguel RD, Cherpes TL. Ephrin-Eph signaling: an important regulator of epithelial integrity and barrier function. Tissue Barriers 2025:2462855. [PMID: 39921660 DOI: 10.1080/21688370.2025.2462855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/20/2025] [Accepted: 01/29/2025] [Indexed: 02/10/2025] Open
Abstract
Eph receptor-interacting proteins (ephrin) ligands and their erythropoietin-producing human hepatocellular (Eph) receptors elicit bidirectional signals that regulate cell migration, angiogenesis, neuronal plasticity, and other developmental processes in the embryo. In adulthood, ephrin-Eph signaling regulates numerous homeostatic events, including epithelial cell proliferation and differentiation. Epithelial surfaces, including those of skin and vagina, are lined by layers of stratified squamous epithelium (SSE) that protect against mechanical stress and microbial pathogen invasion. Ephrin-Eph signaling is known to promote cutaneous epithelial barrier function by regulating the expression of specialized cell-cell adhesion junctions termed desmosomes, but the role of this signaling system in maintaining epithelial integrity and barrier function in the vagina is less explored. This review summarizes current understanding of ephrin-Eph signaling that regulates desmosome expression and barrier function in the skin and considers evidence that suggests ephrin-Eph signaling similarly regulates these processes in vaginal SSE.
Collapse
Affiliation(s)
- Mohan Liu
- Comparative Biomedical Sciences Graduate Program, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
- Department of Otolaryngology - Head and Neck Surgery, The Ohio State University, Columbus, OH, USA
| | - Joseph G Charek
- Department of Otolaryngology - Head and Neck Surgery, The Ohio State University, Columbus, OH, USA
| | - Rodolfo D Vicetti Miguel
- Department of Otolaryngology - Head and Neck Surgery, The Ohio State University, Columbus, OH, USA
| | - Thomas L Cherpes
- Department of Otolaryngology - Head and Neck Surgery, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
3
|
Offenhäuser C, Dave KA, Beckett KJ, Smith FM, Jayakody BA, Cooper LT, Agyei-Yeboah H, McCarron JK, Li Y, Bastick K, Al-Ejeh F, Cullen JK, Coulthard MG, Gorman JJ, Boyd AW, Day BW. EphA2 regulates vascular permeability and prostate cancer metastasis via modulation of cell junction protein phosphorylation. Oncogene 2025; 44:208-227. [PMID: 39511410 PMCID: PMC11753358 DOI: 10.1038/s41388-024-03206-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024]
Abstract
Prostate cancer morbidity and mortality demonstrate a need for more effective targeted therapies. One potential target is EphA2, although paradoxically, pro- and anti-oncogenic effects have been shown to be mediated by EphA2. We demonstrate that unique activating and blocking EphA2-targeting monoclonal antibodies display opposing tumor-suppressive and oncogenic properties in vivo. To further explore this complexity, we performed detailed phosphoproteomic analysis following ligand-induced EphA2 activation. Our analysis identified altered phosphorylation of 73 downstream proteins related to the PI3K/AKT/mTOR and ERK/MAPK pathways, with the majority implicated in cell junction and cytoskeletal organization, cell motility, and tumor metastasis. We demonstrate that the adapter protein SHB is an essential component in mediating the inhibition of the ERK/MAPK pathway in response to EphA2 receptor activation. Furthermore, we identify the adherence junction protein afadin as an EphA2-regulated phosphoprotein which is involved in prostate cancer migration and invasion.
Collapse
Affiliation(s)
- Carolin Offenhäuser
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia.
| | - Keyur A Dave
- Protein Discovery Center, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Kirrilee J Beckett
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Fiona M Smith
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Buddhika A Jayakody
- Protein Discovery Center, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Leanne T Cooper
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Helen Agyei-Yeboah
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Jennifer K McCarron
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Yuchen Li
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Kate Bastick
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Fares Al-Ejeh
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Jason K Cullen
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Mark G Coulthard
- Mayne Academy of Paediatrics, Faculty of Medicine, The University of Queensland, Queensland Children's Hospital, Brisbane, QLD, 4101, Australia
- Paediatric Intensive Care Unit, Queensland Children's Hospital, Brisbane, QLD, 4101, Australia
| | - Jeffrey J Gorman
- Protein Discovery Center, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Andrew W Boyd
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- School of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Bryan W Day
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia.
- School of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia.
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, 4059, Australia.
| |
Collapse
|
4
|
Horner JL, Vu MP, Clark JT, Innis IJ, Cheng C. Canonical ligand-dependent and non-canonical ligand-independent EphA2 signaling in the eye lens of wild-type, knockout, and aging mice. Aging (Albany NY) 2024; 16:13039-13075. [PMID: 39466050 PMCID: PMC11552635 DOI: 10.18632/aging.206144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/29/2024] [Indexed: 10/29/2024]
Abstract
Disruption of Eph-ephrin bidirectional signaling leads to human congenital and age-related cataracts, but the mechanisms for these opacities in the eye lens remain unclear. Eph receptors bind to ephrin ligands on neighboring cells to induce canonical ligand-mediated signaling. The EphA2 receptor also signals non-canonically without ligand binding in cancerous cells, leading to epithelial-to-mesenchymal transition (EMT). We have previously shown that the receptor EphA2 and the ligand ephrin-A5 have diverse functions in maintaining lens transparency in mice. Loss of ephrin-A5 leads to anterior cataracts due to EMT. Surprisingly, both canonical and non-canonical EphA2 activation are present in normal wild-type lenses and in the ephrin-A5 knockout lenses. Canonical EphA2 signaling is localized exclusively to lens epithelial cells and does not change with age. Non-canonical EphA2 signaling is in both epithelial and fiber cells and increases significantly with age. We hypothesize that canonical ligand-dependent EphA2 signaling is required for the morphogenesis and organization of hexagonal equatorial epithelial cells while non-canonical ligand-independent EphA2 signaling is needed for complex membrane interdigitations that change during fiber cell differentiation and maturation. This is the first demonstration of non-canonical EphA2 activation in a non-cancerous tissue or cell and suggests a possible physiological function for ligand-independent EphA2 signaling.
Collapse
Affiliation(s)
- Jenna L. Horner
- School of Optometry and Vision Science Program, Indiana University, Bloomington, IN 47405, USA
| | - Michael P. Vu
- School of Optometry and Vision Science Program, Indiana University, Bloomington, IN 47405, USA
| | - Jackson T. Clark
- School of Optometry and Vision Science Program, Indiana University, Bloomington, IN 47405, USA
| | - Isaiah J. Innis
- School of Optometry and Vision Science Program, Indiana University, Bloomington, IN 47405, USA
| | - Catherine Cheng
- School of Optometry and Vision Science Program, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
5
|
Giordano G, Tucciarello C, Merlini A, Cutrupi S, Pignochino Y. Targeting the EphA2 pathway: could it be the way for bone sarcomas? Cell Commun Signal 2024; 22:433. [PMID: 39252029 PMCID: PMC11382444 DOI: 10.1186/s12964-024-01811-7] [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: 04/09/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
Bone sarcomas are malignant tumors of mesenchymal origin. Complete surgical resection is the cornerstone of multidisciplinary treatment. However, advanced, unresectable forms remain incurable. A crucial step towards addressing this challenge involves comprehending the molecular mechanisms underpinning tumor progression and metastasis, laying the groundwork for innovative precision medicine-based interventions. We previously showed that tyrosine kinase receptor Ephrin Type-A Receptor 2 (EphA2) is overexpressed in bone sarcomas. EphA2 is a key oncofetal protein implicated in metastasis, self-renewal, and chemoresistance. Molecular, genetic, biochemical, and pharmacological approaches have been developed to target EphA2 and its signaling pathway aiming to interfere with its tumor-promoting effects or as a carrier for drug delivery. This review synthesizes the main functions of EphA2 and their relevance in bone sarcomas, providing strategies devised to leverage this receptor for diagnostic and therapeutic purposes, with a focus on its applicability in the three most common bone sarcoma histotypes: osteosarcoma, chondrosarcoma, and Ewing sarcoma.
Collapse
Affiliation(s)
- Giorgia Giordano
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, TO, Italy
- Department of Oncology, University of Turin, 10043, Orbassano, TO, Italy
| | - Cristina Tucciarello
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, TO, Italy
- Department of Clinical and Biological Sciences, University of Turin, 10043, Orbassano, TO, Italy
| | - Alessandra Merlini
- Department of Oncology, University of Turin, 10043, Orbassano, TO, Italy
| | - Santina Cutrupi
- Department of Clinical and Biological Sciences, University of Turin, 10043, Orbassano, TO, Italy
| | - Ymera Pignochino
- Sarcoma Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060, Candiolo, TO, Italy.
- Department of Clinical and Biological Sciences, University of Turin, 10043, Orbassano, TO, Italy.
| |
Collapse
|
6
|
Wirth D, Özdemir E, Wimley WC, Pasquale EB, Hristova K. Transmembrane helix interactions regulate oligomerization of the receptor tyrosine kinase EphA2. J Biol Chem 2024; 300:107441. [PMID: 38838777 PMCID: PMC11263659 DOI: 10.1016/j.jbc.2024.107441] [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: 04/02/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/07/2024] Open
Abstract
The transmembrane helices of receptor tyrosine kinases (RTKs) have been proposed to switch between two different dimeric conformations, one associated with the inactive RTK and the other with the active RTK. Furthermore, recent work has demonstrated that some full-length RTKs are associated into oligomers that are larger than dimers, raising questions about the roles of the TM helices in the assembly and function of these oligomers. Here we probe the roles of the TM helices in the assembly of EphA2 RTK oligomers in the plasma membrane. We employ mutagenesis to evaluate the relevance of a published NMR dimeric structure of the isolated EphA2 TM helix in the context of the full-length EphA2 in the plasma membrane. We use two fluorescence methods, Förster Resonance Energy Transfer and Fluorescence Intensity Fluctuations spectrometry, which yield complementary information about the EphA2 oligomerization process. These studies reveal that the TM helix mutations affect the stability, structure, and size of EphA2 oligomers. However, the effects are multifaceted and point to a more complex role of the TM helix than the one expected from the "TM dimer switch" model.
Collapse
Affiliation(s)
- Daniel Wirth
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ece Özdemir
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - William C Wimley
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Elena B Pasquale
- Cancer Metabolism and Microenvironment Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Kalina Hristova
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA.
| |
Collapse
|
7
|
Khan N, Kumar V, Li P, Schlapbach LJ, Boyd AW, Coulthard MG, Woodruff TM. Inhibiting Eph/ephrin signaling reduces vascular leak and endothelial cell dysfunction in mice with sepsis. Sci Transl Med 2024; 16:eadg5768. [PMID: 38657024 DOI: 10.1126/scitranslmed.adg5768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/02/2024] [Indexed: 04/26/2024]
Abstract
Sepsis is a life-threatening disease caused by a dysregulated host response to infection, resulting in 11 million deaths globally each year. Vascular endothelial cell dysfunction results in the loss of endothelial barrier integrity, which contributes to sepsis-induced multiple organ failure and mortality. Erythropoietin-producing hepatocellular carcinoma (Eph) receptors and their ephrin ligands play a key role in vascular endothelial barrier disruption but are currently not a therapeutic target in sepsis. Using a cecal ligation and puncture (CLP) mouse model of sepsis, we showed that prophylactic or therapeutic treatment of mice with EphA4-Fc, a decoy receptor and pan-ephrin inhibitor, resulted in improved survival and a reduction in vascular leak, lung injury, and endothelial cell dysfunction. EphA2-/- mice also exhibited reduced mortality and pathology after CLP compared with wild-type mice. Proteomics of plasma samples from mice with sepsis after CLP revealed dysregulation of a number of Eph/ephrins, including EphA2/ephrin A1. Administration of EphA4-Fc to cultured human endothelial cells pretreated with TNF-α or ephrin-A1 prevented loss of endothelial junction proteins, specifically VE-cadherin, with maintenance of endothelial barrier integrity. In children admitted to hospital with fever and suspected infection, we observed that changes in EphA2/ephrin A1 in serum samples correlated with endothelial and organ dysfunction. Targeting Eph/ephrin signaling may be a potential therapeutic strategy to reduce sepsis-induced endothelial dysfunction and mortality.
Collapse
Affiliation(s)
- Nemat Khan
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
- Mayne Academy of Paediatrics, Faculty of Medicine, University of Queensland, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
| | - Vinod Kumar
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
| | - Pengcheng Li
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
- Mayne Academy of Paediatrics, Faculty of Medicine, University of Queensland, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
| | - Luregn J Schlapbach
- Children's Intensive Care Research Program, Child Health Research Centre, University of Queensland, Brisbane, QLD 4101, Australia
- Paediatric Intensive Care Unit, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
- Department of Intensive Care and Neonatology, and Children's Research Center, University Children's Hospital Zürich, University of Zürich, 8032 Zürich, Switzerland
| | - Andrew W Boyd
- Faculty of Medicine, University of Queensland, Brisbane, QLD 4006, Australia
| | - Mark G Coulthard
- Mayne Academy of Paediatrics, Faculty of Medicine, University of Queensland, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
- Paediatric Intensive Care Unit, Queensland Children's Hospital, Brisbane, QLD 4101, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
8
|
Hara M, Ishii K, Hattori M, Kohno T. EphA4 Induces the Phosphorylation of an Intracellular Adaptor Protein Dab1 via Src Family Kinases. Biol Pharm Bull 2024; 47:1314-1320. [PMID: 39019611 DOI: 10.1248/bpb.b24-00273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/19/2024]
Abstract
Dab1 is an intracellular adaptor protein essential for brain formation during development. Tyrosine phosphorylation in Dab1 plays important roles in neuronal migration, dendrite development, and synapse formation by affecting several downstream pathways. Reelin is the best-known extracellular protein that induces Dab1 phosphorylation. However, whether other upstream molecule(s) contribute to Dab1 phosphorylation remains largely unknown. Here, we found that EphA4, a member of the Eph family of receptor-type tyrosine kinases, induced Dab1 phosphorylation when co-expressed in cultured cells. Tyrosine residues phosphorylated by EphA4 were the same as those phosphorylated by Reelin in neurons. The autophosphorylation of EphA4 was necessary for Dab1 phosphorylation. We also found that EphA4-induced Dab1 phosphorylation was mediated by the activation of the Src family tyrosine kinases. Interestingly, Dab1 phosphorylation was not observed when EphA4 was activated by ephrin-A5 in cultured cortical neurons, suggesting that Dab1 is localized in a different compartment in them. EphA4-induced Dab1 phosphorylation may occur under limited and/or pathological conditions in the brain.
Collapse
Affiliation(s)
- Mitsuki Hara
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Keisuke Ishii
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Mitsuharu Hattori
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | - Takao Kohno
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University
| |
Collapse
|
9
|
Gomez-Soler M, Olson EJ, de la Torre ER, Zhao C, Lamberto I, Flood DT, Danho W, Lechtenberg BC, Riedl SJ, Dawson PE, Pasquale EB. Lipidation and PEGylation Strategies to Prolong the in Vivo Half-Life of a Nanomolar EphA4 Receptor Antagonist. Eur J Med Chem 2023; 262:115876. [PMID: 38523699 PMCID: PMC10959496 DOI: 10.1016/j.ejmech.2023.115876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/08/2023] [Accepted: 10/12/2023] [Indexed: 03/26/2024]
Abstract
The EphA4 receptor tyrosine kinase plays a role in neurodegenerative diseases, inhibition of nerve regeneration, cancer progression and other diseases. Therefore, EphA4 inhibition has potential therapeutic value. Selective EphA4 kinase inhibitors are not available, but we identified peptide antagonists that inhibit ephrin ligand binding to EphA4 with high specificity. One of these peptides is the cyclic APY-d3 (βAPYCVYRβASWSC-NH2), which inhibits ephrin-A5 ligand binding to EphA4 with low nanomolar binding affinity and is highly protease resistant. Here we describe modifications of APY-d3 that yield two different key derivatives with greatly increased half-lives in the mouse circulation, the lipidated APY-d3-laur8 and the PEGylated APY-d3-PEG4. These two derivatives inhibit ligand induced EphA4 activation in cells with sub-micromolar potency. Since they retain high potency and specificity for EphA4, lipidated and PEGylated APY-d3 derivatives represent new tools for discriminating EphA4 activities in vivo and for preclinical testing of EphA4 inhibition in animal disease models.
Collapse
Affiliation(s)
- Maricel Gomez-Soler
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Erika J. Olson
- Departments of Chemistry and Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Elena Rubio de la Torre
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Chunxia Zhao
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Ilaria Lamberto
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Dillon T. Flood
- Departments of Chemistry and Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Waleed Danho
- Del Mar, California 92014, United States
- Deceased
| | - Bernhard C. Lechtenberg
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Stefan J. Riedl
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Philip E. Dawson
- Departments of Chemistry and Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Elena B. Pasquale
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| |
Collapse
|
10
|
Kim Y, Ahmed S, Miller WT. Colorectal cancer-associated mutations impair EphB1 kinase function. J Biol Chem 2023; 299:105115. [PMID: 37527777 PMCID: PMC10463257 DOI: 10.1016/j.jbc.2023.105115] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/03/2023] Open
Abstract
Erythropoietin-producing hepatoma (Eph) receptor tyrosine kinases regulate the migration and adhesion of cells that are required for many developmental processes and adult tissue homeostasis. In the intestinal epithelium, Eph signaling controls the positioning of cell types along the crypt-villus axis. Eph activity can suppress the progression of colorectal cancer (CRC). The most frequently mutated Eph receptor in metastatic CRC is EphB1. However, the functional effects of EphB1 mutations are mostly unknown. We expressed and purified the kinase domains of WT and five cancer-associated mutant EphB1 and developed assays to assess the functional effects of the mutations. Using purified proteins, we determined that CRC-associated mutations reduce the activity and stability of the folded structure of EphB1. By mammalian cell expression, we determined that CRC-associated mutant EphB1 receptors inhibit signal transducer and activator of transcription 3 and extracellular signal-regulated kinases 1 and 2 signaling. In contrast to the WT, the mutant EphB1 receptors are unable to suppress the migration of human CRC cells. The CRC-associated mutations also impair cell compartmentalization in an assay in which EphB1-expressing cells are cocultured with ligand (ephrin B1)-expressing cells. These results suggest that somatic mutations impair the kinase-dependent tumor suppressor function of EphB1 in CRC.
Collapse
Affiliation(s)
- Yunyoung Kim
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, New York, USA
| | - Sultan Ahmed
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, New York, USA
| | - W Todd Miller
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, New York, USA; Department of Veterans Affairs Medical Center, Northport, New York, USA.
| |
Collapse
|
11
|
Yan K, Bormuth I, Bormuth O, Tutukova S, Renner A, Bessa P, Schaub T, Rosário M, Tarabykin V. TrkB-dependent EphrinA reverse signaling regulates callosal axon fasciculate growth downstream of Neurod2/6. Cereb Cortex 2023; 33:1752-1767. [PMID: 35462405 DOI: 10.1093/cercor/bhac170] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 11/14/2022] Open
Abstract
Abnormal development of corpus callosum is relatively common and causes a broad spectrum of cognitive impairments in humans. We use acallosal Neurod2/6-deficient mice to study callosal axon guidance within the ipsilateral cerebral cortex. Initial callosal tracts form but fail to traverse the ipsilateral cingulum and are not attracted towards the midline in the absence of Neurod2/6. We show that the restoration of Ephrin-A4 (EfnA4) expression in the embryonic neocortex of Neurod2/6-deficient embryos is sufficient to partially rescue targeted callosal axon growth towards the midline. EfnA4 cannot directly mediate reverse signaling within outgrowing axons, but it forms co-receptor complexes with TrkB (Ntrk2). The ability of EfnA4 to rescue the guided growth of a subset of callosal axons in Neurod2/6-deficient mice is abolished by the co-expression of dominant negative TrkBK571N (kinase-dead) or TrkBY515F (SHC-binding deficient) variants, but not by TrkBY816F (PLCγ1-binding deficient). Additionally, EphA4 is repulsive to EfnA4-positive medially projecting axons in organotypic brain slice culture. Collectively, we suggest that EfnA4-mediated reverse signaling acts via TrkB-SHC and is required for ipsilateral callosal axon growth accuracy towards the midline downstream of Neurod family factors.
Collapse
Affiliation(s)
- Kuo Yan
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany
| | - Ingo Bormuth
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany
| | - Olga Bormuth
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany.,Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 603950, Nizhny Novgorod Oblast, Russia
| | - Svetlana Tutukova
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 603950, Nizhny Novgorod Oblast, Russia.,Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634009, Tomsk, Russia
| | - Ana Renner
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany
| | - Paraskevi Bessa
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany
| | - Theres Schaub
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany
| | - Marta Rosário
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany
| | - Victor Tarabykin
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, D-10117, Berlin, Germany.,Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 603950, Nizhny Novgorod Oblast, Russia.,Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634009, Tomsk, Russia
| |
Collapse
|
12
|
Mapping the Universe of Eph Receptor and Ephrin Ligand Transcripts in Epithelial and Fiber Cells of the Eye Lens. Cells 2022; 11:cells11203291. [PMID: 36291158 PMCID: PMC9600312 DOI: 10.3390/cells11203291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/14/2022] [Accepted: 10/15/2022] [Indexed: 11/17/2022] Open
Abstract
The eye lens is a transparent, ellipsoid organ in the anterior chamber of the eye that is required for fine focusing of light onto the retina to transmit a clear image. Cataracts, defined as any opacity in the lens, remains the leading cause of blindness in the world. Recent studies in humans and mice indicate that Eph–ephrin bidirectional signaling is important for maintaining lens transparency. Specifically, mutations and polymorphisms in the EphA2 receptor and the ephrin-A5 ligand have been linked to congenital and age-related cataracts. It is unclear what other variants of Ephs and ephrins are expressed in the lens or whether there is preferential expression in epithelial vs. fiber cells. We performed a detailed analysis of Eph receptor and ephrin ligand mRNA transcripts in whole mouse lenses, epithelial cell fractions, and fiber cell fractions using a new RNA isolation method. We compared control samples with EphA2 knockout (KO) and ephrin-A5 KO samples. Our results revealed the presence of transcripts for 12 out of 14 Eph receptors and 8 out of 8 ephrin ligands in various fractions of lens cells. Using specific primer sets, RT-PCR, and sequencing, we verified the variant of each gene that is expressed, and we found two epithelial-cell-specific genes. Surprisingly, we also identified one Eph receptor variant that is expressed in KO lens fibers but is absent from control lens fibers. We also identified one low expression ephrin variant that is only expressed in ephrin-A5 control samples. These results indicate that the lens expresses almost all Ephs and ephrins, and there may be many receptor–ligand pairs that play a role in lens homeostasis.
Collapse
|
13
|
Banerjee SL, Lessard F, Chartier FJM, Jacquet K, Osornio-Hernandez AI, Teyssier V, Ghani K, Lavoie N, Lavoie JN, Caruso M, Laprise P, Elowe S, Lambert JP, Bisson N. EPH receptor tyrosine kinases phosphorylate the PAR-3 scaffold protein to modulate downstream signaling networks. Cell Rep 2022; 40:111031. [PMID: 35793621 DOI: 10.1016/j.celrep.2022.111031] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 04/26/2022] [Accepted: 06/13/2022] [Indexed: 11/03/2022] Open
Abstract
EPH receptors (EPHRs) constitute the largest family among receptor tyrosine kinases in humans. They are mainly involved in short-range cell-cell communication events that regulate cell adhesion, migration, and boundary formation. However, the molecular mechanisms by which EPHRs control these processes are less understood. To address this, we unravel EPHR-associated complexes under native conditions using mass-spectrometry-based BioID proximity labeling. We obtain a composite proximity network from EPHA4, -B2, -B3, and -B4 that comprises 395 proteins, most of which were not previously linked to EPHRs. We examine the contribution of several BioID-identified candidates via loss-of-function in an EPHR-dependent cell-segregation assay. We find that the signaling scaffold PAR-3 is required for cell sorting and that EPHRs directly phosphorylate PAR-3. We also delineate a signaling complex involving the C-terminal SRC kinase (CSK), whose recruitment to PAR-3 is dependent on EPHR signals. Our work describes signaling networks by which EPHRs regulate cellular phenotypes.
Collapse
Affiliation(s)
- Sara L Banerjee
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Quebec-Université Laval, Division Oncologie, Québec, QC, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, QC, Canada; PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec, QC, Canada
| | - Frédéric Lessard
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Quebec-Université Laval, Division Oncologie, Québec, QC, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, QC, Canada; PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec, QC, Canada
| | - François J M Chartier
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Quebec-Université Laval, Division Oncologie, Québec, QC, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, QC, Canada; PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec, QC, Canada
| | - Kévin Jacquet
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Quebec-Université Laval, Division Oncologie, Québec, QC, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, QC, Canada
| | - Ana I Osornio-Hernandez
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Quebec-Université Laval, Division Oncologie, Québec, QC, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, QC, Canada; PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec, QC, Canada
| | - Valentine Teyssier
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Quebec-Université Laval, Division Oncologie, Québec, QC, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, QC, Canada; PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec, QC, Canada
| | - Karim Ghani
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Quebec-Université Laval, Division Oncologie, Québec, QC, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, QC, Canada
| | - Noémie Lavoie
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Quebec-Université Laval, Division Oncologie, Québec, QC, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, QC, Canada; PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec, QC, Canada
| | - Josée N Lavoie
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Quebec-Université Laval, Division Oncologie, Québec, QC, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, QC, Canada; Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, QC, Canada
| | - Manuel Caruso
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Quebec-Université Laval, Division Oncologie, Québec, QC, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, QC, Canada; Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, QC, Canada
| | - Patrick Laprise
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Quebec-Université Laval, Division Oncologie, Québec, QC, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, QC, Canada; Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, QC, Canada
| | - Sabine Elowe
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Quebec-Université Laval, Division Oncologie, Québec, QC, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, QC, Canada; PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec, QC, Canada; Department of Pediatrics, Université Laval, Québec, QC, Canada
| | - Jean-Philippe Lambert
- Centre de recherche sur le cancer de l'Université Laval, Québec, QC, Canada; Department of Molecular Medicine, Université Laval, Québec, QC, Canada; Centre de recherche en données massives de l'Université Laval, Québec, QC, Canada; Centre de recherche du Centre Hospitalier Universitaire (CHU) de Quebec-Université Laval, Division Endocrinologie-néphrologie, Québec, QC, Canada
| | - Nicolas Bisson
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Quebec-Université Laval, Division Oncologie, Québec, QC, Canada; Centre de recherche sur le cancer de l'Université Laval, Québec, QC, Canada; PROTEO-Quebec Network for Research on Protein Function, Engineering, and Applications, Québec, QC, Canada; Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, QC, Canada.
| |
Collapse
|
14
|
Cai C, Zhang M, Liu L, Zhang H, Guo Y, Lan T, Xu Y, Ma P, Li S. ADAM10-cleaved ephrin-A5 contributes to prostate cancer metastasis. Cell Death Dis 2022; 13:453. [PMID: 35551177 PMCID: PMC9098485 DOI: 10.1038/s41419-022-04893-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/27/2022] [Accepted: 04/28/2022] [Indexed: 12/14/2022]
Abstract
A disintegrin and metalloprotease-10(ADAM10) promotes the metastasis of prostate cancer (PCa), but the specific mechanism is indistinct. Herein, DU145 cell lines with stable overexpression and knockdown of ADAM10 were constructed. We found that ectopic expression of ADAM10 not only significantly facilitated cell proliferation, migration, invasion, and inhibited apoptosis, but also could specifically hydrolyze ephrin-A5 and release the ephrin-A5 soluble ectodomain into extracellular media in vitro. These effects were reversed by ADAM10 depletion or treatment of GI254023X. Meanwhile, the co-location and physical interaction among EphA3, ephrin-A5, and ADAM10 were observed in PCa cells using immunofluorescence and immunoprecipitation techniques. Interestingly, overexpression of EphA3 exerted opposite effects in DU145 (ephrin-A5 + ) cells and PC-3 (ephrin-A5 ± ) cells. In addition, the pro-tumor function of EphA3 was reversed by the treatment with the exogenous ephrin-A5-Fc, which increased the phosphorylation level of EphA3 in PC-3 (ephrin-A5 ± ) cells. In nude mice, ADAM10 accelerated growth of the primary tumor, decreased the level of ephrin-A5 in the tumor tissue, but increased the level of ephrin-A5 in the peripheral blood, accompanied with an increase in the expression of CD31 and VEGF (vascular endothelial growth factor) in the tissue. What is more, the serum ephrin-A5 content of patients with metastatic PCa was significantly higher than that of the non-metastatic group (P < 0.05). The receiver operating characteristic curve(ROC) showed that the area under the curve(AUC) of serum ephrin-A5 as a marker of PCa metastasis was 0.843, with a sensitivity of 93.5% and a specificity of 75%. It is concluded that ADAM10-mediated ephrin-A5 shedding promotes PCa metastasis via transforming the role of EphA3 from ligand-dependent tumor suppressor to ligand-independent promoter, and ephrin-A5 in the blood can be used as a new biomarker for PCa metastasis.
Collapse
Affiliation(s)
- Chenchen Cai
- grid.417303.20000 0000 9927 0537Medical Technology School of Xuzhou Medical University, Xuzhou, 221004 China ,grid.452207.60000 0004 1758 0558Xuzhou Central Hospital, The Affiliated Xuzhou Hospital of Medical College of Southeast University, Xuzhou, 221009 China
| | - Miaomiao Zhang
- grid.417303.20000 0000 9927 0537Medical Technology School of Xuzhou Medical University, Xuzhou, 221004 China ,grid.413389.40000 0004 1758 1622Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 PR China
| | - Lei Liu
- grid.417303.20000 0000 9927 0537Department of Physiology, Xuzhou Medical University, Xuzhou, 221004 PR China
| | - Haoliang Zhang
- grid.413389.40000 0004 1758 1622Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 PR China
| | - Yi Guo
- grid.413389.40000 0004 1758 1622Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 PR China
| | - Ting Lan
- grid.417303.20000 0000 9927 0537Medical Technology School of Xuzhou Medical University, Xuzhou, 221004 China
| | - Yinhai Xu
- grid.413389.40000 0004 1758 1622Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 PR China
| | - Ping Ma
- grid.417303.20000 0000 9927 0537Medical Technology School of Xuzhou Medical University, Xuzhou, 221004 China ,grid.413389.40000 0004 1758 1622Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 PR China
| | - Shibao Li
- grid.417303.20000 0000 9927 0537Medical Technology School of Xuzhou Medical University, Xuzhou, 221004 China ,grid.413389.40000 0004 1758 1622Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 PR China
| |
Collapse
|
15
|
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: 1.0] [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
|
16
|
Murugan S, Cheng C. Roles of Eph-Ephrin Signaling in the Eye Lens Cataractogenesis, Biomechanics, and Homeostasis. Front Cell Dev Biol 2022; 10:852236. [PMID: 35295853 PMCID: PMC8918484 DOI: 10.3389/fcell.2022.852236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/04/2022] [Indexed: 01/26/2023] Open
Abstract
The eye lens is responsible for fine focusing of light onto the retina, and its function relies on tissue transparency and biomechanical properties. Recent studies have demonstrated the importance of Eph-ephrin signaling for the maintenance of life-long lens homeostasis. The binding of Eph receptor tyrosine kinases to ephrin ligands leads to a bidirectional signaling pathway that controls many cellular processes. In particular, dysfunction of the receptor EphA2 or the ligand ephrin-A5 lead to a variety of congenital and age-related cataracts, defined as any opacity in the lens, in human patients. In addition, a wealth of animal studies reveal the unique and overlapping functions of EphA2 and ephrin-A5 in lens cell shape, cell organization and patterning, and overall tissue optical and biomechanical properties. Significant differences in lens phenotypes of mouse models with disrupted EphA2 or ephrin-A5 signaling indicate that genetic modifiers likely affect cataract phenotypes and progression, suggesting a possible reason for the variability of human cataracts due to Eph-ephrin dysfunction. This review summarizes the roles of EphA2 and ephrin-A5 in the lens and suggests future avenues of study.
Collapse
|
17
|
Chakraborty S, Varma AK. Crystal structure of clinically reported mutations Gly656Arg, Gly656Glu and Asp751His identified in the kinase domain of EphA7. Biochem Biophys Res Commun 2021; 568:62-67. [PMID: 34186436 DOI: 10.1016/j.bbrc.2021.06.048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 06/13/2021] [Indexed: 10/21/2022]
Abstract
Erythropoietin producing hepatocellular (Eph) forms the largest family of receptor tyrosine kinases (RTK). As a family, Eph regulates physiological events such as cell-cell interaction, cell migration, and adhesion. The Kinase domain is the catalytic core of the Eph receptor and is highly conserved sequentially. EphA7 has been recently regarded as a cancer driver gene and comprises several clinically important mutations. Three of the EphA7 mutations Gly656Glu, Gly656Arg, and Asp751His, present in the kinase domain, are predicted to be highly pathogenic. Furthermore, Gly656Glu and Gly656Arg are reported to be hotspot mutations. Considering the importance of mutations, crystals structure of EphA7 Gly656Glu, Gly656Arg, and Asp751His mutants has been explored. Changes in folding pattern and intramolecular interactions were observed in mutant structures. Secondary structural changes were observed in the hinge region of EphA7 Gly656Arg and Asp751His structure, affecting the transition of kinase domain between open and closed conformations. EphA7 Asp751His mutant structure shows a distorted nucleotide-binding groove. Differences were observed in hydrogen bonding and hydrophobic interactions between the catalytic and highly conserved DFG motif in the EphA7 mutants, which may influence the catalytic activity of kinase domain.
Collapse
Affiliation(s)
- Shubhashish Chakraborty
- Advanced Centre for Treatment, Research and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra, 410210, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, Maharashtra, 400094, India
| | - Ashok K Varma
- Advanced Centre for Treatment, Research and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra, 410210, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, Maharashtra, 400094, India.
| |
Collapse
|
18
|
Fagotto F. Tissue segregation in the early vertebrate embryo. Semin Cell Dev Biol 2020; 107:130-146. [DOI: 10.1016/j.semcdb.2020.05.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 11/30/2022]
|
19
|
Tamura K, Chiu YW, Shiohara A, Hori Y, Tomita T. EphA4 regulates Aβ production via BACE1 expression in neurons. FASEB J 2020; 34:16383-16396. [PMID: 33090569 DOI: 10.1096/fj.202001510r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/28/2020] [Accepted: 10/02/2020] [Indexed: 01/05/2023]
Abstract
Several lines of evidence suggest that the aggregation and deposition of amyloid-β peptide (Aβ) initiate the pathology of Alzheimer's disease (AD). Recently, a genome-wide association study demonstrated that a single-nucleotide polymorphism proximal to the EPHA4 gene, which encodes a receptor tyrosine kinase, is associated with AD risk. However, the molecular mechanism of EphA4 in the pathogenesis of AD, particularly in Aβ production, remains unknown. Here, we performed several pharmacological and biological experiments both in vitro and in vivo and demonstrated that EphA4 is responsible for the regulation of Aβ production. Pharmacological inhibition of EphA4 signaling and knockdown of Epha4 led to increased Aβ levels accompanied by increased expression of β-site APP cleaving enzyme 1 (BACE1), which is an enzyme responsible for Aβ production. Moreover, EPHA4 overexpression and activation of EphA4 signaling via ephrin ligands decreased Aβ levels. In particular, the sterile-alpha motif domain of EphA4 was necessary for the regulation of Aβ production. Finally, EPHA4 mRNA levels were significantly reduced in the brains of AD patients, and negatively correlated with BACE1 mRNA levels. Our results indicate a novel mechanism of Aβ regulation by EphA4, which is involved in AD pathogenesis.
Collapse
Affiliation(s)
- Kensuke Tamura
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yung-Wen Chiu
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Azusa Shiohara
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yukiko Hori
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
20
|
London M, Gallo E. Critical role of EphA3 in cancer and current state of EphA3 drug therapeutics. Mol Biol Rep 2020; 47:5523-5533. [PMID: 32621117 DOI: 10.1007/s11033-020-05571-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 06/05/2020] [Indexed: 12/14/2022]
Abstract
The erythropoietin-producing human hepatocellular (Eph) receptors are transmembrane glycoprotein members of the tyrosine kinase receptors family. The Ephs may bind to various ephrin ligands resulting in the phosphorylation of their tyrosine kinase domain and the activation of the Eph receptor. In this review we focus on EphA3, one receptor of the 14 different Ephs, as it carries out both redundant and restricted functions in the germline development of mammals and in the maintenance of various adult tissues. The loss of EphA3 regulation is correlated with various human malignancies, the most notable being cancer. This receptor is overexpressed and/or mutated in multiple tumors, and is also associated with poor prognosis and decreased survival in patients. Here we highlight the role of EphA3 in normal and malignant tissues that are specific to cancer; these include hematologic disorders, gastric cancer, glioblastoma multiforme, colorectal cancer, lung cancer, renal cell carcinoma, and prostate cancer. Moreover, various anticancer agents against EphA3 have been developed to either inhibit its kinase domain activity or to function as agonists. Thus, we examine the most potent small molecule drugs and mAb-based therapeutics against EphA3 that are currently in pre-clinical or clinical stages.
Collapse
Affiliation(s)
- Max London
- Department of Molecular Genetics, Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - Eugenio Gallo
- Department of Molecular Genetics, Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada.
| |
Collapse
|
21
|
DiPrima M, Wang D, Tröster A, Maric D, Terrades-Garcia N, Ha T, Kwak H, Sanchez-Martin D, Kudlinzki D, Schwalbe H, Tosato G. Identification of Eph receptor signaling as a regulator of autophagy and a therapeutic target in colorectal carcinoma. Mol Oncol 2019; 13:2441-2459. [PMID: 31545551 PMCID: PMC6822245 DOI: 10.1002/1878-0261.12576] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 08/02/2019] [Accepted: 09/20/2019] [Indexed: 01/07/2023] Open
Abstract
Advanced colorectal carcinoma is currently incurable, and new therapies are urgently needed. We report that phosphotyrosine‐dependent Eph receptor signaling sustains colorectal carcinoma cell survival, thereby uncovering a survival pathway active in colorectal carcinoma cells. We find that genetic and biochemical inhibition of Eph tyrosine kinase activity or depletion of the Eph ligand EphrinB2 reproducibly induces colorectal carcinoma cell death by autophagy. Spautin and 3‐methyladenine, inhibitors of early steps in the autophagic pathway, significantly reduce autophagy‐mediated cell death that follows inhibition of phosphotyrosine‐dependent Eph signaling in colorectal cancer cells. A small‐molecule inhibitor of the Eph kinase, NVP‐BHG712 or its regioisomer NVP‐Iso, reduces human colorectal cancer cell growth in vitro and tumor growth in mice. Colorectal cancers express the EphrinB ligand and its Eph receptors at significantly higher levels than numerous other cancer types, supporting Eph signaling inhibition as a potential new strategy for the broad treatment of colorectal carcinoma.
Collapse
Affiliation(s)
- Michael DiPrima
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), Bethesda, MD, USA
| | - Dunrui Wang
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), Bethesda, MD, USA
| | - Alix Tröster
- Center for Biomolecular Magnetic Resonance, Institute for Organic Chemistry and Chemical Biology, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Dragan Maric
- National Institutes of Neurological Disorders and Stroke, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Nekane Terrades-Garcia
- Vasculitis Research Unit, Department of Autoimmune Diseases, Hospital Clinic, University of Barcelona, Spain
| | - Taekyu Ha
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), Bethesda, MD, USA
| | - Hyeongil Kwak
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), Bethesda, MD, USA
| | - David Sanchez-Martin
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), Bethesda, MD, USA
| | - Denis Kudlinzki
- Center for Biomolecular Magnetic Resonance, Institute for Organic Chemistry and Chemical Biology, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Harald Schwalbe
- Center for Biomolecular Magnetic Resonance, Institute for Organic Chemistry and Chemical Biology, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Giovanna Tosato
- Laboratory of Cellular Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), Bethesda, MD, USA
| |
Collapse
|
22
|
Magic Z, Sandström J, Perez-Tenorio G. Ephrin‑B2 inhibits cell proliferation and motility in vitro and predicts longer metastasis‑free survival in breast cancer. Int J Oncol 2019; 55:1275-1286. [PMID: 31638179 PMCID: PMC6831205 DOI: 10.3892/ijo.2019.4892] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/12/2019] [Indexed: 12/14/2022] Open
Abstract
The tyrosine kinase receptor EphB4 and its ligand ephrin‑B2 interact through cell‑to‑cell contacts. Upon interaction, EphB4 transmits bidirectional signals. A forward signal inside EphB4‑expressing cells is believed to suppress tumor growth, while inside the ephrin‑expressing cells, an oncogenic reverse signal arises. In breast cancer cells with a high EphB4 receptor expression the forward signal is low, in part due to the low expression of the ligand ephrin‑B2. Therefore, we hypothesized that by re‑introducing the ligand in EphB4‑positive cells, tumor suppression could be induced by the stimulation of the forward signal. This question was addressed in vitro by the stable lentiviral infection of breast cancer cells with either wild‑type EFNB2 or with a mutant EFNB2‑5F, unable to transmit reverse signaling. Furthermore, we investigated ephrin‑B and EphB4 protein expression in 216 paraffin‑embedded tumors using immunohistochemistry. The in vitro results indicated that ephrin‑B2 expression was associated with a lower cell proliferation, migration and motility compared with the control cells. These effects were more pronounced when the cells lacked the ability to transmit the reverse signal (B2‑5F). In clinical material, ephrin‑B protein expression was associated with a positive estrogen receptor (ER) status, a low HER‑2 expression and was negatively associated with Nottingham histologic grade (NHG) III. Ephrin‑B expression indicated a good prognosis, whereas EphB4 expression was associated with a shorter metastasis‑free survival in univariate and multivariate analysis. Furthermore, the prognostic value of EFNB2 and EPHB4 was confirmed at the gene expression level in public datasets. Thus, on the whole, the findings of this study suggest that ephrin‑B2 expression is associated with less proliferation and lower motility of breast cancer cells and with a longer patient survival in breast cancer.
Collapse
Affiliation(s)
- Zeljana Magic
- Department of Clinical and Experimental Medicine and Department of Oncology, Linköping University, SE‑58185 Linköping, Sweden
| | - Josefine Sandström
- Department of Clinical and Experimental Medicine and Department of Oncology, Linköping University, SE‑58185 Linköping, Sweden
| | - Gizeh Perez-Tenorio
- Department of Clinical and Experimental Medicine and Department of Oncology, Linköping University, SE‑58185 Linköping, Sweden
| |
Collapse
|
23
|
Cayuso J, Xu Q, Addison M, Wilkinson DG. Actomyosin regulation by Eph receptor signaling couples boundary cell formation to border sharpness. eLife 2019; 8:49696. [PMID: 31502954 PMCID: PMC6739871 DOI: 10.7554/elife.49696] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 08/23/2019] [Indexed: 02/06/2023] Open
Abstract
The segregation of cells with distinct regional identity underlies formation of a sharp border, which in some tissues serves to organise a boundary signaling centre. It is unclear whether or how border sharpness is coordinated with induction of boundary-specific gene expression. We show that forward signaling of EphA4 is required for border sharpening and induction of boundary cells in the zebrafish hindbrain, which we find both require kinase-dependent signaling, with a lesser input of PDZ domain-dependent signaling. We find that boundary-specific gene expression is regulated by myosin II phosphorylation, which increases actomyosin contraction downstream of EphA4 signaling. Myosin phosphorylation leads to nuclear translocation of Taz, which together with Tead1a is required for boundary marker expression. Since actomyosin contraction maintains sharp borders, there is direct coupling of border sharpness to boundary cell induction that ensures correct organisation of signaling centres.
Collapse
Affiliation(s)
- Jordi Cayuso
- The Francis Crick Institute, London, United Kingdom
| | - Qiling Xu
- The Francis Crick Institute, London, United Kingdom
| | | | | |
Collapse
|
24
|
Liang LY, Patel O, Janes PW, Murphy JM, Lucet IS. Eph receptor signalling: from catalytic to non-catalytic functions. Oncogene 2019; 38:6567-6584. [PMID: 31406248 DOI: 10.1038/s41388-019-0931-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 07/23/2019] [Accepted: 07/24/2019] [Indexed: 12/26/2022]
Abstract
Eph receptors, the largest subfamily of receptor tyrosine kinases, are linked with proliferative disease, such as cancer, as a result of their deregulated expression or mutation. Unlike other tyrosine kinases that have been clinically targeted, the development of therapeutics against Eph receptors remains at a relatively early stage. The major reason is the limited understanding on the Eph receptor regulatory mechanisms at a molecular level. The complexity in understanding Eph signalling in cells arises due to following reasons: (1) Eph receptors comprise 14 members, two of which are pseudokinases, EphA10 and EphB6, with relatively uncharacterised function; (2) activation of Eph receptors results in dimerisation, oligomerisation and formation of clustered signalling centres at the plasma membrane, which can comprise different combinations of Eph receptors, leading to diverse downstream signalling outputs; (3) the non-catalytic functions of Eph receptors have been overlooked. This review provides a structural perspective of the intricate molecular mechanisms that drive Eph receptor signalling, and investigates the contribution of intra- and inter-molecular interactions between Eph receptors intracellular domains and their major binding partners. We focus on the non-catalytic functions of Eph receptors with relevance to cancer, which are further substantiated by exploring the role of the two pseudokinase Eph receptors, EphA10 and EphB6. Throughout this review, we carefully analyse and reconcile the existing/conflicting data in the field, to allow researchers to further the current understanding of Eph receptor signalling.
Collapse
Affiliation(s)
- Lung-Yu Liang
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Onisha Patel
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Peter W Janes
- Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, VIC, 3084, Australia
| | - James M Murphy
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia. .,Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia.
| | - Isabelle S Lucet
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia. .,Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia.
| |
Collapse
|
25
|
Rué L, Oeckl P, Timmers M, Lenaerts A, van der Vos J, Smolders S, Poppe L, de Boer A, Van Den Bosch L, Van Damme P, Weishaupt JH, Ludolph AC, Otto M, Robberecht W, Lemmens R. Reduction of ephrin-A5 aggravates disease progression in amyotrophic lateral sclerosis. Acta Neuropathol Commun 2019; 7:114. [PMID: 31300041 PMCID: PMC6626434 DOI: 10.1186/s40478-019-0759-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 06/22/2019] [Indexed: 12/30/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that affects motor neurons in the brainstem, spinal cord and motor cortex. ALS is characterized by genetic and clinical heterogeneity, suggesting the existence of genetic factors that modify the phenotypic expression of the disease. We previously identified the axonal guidance EphA4 receptor, member of the Eph-ephrin system, as an ALS disease-modifying factor. EphA4 genetic inhibition rescued the motor neuron phenotype in zebrafish and a rodent model of ALS. Preventing ligands from binding to the EphA4 receptor also successfully improved disease, suggesting a role for EphA4 ligands in ALS. One particular ligand, ephrin-A5, is upregulated in reactive astrocytes after acute neuronal injury and inhibits axonal regeneration. Moreover, it plays a role during development in the correct pathfinding of motor axons towards their target limb muscles. We hypothesized that a constitutive reduction of ephrin-A5 signalling would benefit disease progression in a rodent model for ALS. We discovered that in the spinal cord of control and symptomatic ALS mice ephrin-A5 was predominantly expressed in neurons. Surprisingly, reduction of ephrin-A5 levels in SOD1G93A mice accelerated disease progression and reduced survival without affecting disease onset, motor neuron numbers or innervated neuromuscular junctions in symptomatic mice. These findings suggest ephrin-A5 as a modifier of disease progression that might play a role in the later stages of the disease. Similarly, we identified a more aggressive disease progression in patients with lower ephrin-A5 protein levels in the cerebrospinal fluid without modifying disease onset. In summary, we identified reduced expression of ephrin-A5 to accelerate disease progression in a mouse model of ALS as well as in humans. Combined with our previous findings on the role of EphA4 in ALS our current data suggests different contribution for various members of the Eph-ephrin system in the pathophysiology of a motor neuron disease.
Collapse
|
26
|
Darling TK, Lamb TJ. Emerging Roles for Eph Receptors and Ephrin Ligands in Immunity. Front Immunol 2019; 10:1473. [PMID: 31333644 PMCID: PMC6620610 DOI: 10.3389/fimmu.2019.01473] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/13/2019] [Indexed: 12/30/2022] Open
Abstract
Eph receptors are the largest family of receptor tyrosine kinases and mediate a myriad of essential processes in humans from embryonic development to adult tissue homeostasis through interactions with membrane-bound ephrin ligands. The ubiquitous expression of Eph receptors and ephrin ligands among the cellular players of the immune system underscores the importance of these molecules in orchestrating an optimal immune response. This review provides an overview of the various roles of Eph receptors and ephrin ligands in immune cell development, activation, and migration. We also discuss the role of Eph receptors in disease pathogenesis as well as the implications of Eph receptors as future immunotherapy targets. Given the diverse and critical roles of Eph receptors and ephrin ligands throughout the immune system during both resting and activated states, this review aims to highlight the critical yet underappreciated roles of this family of signaling molecules in the immune system.
Collapse
Affiliation(s)
- Thayer K Darling
- Immunology and Molecular Pathogenesis Program, Emory University Laney Graduate School, Atlanta, GA, United States.,Department of Pathology, University of Utah, Salt Lake City, UT, United States
| | - Tracey J Lamb
- Department of Pathology, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
27
|
Peng X, Emiliani F, Smallwood PM, Rattner A, Lei H, Sabbagh MF, Nathans J. Affinity capture of polyribosomes followed by RNAseq (ACAPseq), a discovery platform for protein-protein interactions. eLife 2018; 7:40982. [PMID: 30345971 PMCID: PMC6197854 DOI: 10.7554/elife.40982] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 10/02/2018] [Indexed: 02/05/2023] Open
Abstract
Defining protein-protein interactions (PPIs) is central to the biological sciences. Here, we present a novel platform - Affinity Capture of Polyribosomes followed by RNA sequencing (ACAPseq) - for identifying PPIs. ACAPseq harnesses the power of massively parallel RNA sequencing (RNAseq) to quantify the enrichment of polyribosomes based on the affinity of their associated nascent polypeptides for an immobilized protein 'bait'. This method was developed and tested using neonatal mouse brain polyribosomes and a variety of extracellular domains as baits. Of 92 baits tested, 25 identified one or more binding partners that appear to be biologically relevant; additional candidate partners remain to be validated. ACAPseq can detect binding to targets that are present at less than 1 part in 100,000 in the starting polyribosome preparation. One of the observed PPIs was analyzed in detail, revealing the mode of homophilic binding for Protocadherin-9 (PCDH9), a non-clustered Protocadherin family member.
Collapse
Affiliation(s)
- Xi Peng
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States.,Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Francesco Emiliani
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Philip M Smallwood
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States.,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Amir Rattner
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Hong Lei
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Mark F Sabbagh
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Jeremy Nathans
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States.,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States.,Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
28
|
Kaplan N, Ventrella R, Peng H, Pal-Ghosh S, Arvanitis C, Rappoport JZ, Mitchell BJ, Stepp MA, Lavker RM, Getsios S. EphA2/Ephrin-A1 Mediate Corneal Epithelial Cell Compartmentalization via ADAM10 Regulation of EGFR Signaling. Invest Ophthalmol Vis Sci 2018; 59:393-406. [PMID: 29351356 PMCID: PMC5774870 DOI: 10.1167/iovs.17-22941] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Purpose Progenitor cells of the limbal epithelium reside in a discrete area peripheral to the more differentiated corneal epithelium and maintain tissue homeostasis. What regulates the limbal-corneal epithelial boundary is a major unanswered question. Ephrin-A1 ligand is enriched in the limbal epithelium, whereas EphA2 receptor is concentrated in the corneal epithelium. This reciprocal pattern led us to assess the role of ephrin-A1 and EphA2 in limbal-corneal epithelial boundary organization. Methods EphA2-expressing corneal epithelial cells engineered to express ephrin-A1 were used to study boundary formation in vitro in a manner that mimicked the relative abundance of these juxtamembrane signaling proteins in the limbal and corneal epithelium in vivo. Interaction of these two distinct cell populations following initial seeding into discrete culture compartments was assessed by live cell imaging. Immunofluoresence and immunoblotting was used to evaluate the contribution of downstream growth factor signaling and cell-cell adhesion systems to boundary formation at sites of heterotypic contact between ephrin-A1 and EphA2 expressing cells. Results Ephrin-A1-expressing cells impeded and reversed the migration of EphA2-expressing corneal epithelial cells upon heterotypic contact formation leading to coordinated migration of the two cell populations in the direction of an ephrin-A1-expressing leading front. Genetic silencing and pharmacologic inhibitor studies demonstrated that the ability of ephrin-A1 to direct migration of EphA2-expressing cells depended on an a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) and epidermal growth factor receptor (EGFR) signaling pathway that limited E-cadherin-mediated adhesion at heterotypic boundaries. Conclusions Ephrin-A1/EphA2 signaling complexes play a key role in limbal-corneal epithelial compartmentalization and the response of these tissues to injury.
Collapse
Affiliation(s)
- Nihal Kaplan
- Department of Dermatology, Northwestern University, Chicago, Illinois, United States
| | - Rosa Ventrella
- Department of Dermatology, Northwestern University, Chicago, Illinois, United States
| | - Han Peng
- Department of Dermatology, Northwestern University, Chicago, Illinois, United States
| | - Sonali Pal-Ghosh
- Department of Anatomy and Regenerative Biology, The George Washington University Medical Center, Washington, District of Columbia, United States
| | - Constadina Arvanitis
- Department of Cell and Molecular Biology, Northwestern University, Chicago, Illinois, United States
| | - Joshua Z Rappoport
- Department of Cell and Molecular Biology, Northwestern University, Chicago, Illinois, United States
| | - Brian J Mitchell
- Department of Cell and Molecular Biology, Northwestern University, Chicago, Illinois, United States
| | - Mary Ann Stepp
- Department of Anatomy and Regenerative Biology, The George Washington University Medical Center, Washington, District of Columbia, United States
| | - Robert M Lavker
- Department of Dermatology, Northwestern University, Chicago, Illinois, United States
| | - Spiro Getsios
- Department of Dermatology, Northwestern University, Chicago, Illinois, United States
| |
Collapse
|
29
|
Li S, Zhao Y, Chen W, Yin L, Zhu J, Zhang H, Cai C, Li P, Huang L, Ma P. Exosomal ephrinA2 derived from serum as a potential biomarker for prostate cancer. J Cancer 2018; 9:2659-2665. [PMID: 30087706 PMCID: PMC6072821 DOI: 10.7150/jca.25201] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/03/2018] [Indexed: 12/11/2022] Open
Abstract
Up-regulation of serum ephrinA2 is common in various malignancies and has been suggested as a potential biomarker for the diagnosis and prognosis of prostate cancer (PCa). However, the type of serum ephrinA2 expressed in PCa patients remains elusive. Furthermore, the level of exosomal ephrinA2 derived from serum is increased in patients with osteoporosis, a common complication of PCa patients undergoing androgen deprivation therapy. It is unknown whether exosomes derived from PCa patient serum contains ephrinA2. In this study, we explored the ephrinA2 expression in whole serum and tissues and identified the circulating exosomal ephrinA2 as a potential biomarker for PCa. Exosomes were isolated from patient sera by differential centrifugation and the presence of ephrinA2 was confirmed via electron microscopy and western blotting. The type of ephrinA2 in serum was evaluated by western blotting. The expression of serum ephrinA2 including secreted and cleaved ephrinA2 and exosomal ephrinA2 were detected by ELISA and western blotting. Compared with benign prostatic hyperplasia (BPH) and controls, the levels of whole serum ephrinA2 and exosomal ephrinA2 were significantly higher in PCa patients. Moreover, exosomal ephrinA2 expression was positively correlated with TNM staging and Gleason score of PCa patients. The diagnostic efficiency of exosomal ephrinA2 was superior to that of whole serum ephrinA2 and serum PSA in distinguishing PCa patients from those from BPH patents. Our study indicates that exosomal ephrinA2 has high potential as a biomarker for the presence of PCa and offers a new therapeutic target for this disease.
Collapse
Affiliation(s)
- Shibao Li
- Medical Technology Institute of Xuzhou Medical University, Xuzhou 221004, China.,Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Yao Zhao
- Medical Technology Institute of Xuzhou Medical University, Xuzhou 221004, China
| | - Wenbai Chen
- Department of Nuclear Medicine Laboratory, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Lingyu Yin
- Medical Technology Institute of Xuzhou Medical University, Xuzhou 221004, China
| | - Jie Zhu
- Medical Technology Institute of Xuzhou Medical University, Xuzhou 221004, China
| | - Haoliang Zhang
- Medical Technology Institute of Xuzhou Medical University, Xuzhou 221004, China
| | - Chenchen Cai
- Medical Technology Institute of Xuzhou Medical University, Xuzhou 221004, China
| | - Pengpeng Li
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Lingyan Huang
- Medical Technology Institute of Xuzhou Medical University, Xuzhou 221004, China
| | - Ping Ma
- Medical Technology Institute of Xuzhou Medical University, Xuzhou 221004, China.,Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| |
Collapse
|
30
|
Ventrella R, Kaplan N, Hoover P, Perez White BE, Lavker RM, Getsios S. EphA2 Transmembrane Domain Is Uniquely Required for Keratinocyte Migration by Regulating Ephrin-A1 Levels. J Invest Dermatol 2018; 138:2133-2143. [PMID: 29705292 DOI: 10.1016/j.jid.2018.04.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 04/11/2018] [Accepted: 04/12/2018] [Indexed: 12/19/2022]
Abstract
EphA2 receptor tyrosine kinase is activated by ephrin-A1 ligand, which harbors a glycosylphosphatidylinositol anchor that enhances lipid raft localization. Although EphA2 and ephrin-A1 modulate keratinocyte migration and differentiation, the ability of this cell-cell communication complex to localize to different membrane regions in keratinocytes remains unknown. Using a combination of biochemical and imaging approaches, we provide evidence that ephrin-A1 and a ligand-activated form of EphA2 partition outside of lipid raft domains in response to calcium-mediated cell-cell contact stabilization in normal human epidermal keratinocytes. EphA2 transmembrane domain swapping with a shorter and molecularly distinct transmembrane domain of EphA1 resulted in decreased localization of this receptor tyrosine kinase at cell-cell junctions and increased expression of ephrin-A1, which is a negative regulator of keratinocyte migration. Accordingly, altered EphA2 membrane distribution at cell-cell contacts limited the ability of keratinocytes to seal linear scratch wounds in vitro in an ephrin-A1-dependent manner. Collectively, these studies highlight a key role for the EphA2 transmembrane domain in receptor-ligand membrane distribution at cell-cell contacts that modulates ephrin-A1 levels to allow for efficient keratinocyte migration with relevance for cutaneous wound healing.
Collapse
Affiliation(s)
- Rosa Ventrella
- Department of Dermatology, 303 East Chicago Avenue, Ward 9, Northwestern University, Chicago, Illinois, USA
| | - Nihal Kaplan
- Department of Dermatology, 303 East Chicago Avenue, Ward 9, Northwestern University, Chicago, Illinois, USA
| | - Paul Hoover
- Department of Dermatology, 303 East Chicago Avenue, Ward 9, Northwestern University, Chicago, Illinois, USA
| | - Bethany E Perez White
- Department of Dermatology, 303 East Chicago Avenue, Ward 9, Northwestern University, Chicago, Illinois, USA
| | - Robert M Lavker
- Department of Dermatology, 303 East Chicago Avenue, Ward 9, Northwestern University, Chicago, Illinois, USA
| | - Spiro Getsios
- Department of Dermatology, 303 East Chicago Avenue, Ward 9, Northwestern University, Chicago, Illinois, USA.
| |
Collapse
|
31
|
Apostolova LG, Risacher SL, Duran T, Stage EC, Goukasian N, West JD, Do TM, Grotts J, Wilhalme H, Nho K, Phillips M, Elashoff D, Saykin AJ. Associations of the Top 20 Alzheimer Disease Risk Variants With Brain Amyloidosis. JAMA Neurol 2018; 75:328-341. [PMID: 29340569 PMCID: PMC5885860 DOI: 10.1001/jamaneurol.2017.4198] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 10/19/2017] [Indexed: 01/28/2023]
Abstract
Importance Late-onset Alzheimer disease (AD) is highly heritable. Genome-wide association studies have identified more than 20 AD risk genes. The precise mechanism through which many of these genes are associated with AD remains unknown. Objective To investigate the association of the top 20 AD risk variants with brain amyloidosis. Design, Setting, and Participants This study analyzed the genetic and florbetapir F 18 data from 322 cognitively normal control individuals, 496 individuals with mild cognitive impairment, and 159 individuals with AD dementia who had genome-wide association studies and 18F-florbetapir positron emission tomographic data from the Alzheimer's Disease Neuroimaging Initiative (ADNI), a prospective, observational, multisite tertiary center clinical and biomarker study. This ongoing study began in 2005. Main Outcomes and Measures The study tested the association of AD risk allele carrier status (exposure) with florbetapir mean standard uptake value ratio (outcome) using stepwise multivariable linear regression while controlling for age, sex, and apolipoprotein E ε4 genotype. The study also reports on an exploratory 3-dimensional stepwise regression model using an unbiased voxelwise approach in Statistical Parametric Mapping 8 with cluster and significance thresholds at 50 voxels and uncorrected P < .01. Results This study included 977 participants (mean [SD] age, 74 [7.5] years; 535 [54.8%] male and 442 [45.2%] female) from the ADNI-1, ADNI-2, and ADNI-Grand Opportunity. The adenosine triphosphate-binding cassette subfamily A member 7 (ABCA7) gene had the strongest association with amyloid deposition (χ2 = 8.38, false discovery rate-corrected P < .001), after apolioprotein E ε4. Significant associations were found between ABCA7 in the asymptomatic and early symptomatic disease stages, suggesting an association with rapid amyloid accumulation. The fermitin family homolog 2 (FERMT2) gene had a stage-dependent association with brain amyloidosis (FERMT2 × diagnosis χ2 = 3.53, false discovery rate-corrected P = .05), which was most pronounced in the mild cognitive impairment stage. Conclusions and Relevance This study found an association of several AD risk variants with brain amyloidosis. The data also suggest that AD genes might differentially regulate AD pathologic findings across the disease stages.
Collapse
Affiliation(s)
- Liana G. Apostolova
- Department of Neurology, School of Medicine, Indiana University, Indianapolis
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, School of Medicine, Indiana University, Indianapolis
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis
| | - Shannon L. Risacher
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, School of Medicine, Indiana University, Indianapolis
| | - Tugce Duran
- Department of Neurology, School of Medicine, Indiana University, Indianapolis
| | - Eddie C. Stage
- Department of Neurology, School of Medicine, Indiana University, Indianapolis
| | - Naira Goukasian
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - John D. West
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, School of Medicine, Indiana University, Indianapolis
| | - Triet M. Do
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Jonathan Grotts
- Department of Medicine Statistics Core, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Holly Wilhalme
- Department of Medicine Statistics Core, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, School of Medicine, Indiana University, Indianapolis
| | - Meredith Phillips
- Department of Neurology, School of Medicine, Indiana University, Indianapolis
| | - David Elashoff
- Department of Medicine Statistics Core, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, School of Medicine, Indiana University, Indianapolis
- Department of Medical and Molecular Genetics, School of Medicine, Indiana University, Indianapolis
| |
Collapse
|
32
|
Taylor HB, Khuong A, Wu Z, Xu Q, Morley R, Gregory L, Poliakov A, Taylor WR, Wilkinson DG. Cell segregation and border sharpening by Eph receptor-ephrin-mediated heterotypic repulsion. J R Soc Interface 2018; 14:rsif.2017.0338. [PMID: 28747399 PMCID: PMC5550979 DOI: 10.1098/rsif.2017.0338] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 06/26/2017] [Indexed: 01/06/2023] Open
Abstract
Eph receptor and ephrin signalling has a major role in cell segregation and border formation, and may act through regulation of cell adhesion, repulsion or tension. To elucidate roles of cell repulsion and adhesion, we combined experiments in cell culture assays with quantitations of cell behaviour which are used in computer simulations. Cells expressing EphB2, or kinase-inactive EphB2 (kiEphB2), segregate and form a sharp border with ephrinB1-expressing cells, and this is disrupted by knockdown of N-cadherin. Measurements of contact inhibition of locomotion reveal that EphB2-, kiEphB2- and ephrinB1-expressing cells have strong heterotypic and weak homotypic repulsion. EphB2 cells have a transient increase in migration after heterotypic activation, which underlies a shift in the EphB2–ephrinB1 border but is not required for segregation or border sharpening. Simulations with the measured values of cell behaviour reveal that heterotypic repulsion can account for cell segregation and border sharpening, and is more efficient than decreased heterotypic adhesion. By suppressing homotypic repulsion, N-cadherin creates a sufficient difference between heterotypic and homotypic repulsion, and enables homotypic cohesion, both of which are required to sharpen borders.
Collapse
Affiliation(s)
- Harriet B Taylor
- Neural Development Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.,Previously at MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Anaïs Khuong
- Neural Development Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.,Computational Cell and Molecular Biology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.,Previously at MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Zhonglin Wu
- Neural Development Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.,Previously at MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Qiling Xu
- Neural Development Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.,Previously at MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Rosalind Morley
- Previously at MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Lauren Gregory
- Previously at MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Alexei Poliakov
- Previously at MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - William R Taylor
- Computational Cell and Molecular Biology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK .,Previously at MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - David G Wilkinson
- Neural Development Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK .,Previously at MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| |
Collapse
|
33
|
The EphA2 receptor is activated through induction of distinct, ligand-dependent oligomeric structures. Commun Biol 2018; 1:15. [PMID: 30271902 PMCID: PMC6123813 DOI: 10.1038/s42003-018-0017-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/28/2018] [Indexed: 01/19/2023] Open
Abstract
The EphA2 receptor tyrosine kinase is capable of activating multiple diverse signaling pathways with roles in processes such as tissue homeostasis and cancer. EphA2 is known to form activated oligomers in the presence of ephrin-A ligands. Here, we characterize the lateral interactions between full-length EphA2 molecules in the plasma membrane in the presence of three types of ligands (dimeric ephrinA1-Fc, monomeric ephrinA1, and an engineered peptide ligand) as well as in the absence of ligand, using a quantitative FRET technique. The data show that EphA2 forms higher-order oligomers and two different types of dimers that all lead to increased EphA2 tyrosine phosphorylation, which is indicative of increased kinase-dependent signaling. We find that different ligands stabilize conformationally distinct oligomers that are assembled through two different interfaces. Our results suggest that these different oligomeric assemblies could have distinct signaling properties, contributing to the diverse activities of the EphA2 receptor. Deo Singh et al. use Fully Quantified Spectral Imaging-FRET to show that the EphA2 receptor forms dimers or higher order oligomers depending on the type of ligand, and that different ligands stabilize EphA2 dimers through distinct interfaces. These findings may explain how EphA2 activates diverse signaling pathways.
Collapse
|
34
|
Locke C, Machida K, Tucker CL, Wu Y, Yu J. Optogenetic activation of EphB2 receptor in dendrites induced actin polymerization by activating Arg kinase. Biol Open 2017; 6:1820-1830. [PMID: 29158322 PMCID: PMC5769660 DOI: 10.1242/bio.029900] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Erythropoietin-producing hepatocellular (Eph) receptors regulate a wide array of developmental processes by responding to cell-cell contacts. EphB2 is well-expressed in the brain and known to be important for dendritic spine development, as well as for the maintenance of the synapses, although the mechanisms of these functions have not been fully understood. Here we studied EphB2's functions in hippocampal neurons with an optogenetic approach, which allowed us to specify spatial regions of signal activation and monitor in real-time the consequences of signal activation. We designed and constructed OptoEphB2, a genetically encoded photoactivatable EphB2. Photoactivation of OptoEphB2 in fibroblast cells induced receptor phosphorylation and resulted in cell rounding ------- a well-known cellular response to EphB2 activation. In contrast, local activation of OptoEphb2 in dendrites of hippocampal neurons induces rapid actin polymerization, resulting dynamic dendritic filopodial growth. Inhibition of Rac1 and CDC42 did not abolish OptoEphB2-induced actin polymerization. Instead, we identified Abelson tyrosine-protein kinase 2 (Abl2/Arg) as a necessary effector in OptoEphB2-induced filopodia growth in dendrites. These findings provided new mechanistic insight into EphB2's role in neural development and demonstrated the advantage of OptoEphB as a new tool for studying EphB signaling.
Collapse
Affiliation(s)
- Clifford Locke
- Richard D. Berlin Center for Cell Analysis and Modeling, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Kazuya Machida
- Richard D. Berlin Center for Cell Analysis and Modeling, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | - Yi Wu
- Richard D. Berlin Center for Cell Analysis and Modeling, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Ji Yu
- Richard D. Berlin Center for Cell Analysis and Modeling, University of Connecticut Health Center, Farmington, CT 06030, USA
| |
Collapse
|
35
|
Ligand-dependent EphA7 signaling inhibits prostate tumor growth and progression. Cell Death Dis 2017; 8:e3122. [PMID: 29022918 PMCID: PMC5682672 DOI: 10.1038/cddis.2017.507] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 08/08/2017] [Accepted: 08/17/2017] [Indexed: 01/02/2023]
Abstract
The downregulation of receptor tyrosine kinase EphA7 is frequent in epithelial cancers and linked to tumor progression. However, the detailed mechanism of EphA7-mediated prostate tumor progression remains elusive. To test the role of EphA7 receptor in prostate cancer (PCa) progression directly, we generated EphA7 receptor variants that were either lacking the cytoplasmic domain or carrying a point mutation that inhibits its phosphorylation by site-directed mutagenesis. Overexpression of wild-type (WT) EphA7 in PCa cells resulted in decreased tumor volume and increased tumor apoptosis in primary tumors. In addition, ectopic expression of WT EphA7 both can delay PCa cell proliferation and could inhibit PCa cell migration and invasion. This protein can also induce PCa cell apoptosis that correlated with increasing the protein expression levels of Bax, elevating the caspase-3 activities, reducing the protein expression levels of Bcl-2 and facilitating the dephosphorylation of Akt, which is further increased by the stimulation of ephrinA5-Fc. However, expression of these EphA7 mutants in PCa cells has no effect in vivo and in vitro. The expression of EphA7 and ephrinA5 was significantly decreased in PCa specimens compared with BPH tissues or paired normal tissues. Moreover, the phosphorylation of EphA7 was positively related with ephrinA5 expression in human prostate tissues. In sum, receptor phosphorylation of EphA7, at least in part, suppress PCa tumor malignancy through targeting PI3K/Akt signaling pathways.
Collapse
|
36
|
Pegg CL, Cooper LT, Zhao J, Gerometta M, Smith FM, Yeh M, Bartlett PF, Gorman JJ, Boyd AW. Glycoengineering of EphA4 Fc leads to a unique, long-acting and broad spectrum, Eph receptor therapeutic antagonist. Sci Rep 2017; 7:6519. [PMID: 28747680 PMCID: PMC5529513 DOI: 10.1038/s41598-017-06685-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/15/2017] [Indexed: 11/09/2022] Open
Abstract
Eph receptors have emerged as targets for therapy in both neoplastic and non-neoplastic disease, however, particularly in non-neoplastic diseases, redundancy of function limits the effectiveness of targeting individual Eph proteins. We have shown previously that a soluble fusion protein, where the EphA4 ectodomain was fused to IgG Fc (EphA4 Fc), was an effective therapy in acute injuries and demonstrated that EphA4 Fc was a broad spectrum Eph/ephrin antagonist. However, a very short in vivo half-life effectively limited its therapeutic development. We report a unique glycoengineering approach to enhance the half-life of EphA4 Fc. Progressive deletion of three demonstrated N-linked sites in EphA4 progressively increased in vivo half-life such that the triple mutant protein showed dramatically improved pharmacokinetic characteristics. Importantly, protein stability, affinity for ephrin ligands and antagonism of cell expressed EphA4 was fully preserved, enabling it to be developed as a broad spectrum Eph/ephrin antagonist for use in both acute and chronic diseases.
Collapse
Affiliation(s)
- Cassandra L Pegg
- Protein Discovery Centre, QIMR Berghofer Medical Research Institute, Queensland, 4006, Australia.
- School of Chemistry and Molecular Biosciences, University of Queensland, Queensland, 4072, Australia.
| | - Leanne T Cooper
- Leukaemia Foundation Research Laboratory, QIMR Berghofer Medical Research Institute, Queensland, 4006, Australia
| | - Jing Zhao
- Queensland Brain Institute, University of Queensland, Queensland, 4072, Australia
| | - Michael Gerometta
- Queensland Brain Institute, University of Queensland, Queensland, 4072, Australia
| | - Fiona M Smith
- Leukaemia Foundation Research Laboratory, QIMR Berghofer Medical Research Institute, Queensland, 4006, Australia
| | - Michael Yeh
- The Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Queensland, 4006, Australia
| | - Perry F Bartlett
- Queensland Brain Institute, University of Queensland, Queensland, 4072, Australia
| | - Jeffrey J Gorman
- Protein Discovery Centre, QIMR Berghofer Medical Research Institute, Queensland, 4006, Australia
- School of Chemistry and Molecular Biosciences, University of Queensland, Queensland, 4072, Australia
| | - Andrew W Boyd
- Leukaemia Foundation Research Laboratory, QIMR Berghofer Medical Research Institute, Queensland, 4006, Australia
- Faculty of Medicine and Biomedical Sciences, University of Queensland, Queensland, 4006, Australia
| |
Collapse
|
37
|
Barquilla A, Lamberto I, Noberini R, Heynen-Genel S, Brill LM, Pasquale EB. Protein kinase A can block EphA2 receptor-mediated cell repulsion by increasing EphA2 S897 phosphorylation. Mol Biol Cell 2016; 27:2757-70. [PMID: 27385333 PMCID: PMC5007095 DOI: 10.1091/mbc.e16-01-0048] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 06/24/2016] [Indexed: 12/18/2022] Open
Abstract
The EphA2 receptor plays multiple roles in cancer through two distinct signaling mechanisms. In a novel cross-talk, the β2-adrenoceptor/cAMP/PKA axis can promote EphA2 pro-oncogenic, ligand-independent signaling, blocking cell repulsion induced by ligand-dependent signaling. PKA emerges as a third kinase, besides AKT and RSK, that can regulate EphA2. The EphA2 receptor tyrosine kinase plays key roles in tissue homeostasis and disease processes such as cancer, pathological angiogenesis, and inflammation through two distinct signaling mechanisms. EphA2 “canonical” signaling involves ephrin-A ligand binding, tyrosine autophosphorylation, and kinase activity; EphA2 “noncanonical” signaling involves phosphorylation of serine 897 (S897) by AKT and RSK kinases. To identify small molecules counteracting EphA2 canonical signaling, we developed a high-content screening platform measuring inhibition of ephrin-A1–induced PC3 prostate cancer cell retraction. Surprisingly, most hits from a screened collection of pharmacologically active compounds are agents that elevate intracellular cAMP by activating G protein–coupled receptors such as the β2-adrenoceptor. We found that cAMP promotes phosphorylation of S897 by protein kinase A (PKA) as well as increases the phosphorylation of several nearby serine/threonine residues, which constitute a phosphorylation hotspot. Whereas EphA2 canonical and noncanonical signaling have been viewed as mutually exclusive, we show that S897 phosphorylation by PKA can coexist with EphA2 tyrosine phosphorylation and block cell retraction induced by EphA2 kinase activity. Our findings reveal a novel paradigm in EphA2 function involving the interplay of canonical and noncanonical signaling and highlight the ability of the β2-adrenoceptor/cAMP/PKA axis to rewire EphA2 signaling in a subset of cancer cells.
Collapse
Affiliation(s)
- Antonio Barquilla
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037
| | - Ilaria Lamberto
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037
| | - Roberta Noberini
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037
| | - Susanne Heynen-Genel
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037
| | - Laurence M Brill
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037
| | - Elena B Pasquale
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037 Pathology Department, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
38
|
Stark DA, Coffey NJ, Pancoast HR, Arnold LL, Walker JPD, Vallée J, Robitaille R, Garcia ML, Cornelison DDW. Ephrin-A3 promotes and maintains slow muscle fiber identity during postnatal development and reinnervation. J Cell Biol 2015; 211:1077-91. [PMID: 26644518 PMCID: PMC4674275 DOI: 10.1083/jcb.201502036] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 10/22/2015] [Indexed: 11/28/2022] Open
Abstract
Each adult mammalian skeletal muscle has a unique complement of fast and slow myofibers, reflecting patterns established during development and reinforced via their innervation by fast and slow motor neurons. Existing data support a model of postnatal "matching" whereby predetermined myofiber type identity promotes pruning of inappropriate motor axons, but no molecular mechanism has yet been identified. We present evidence that fiber type-specific repulsive interactions inhibit innervation of slow myofibers by fast motor axons during both postnatal maturation of the neuromuscular junction and myofiber reinnervation after injury. The repulsive guidance ligand ephrin-A3 is expressed only on slow myofibers, whereas its candidate receptor, EphA8, localizes exclusively to fast motor endplates. Adult mice lacking ephrin-A3 have dramatically fewer slow myofibers in fast and mixed muscles, and misexpression of ephrin-A3 on fast myofibers followed by denervation/reinnervation promotes their respecification to a slow phenotype. We therefore conclude that Eph/ephrin interactions guide the fiber type specificity of neuromuscular interactions during development and adult life.
Collapse
Affiliation(s)
- Danny A Stark
- Division of Biological Sciences, University of Missouri, Columbia, MO 65211 Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211
| | - Nathan J Coffey
- Division of Biological Sciences, University of Missouri, Columbia, MO 65211
| | - Hannah R Pancoast
- Division of Biological Sciences, University of Missouri, Columbia, MO 65211
| | - Laura L Arnold
- Division of Biological Sciences, University of Missouri, Columbia, MO 65211 Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211
| | - J Peyton D Walker
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211
| | - Joanne Vallée
- Département de Neurosciences, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Richard Robitaille
- Département de Neurosciences, Université de Montréal, Montréal, Québec H3C 3J7, Canada Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Michael L Garcia
- Division of Biological Sciences, University of Missouri, Columbia, MO 65211 Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211
| | - D D W Cornelison
- Division of Biological Sciences, University of Missouri, Columbia, MO 65211 Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211
| |
Collapse
|
39
|
Li S, Wu Z, Chen Y, Kang Z, Wang H, He P, Zhang X, Hu T, Zhang Q, Cai Y, Xu X, Guan M. Diagnostic and prognostic value of tissue and circulating levels of Ephrin-A2 in prostate cancer. Tumour Biol 2015; 37:5365-74. [PMID: 26561474 DOI: 10.1007/s13277-015-4398-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/05/2015] [Indexed: 10/22/2022] Open
Abstract
Ephrin-A2, a member of the Eph/ephrin family, is associated with tumorigenesis and tumor progression. This study aimed to assess the diagnostic and prognostic value of both serum and tissue levels of Ephrin-A2 in prostate cancer (PCa) management. One hundred and forty-five frozen prostate tissues, 55 paraffin-embedded prostate tissues, 88 serum samples, and seven prostate cell lines (RWPE-1, LNCaP, LNCaP-LN3, PC-3, PC-3M, PC-3M-LN4, and DU145) were examined via quantitative reverse transcription-PCR (qRT-PCR), immunohistochemistry, enzyme-linked immunosorbent assay, and western blotting. Induced Ephrin-A2 messenger RNA (mRNA) or protein expression was detected in 8.6 % (5/58) benign prostatic hyperplasia (BPH), 59.8 % (52/87) PCa, and five prostate cancer cell lines. Ephrin-A2 immunostaining was present in 6.7 % (1/15) patients with BPHs and 62.5 % (25/40) clinically localized PCa. Accordingly, serum Ephrin-A2 was significantly higher in PCa patients compared to those in the BPH patients and controls (P < 0.001). The expression of Ephrin-A2 was higher in tumor patients with an elevated Gleason score or T3-T4 staging. Ephrin-A2 expression was correlated with Ki-67 expression in PCa patients, both at the gene scale and protein level. Our data indicate that Ephrin-A2 is a potential diagnostic and prognostic biomarker and a promising molecular therapeutic target to attenuate prostate cancer progression.
Collapse
Affiliation(s)
- Shibao Li
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical School, Fudan University, 12 Central Urumqi Road, Shanghai, China.,Department of Laboratory Medicine, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Zhiyuan Wu
- Department of Laboratory Medicine, Huashan Hospital North, Fudan University, Shanghai, China
| | - Yuming Chen
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical School, Fudan University, 12 Central Urumqi Road, Shanghai, China
| | - Zhihua Kang
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical School, Fudan University, 12 Central Urumqi Road, Shanghai, China
| | - Hua Wang
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical School, Fudan University, 12 Central Urumqi Road, Shanghai, China
| | - Ping He
- Department of Laboratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Xinju Zhang
- Central Laboratory, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China
| | - Tingting Hu
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical School, Fudan University, 12 Central Urumqi Road, Shanghai, China
| | - Qunfeng Zhang
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical School, Fudan University, 12 Central Urumqi Road, Shanghai, China.,Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Yanqun Cai
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical School, Fudan University, 12 Central Urumqi Road, Shanghai, China.,Department of Laboratory Medicine, Taizhou Municipal Hospital, Taizhou, Zhejiang, China
| | - Xiao Xu
- Central Laboratory, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China
| | - Ming Guan
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical School, Fudan University, 12 Central Urumqi Road, Shanghai, China. .,Department of Laboratory Medicine, Huashan Hospital North, Fudan University, Shanghai, China. .,Central Laboratory, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China.
| |
Collapse
|
40
|
Δ(5)-Cholenoyl-amino acids as selective and orally available antagonists of the Eph-ephrin system. Eur J Med Chem 2015; 103:312-24. [PMID: 26363867 DOI: 10.1016/j.ejmech.2015.08.048] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 08/24/2015] [Accepted: 08/25/2015] [Indexed: 01/22/2023]
Abstract
The Eph receptor-ephrin system is an emerging target for the development of novel anti-angiogenic therapies. Research programs aimed at developing small-molecule antagonists of the Eph receptors are still in their initial stage as available compounds suffer from pharmacological drawbacks, limiting their application in vitro and in vivo. In the present work, we report the design, synthesis and evaluation of structure-activity relationships of a class of Δ(5)-cholenoyl-amino acid conjugates as Eph-ephrin antagonists. As a major achievement of our exploration, we identified N-(3β-hydroxy-Δ(5)-cholen-24-oyl)-L-tryptophan (UniPR1331) as the first small molecule antagonist of the Eph-ephrin system effective as an anti-angiogenic agent in endothelial cells, bioavailable in mice by the oral route and devoid of biological activity on G protein-coupled and nuclear receptors targeted by bile acid derivatives.
Collapse
|
41
|
Abstract
Eph:ephrin signaling plays an important role in embryonic development as well as tissue homeostasis in the adult. At the cellular level, this transduction pathway is best known for its role in the control of cell adhesion and repulsion, cell migration and morphogenesis. Yet, a number of publications have also implicated Eph:ephrin signaling in the control of adult and embryonic neurogenesis. As is the case for other biological processes, these studies have reported conflicting and sometimes opposite roles for Eph:ephrin signaling in neurogenesis. Herein, we review these studies and we discuss existing mathematical models of stem cell dynamics and neurogenesis that provide a coherent framework and may help reconcile conflicting results.
Collapse
Affiliation(s)
- J Laussu
- a Centre de Biologie du Développement; CNRS; Université de Toulouse ; Toulouse , France
| | | | | | | |
Collapse
|
42
|
Szepietowska B, Horvath TL, Sherwin RS. Role of synaptic plasticity and EphA5-ephrinA5 interaction within the ventromedial hypothalamus in response to recurrent hypoglycemia. Diabetes 2014; 63:1140-7. [PMID: 24222347 PMCID: PMC3931406 DOI: 10.2337/db13-1259] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hypoglycemia stimulates counterregulatory hormone release to restore euglycemia. This protective response is diminished by recurrent hypoglycemia, limiting the benefits of intensive insulin treatment in patients with diabetes. We previously reported that EphA5 receptor-ephrinA5 interactions within the ventromedial hypothalamus (VMH) influence counterregulatory hormone responses during acute hypoglycemia in nondiabetic rats. In this study, we examined whether recurrent hypoglycemia alters the capacity of the ephrinA5 ligand to activate VMH EphA5 receptors, and if so, whether these changes could contribute to pathogenesis of defective glucose counterregulation in response to a standard hypoglycemic stimulus. The expression of ephrinA5, but not EphA5 receptors within the VMH, was reduced by antecedent recurrent hypoglycemia. In addition, the number of synaptic connections was increased and astroglial synaptic coverage was reduced. Activation of VMH EphA5 receptors via targeted microinjection of ephrinA5-Fc before a hyperinsulinemic hypoglycemic clamp study caused a reduction in the glucose infusion rate in nondiabetic rats exposed to recurrent hypoglycemia. The increase in the counterregulatory response to insulin-induced hypoglycemia was associated with a 150% increase in glucagon release (P < 0.001). These data suggest that changes in ephrinA5/EphA5 interactions and synaptic plasticity within the VMH, a key glucose-sensing region in the brain, may contribute to the impairment in glucagon secretion and counterregulatory responses caused by recurrent hypoglycemia.
Collapse
Affiliation(s)
- Barbara Szepietowska
- Yale University School of Medicine, Department of Internal Medicine and Endocrinology, New Haven, CT
| | - Tamas L. Horvath
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT
| | - Robert S. Sherwin
- Yale University School of Medicine, Department of Internal Medicine and Endocrinology, New Haven, CT
- Corresponding author: Robert S. Sherwin,
| |
Collapse
|
43
|
Fagotto F, Winklbauer R, Rohani N. Ephrin-Eph signaling in embryonic tissue separation. Cell Adh Migr 2014; 8:308-26. [PMID: 25482630 PMCID: PMC4594459 DOI: 10.4161/19336918.2014.970028] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 08/19/2014] [Accepted: 08/25/2014] [Indexed: 01/19/2023] Open
Abstract
The physical separation of the embryonic regions that give rise to the tissues and organs of multicellular organisms is a fundamental aspect of morphogenesis. Pioneer experiments by Holtfreter had shown that embryonic cells can sort based on "tissue affinities," which have long been considered to rely on differences in cell-cell adhesion. However, vertebrate embryonic tissues also express a variety of cell surface cues, in particular ephrins and Eph receptors, and there is now firm evidence that these molecules are systematically used to induce local repulsion at contacts between different cell types, efficiently preventing mixing of adjacent cell populations.
Collapse
Affiliation(s)
| | - Rudolf Winklbauer
- Dpt. of Cell and Systems Biology; University of Toronto; Toronto, Canada
| | - Nazanin Rohani
- Dpt. of Biology; McGill University; Montreal, Quebec, Canada
| |
Collapse
|
44
|
Falivelli G, Lisabeth EM, de la Torre ER, Perez-Tenorio G, Tosato G, Salvucci O, Pasquale EB. Attenuation of eph receptor kinase activation in cancer cells by coexpressed ephrin ligands. PLoS One 2013; 8:e81445. [PMID: 24348920 PMCID: PMC3857839 DOI: 10.1371/journal.pone.0081445] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 10/21/2013] [Indexed: 12/20/2022] Open
Abstract
The Eph receptor tyrosine kinases mediate juxtacrine signals by interacting “in trans” with ligands anchored to the surface of neighboring cells via a GPI-anchor (ephrin-As) or a transmembrane segment (ephrin-Bs), which leads to receptor clustering and increased kinase activity. Additionally, soluble forms of the ephrin-A ligands released from the cell surface by matrix metalloproteases can also activate EphA receptor signaling. Besides these trans interactions, recent studies have revealed that Eph receptors and ephrins coexpressed in neurons can also engage in lateral “cis” associations that attenuate receptor activation by ephrins in trans with critical functional consequences. Despite the importance of the Eph/ephrin system in tumorigenesis, Eph receptor-ephrin cis interactions have not been previously investigated in cancer cells. Here we show that in cancer cells, coexpressed ephrin-A3 can inhibit the ability of EphA2 and EphA3 to bind ephrins in trans and become activated, while ephrin-B2 can inhibit not only EphB4 but also EphA3. The cis inhibition of EphA3 by ephrin-B2 implies that in some cases ephrins that cannot activate a particular Eph receptor in trans can nevertheless inhibit its signaling ability through cis association. We also found that an EphA3 mutation identified in lung cancer enhances cis interaction with ephrin-A3. These results suggest a novel mechanism that may contribute to cancer pathogenesis by attenuating the tumor suppressing effects of Eph receptor signaling pathways activated by ephrins in trans.
Collapse
Affiliation(s)
- Giulia Falivelli
- Sanford-Burnham Medical Research Institute, San Diego, California, United States of America
- Department Pharmacology, University of Bologna, Bologna, Italy
| | - Erika Mathes Lisabeth
- Sanford-Burnham Medical Research Institute, San Diego, California, United States of America
| | | | - Gizeh Perez-Tenorio
- Sanford-Burnham Medical Research Institute, San Diego, California, United States of America
| | - Giovanna Tosato
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ombretta Salvucci
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Elena B. Pasquale
- Sanford-Burnham Medical Research Institute, San Diego, California, United States of America
- Department of Pathology, University of California San Diego, San Diego, California, United States of America
- * E-mail:
| |
Collapse
|
45
|
Russo S, Incerti M, Tognolini M, Castelli R, Pala D, Hassan-Mohamed I, Giorgio C, De Franco F, Gioiello A, Vicini P, Barocelli E, Rivara S, Mor M, Lodola A. Synthesis and structure-activity relationships of amino acid conjugates of cholanic acid as antagonists of the EphA2 receptor. Molecules 2013; 18:13043-60. [PMID: 24152675 PMCID: PMC6270184 DOI: 10.3390/molecules181013043] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 10/11/2013] [Accepted: 10/12/2013] [Indexed: 11/16/2022] Open
Abstract
The Eph–ephrin system plays a critical role in tumor growth and vascular functions during carcinogenesis. We had previously identified cholanic acid as a competitive and reversible EphA2 antagonist able to disrupt EphA2-ephrinA1 interaction and to inhibit EphA2 activation in prostate cancer cells. Herein, we report the synthesis and biological evaluation of a set of cholanic acid derivatives obtained by conjugation of its carboxyl group with a panel of naturally occurring amino acids with the aim to improve EphA2 receptor inhibition. Structure-activity relationships indicate that conjugation of cholanic acid with linear amino acids of small size leads to effective EphA2 antagonists whereas the introduction of aromatic amino acids reduces the potency in displacement studies. The β-alanine derivative 4 was able to disrupt EphA2-ephrinA1 interaction in the micromolar range and to dose-dependently inhibit EphA2 activation on PC3 cells. These findings may help the design of novel EphA2 antagonists active on cancer cell lines.
Collapse
Affiliation(s)
- Simonetta Russo
- Dipartimento di Farmacia, Università degli Studi di Parma, Viale delle Scienze 27/A, Parma I-43124, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Tognolini M, Incerti M, Pala D, Russo S, Castelli R, Hassan-Mohamed I, Giorgio C, Lodola A. Target hopping as a useful tool for the identification of novel EphA2 protein-protein antagonists. ChemMedChem 2013; 9:67-72. [PMID: 24115725 DOI: 10.1002/cmdc.201300305] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Indexed: 11/08/2022]
Abstract
Lithocholic acid (LCA), a physiological ligand for the nuclear receptor FXR and the G-protein-coupled receptor TGR5, has been recently described as an antagonist of the EphA2 receptor, a key member of the ephrin signalling system involved in tumour growth. Given the ability of LCA to recognize FXR, TGR5, and EphA2 receptors, we hypothesized that the structural requirements for a small molecule to bind each of these receptors might be similar. We therefore selected a set of commercially available FXR or TGR5 ligands and tested them for their ability to inhibit EphA2 by targeting the EphA2-ephrin-A1 interface. Among the selected compounds, the stilbene carboxylic acid GW4064 was identified as an effective antagonist of EphA2, being able to block EphA2 activation in prostate carcinoma cells, in the micromolar range. This finding proposes the "target hopping" approach as a new effective strategy to discover new protein-protein interaction inhibitors.
Collapse
Affiliation(s)
- Massimiliano Tognolini
- Dipartimento di Farmacia, Università degli Studi di Parma, V. le delle Scienze 27 A, 43124 Parma (Italy)
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Eph:ephrin-B1 forward signaling controls fasciculation of sensory and motor axons. Dev Biol 2013; 383:264-74. [PMID: 24056079 DOI: 10.1016/j.ydbio.2013.09.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 09/04/2013] [Accepted: 09/07/2013] [Indexed: 11/22/2022]
Abstract
Axon fasciculation is one of the processes controlling topographic innervation during embryonic development. While axon guidance steers extending axons in the accurate direction, axon fasciculation allows sets of co-extending axons to grow in tight bundles. The Eph:ephrin family has been involved both in axon guidance and fasciculation, yet it remains unclear how these two distinct types of responses are elicited. Herein we have characterized the role of ephrin-B1, a member of the ephrinB family in sensory and motor innervation of the limb. We show that ephrin-B1 is expressed in sensory axons and in the limb bud mesenchyme while EphB2 is expressed in motor and sensory axons. Loss of ephrin-B1 had no impact on the accurate dorso-ventral innervation of the limb by motor axons, yet EfnB1 mutants exhibited decreased fasciculation of peripheral motor and sensory nerves. Using tissue-specific excision of EfnB1 and in vitro experiments, we demonstrate that ephrin-B1 controls fasciculation of axons via a surround repulsion mechanism involving growth cone collapse of EphB2-expressing axons. Altogether, our results highlight the complex role of Eph:ephrin signaling in the development of the sensory-motor circuit innervating the limb.
Collapse
|
48
|
Bell MR, Engleka MJ, Malik A, Strickler JE. To fuse or not to fuse: what is your purpose? Protein Sci 2013; 22:1466-77. [PMID: 24038604 DOI: 10.1002/pro.2356] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 08/19/2013] [Accepted: 08/20/2013] [Indexed: 01/13/2023]
Abstract
Since the dawn of time, or at least the dawn of recombinant DNA technology (which for many of today's scientists is the same thing), investigators have been cloning and expressing heterologous proteins in a variety of different cells for a variety of different reasons. These range from cell biological studies looking at protein-protein interactions, post-translational modifications, and regulation, to laboratory-scale production in support of biochemical, biophysical, and structural studies, to large scale production of potential biotherapeutics. In parallel, fusion-tag technology has grown-up to facilitate microscale purification (pull-downs), protein visualization (epitope tags), enhanced expression and solubility (protein partners, e.g., GST, MBP, TRX, and SUMO), and generic purification (e.g., His-tags, streptag, and FLAG™-tag). Frequently, these latter two goals are combined in a single fusion partner. In this review, we examine the most commonly used fusion methodologies from the perspective of the ultimate use of the tagged protein. That is, what are the most commonly used fusion partners for pull-downs, for structural studies, for production of active proteins, or for large-scale purification? What are the advantages and limitations of each? This review is not meant to be exhaustive and the approach undoubtedly reflects the experiences and interests of the authors. For the sake of brevity, we have largely ignored epitope tags although they receive wide use in cell biology for immunopreciptation.
Collapse
Affiliation(s)
- Mark R Bell
- LifeSensors, Inc., Malvern, Pennsylvania, 19083
| | | | | | | |
Collapse
|
49
|
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: 314] [Impact Index Per Article: 26.2] [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
|
50
|
Seiradake E, Schaupp A, del Toro Ruiz D, Kaufmann R, Mitakidis N, Harlos K, Aricescu AR, Klein R, Jones EY. Structurally encoded intraclass differences in EphA clusters drive distinct cell responses. Nat Struct Mol Biol 2013; 20:958-64. [PMID: 23812375 PMCID: PMC3941021 DOI: 10.1038/nsmb.2617] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 05/08/2013] [Indexed: 02/06/2023]
Abstract
The functional outcomes of ephrin binding to Eph receptors (Ephs) range from cell repulsion to adhesion. Here we used cell collapse and stripe assays to show contrasting effects of human ephrinA5 binding to EphA2 and EphA4. Despite equivalent ligand-binding affinities EphA4 triggered greater cell collapse, while EphA2-expressing cells adhered better to ephrinA5-coated surfaces. Chimeric receptors showed the ectodomain is a major determinant of cell response. We report crystal structures of EphA4 ectodomain alone and in complexes with ephrinB3 and ephrinA5. These revealed closed clusters with a dimeric or circular arrangement in the crystal lattice, contrasting with extended arrays previously observed for EphA2 ectodomain. Localization microscopy-based analyses showed ligand-stimulated EphA4 induces smaller clusters than EphA2. Mutant Ephs link these characteristics to interactions observed in the crystal lattices, suggesting a mechanism by which distinctive ectodomain surfaces determine clustering, and thereby signalling, properties.
Collapse
Affiliation(s)
- Elena Seiradake
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | | | | | | | | | | | | | | |
Collapse
|