101
|
Seif F, Little PJ, Niayesh-Mehr R, Zamanpour M, Babaahmadi-Rezaei H. Endothelin-1 increases CHSY-1 expression in aortic endothelial cells via transactivation of transforming growth factor β type I receptor induced by type B receptor endothelin-1. J Pharm Pharmacol 2019; 71:988-995. [DOI: 10.1111/jphp.13081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 01/12/2019] [Indexed: 12/29/2022]
Abstract
Abstract
Objectives
TGF-β through hyperelongation of glycosaminoglycan (GAG) chains leads to binding of low-density lipoproteins to the proteoglycans. The vasoactive peptide, endothelin-1 (ET-1), plays a key role in the development of atherosclerosis. This study addressed the question whether ET-1 by activating the Rho kinase and cytoskeletal rearrangement can transactivate the TGF-β receptor leading to phosphorylation of the transcription factor Smad2 and increased expression of the GAG chain synthesizing enzyme such as chondroitin synthase-1 (CHSY-1) in bovine aortic endothelial cells (BAECs).
Methods
In this study, intermediates in ET-1-induced Smad2C phosphorylation and the protein level of CHSY-1 were identified and quantified by Western blotting.
Key findings
Endothelin-1 caused time-dependent phosphorylation of Smad2C which was inhibited in the presence of the endothelin B receptor antagonist, BQ788. The response to ET-1 was inhibited by the Rho/ROCK kinase antagonist, Y27632 and by cytochalasin D, an inhibitor of actin polymerization but the ET-1-mediated pSmad2C was not inhibited by the matrix metalloproteinase (MMP) inhibitor, GM6001. ET-1 increased CHSY-1 protein level, which was inhibited in the presence of BQ788, cytochalasin D and Y27632.
Conclusions
Endothelin-1 signalling via the ETB receptor utilizes cytoskeletal rearrangement and Rho kinase but not MMPs leading to TβRI transactivation signalling and phosphorylation of Smad2C and through this pathway increased the level of CHSY-1.
Collapse
Affiliation(s)
- Faezeh Seif
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Qld, Australia
- Department of Pharmacy, Xinhua College of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Reyhaneh Niayesh-Mehr
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masoumeh Zamanpour
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Babaahmadi-Rezaei
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
102
|
Clarke DT, Martin-Fernandez ML. A Brief History of Single-Particle Tracking of the Epidermal Growth Factor Receptor. Methods Protoc 2019; 2:mps2010012. [PMID: 31164594 PMCID: PMC6481046 DOI: 10.3390/mps2010012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/21/2019] [Accepted: 01/21/2019] [Indexed: 12/15/2022] Open
Abstract
Single-particle tracking (SPT) has been used and developed over the last 25 years as a method to investigate molecular dynamics, structure, interactions, and function in the cellular context. SPT is able to show how fast and how far individual molecules move, identify different dynamic populations, measure the duration and strength of intermolecular interactions, and map out structures on the nanoscale in cells. In combination with other techniques such as macromolecular crystallography and molecular dynamics simulation, it allows us to build models of complex structures, and develop and test hypotheses of how these complexes perform their biological roles in health as well as in disease states. Here, we use the example of the epidermal growth factor receptor (EGFR), which has been studied extensively by SPT, demonstrating how the method has been used to increase our understanding of the receptor’s organization and function, including its interaction with the plasma membrane, its activation, clustering, and oligomerization, and the role of other receptors and endocytosis. The examples shown demonstrate how SPT might be employed in the investigation of other biomolecules and systems.
Collapse
Affiliation(s)
- David T Clarke
- STFC Central Laser Facility, Research Complex at Harwell, Rutherford Appleton Laboratory, Didcot OX11 0QX, UK.
| | - Marisa L Martin-Fernandez
- STFC Central Laser Facility, Research Complex at Harwell, Rutherford Appleton Laboratory, Didcot OX11 0QX, UK.
| |
Collapse
|
103
|
Gebert-Oberle B, Giles J, Clayton S, Tran QK. Calcium/calmodulin regulates signaling at the α 1A adrenoceptor. Eur J Pharmacol 2019; 848:70-79. [PMID: 30690001 DOI: 10.1016/j.ejphar.2019.01.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 11/16/2022]
Abstract
Cardiovascular functions are mediated by multiple 7-pass transmembrane receptors whose activation promotes contraction or relaxation of the tissues. The α1 adrenoceptor type 1A plays important roles in the control of vascular tone and myocardial contractility via Ca2+-dependent actions. Here, using novel FRET-based biosensors, we identified a novel Ca2+-dependent interaction between calmodulin (CaM) and the human α1A adrenoceptor at the juxtamembranous region of its 4th submembrane domain (SMD4JM, a.a. 333-361). SMD4JM houses the known nuclear localization signal of α1A adrenoceptor (NLS, a.a. 334-349). We found that NLS itself also interacts with CaM, but with lower affinity and Ca2+ sensitivity, indicating that full interaction between CaM and α1A receptor in this region requires segment a.a. 333-361. Combined K353Q/L356A substitutions in the non-NLS segment of SMD4JM cause a 3.5-fold reduction in the affinity of CaM-SMD4JM interaction. Overexpression of wild-type α1A adrenoceptor in cells enhances phosphorylation of the extracellular signal-regulated kinases 1/2 (ERK1/2) stimulated by A61603, while overexpression of the K353Q/L356A α1A receptor mutant significantly reduces this signal. Norepinephrine stimulates intracellular Ca2+ signals that are higher in cells overexpressing wild-type receptor but lower in cells overexpressing the K353Q/L356A receptor compared to non-transfected cells in the same microscopic environments. These data support a novel and important role for Ca2+-dependent CaM interaction at SMD4JM in α1A adrenoceptor-mediated signaling.
Collapse
Affiliation(s)
- Briana Gebert-Oberle
- Department of Physiology and Pharmacology, Des Moines University Osteopathic Medical Center, Ryan Hall 258, 3200 Grand Avenue, Des Moines, IA 50312, United States
| | - Jennifer Giles
- Department of Physiology and Pharmacology, Des Moines University Osteopathic Medical Center, Ryan Hall 258, 3200 Grand Avenue, Des Moines, IA 50312, United States
| | - Sarah Clayton
- Department of Physiology and Pharmacology, Des Moines University Osteopathic Medical Center, Ryan Hall 258, 3200 Grand Avenue, Des Moines, IA 50312, United States
| | - Quang-Kim Tran
- Department of Physiology and Pharmacology, Des Moines University Osteopathic Medical Center, Ryan Hall 258, 3200 Grand Avenue, Des Moines, IA 50312, United States.
| |
Collapse
|
104
|
Parra-Mercado GK, Fuentes-Gonzalez AM, Hernandez-Aranda J, Diaz-Coranguez M, Dautzenberg FM, Catt KJ, Hauger RL, Olivares-Reyes JA. CRF 1 Receptor Signaling via the ERK1/2-MAP and Akt Kinase Cascades: Roles of Src, EGF Receptor, and PI3-Kinase Mechanisms. Front Endocrinol (Lausanne) 2019; 10:869. [PMID: 31920979 PMCID: PMC6921279 DOI: 10.3389/fendo.2019.00869] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 11/27/2019] [Indexed: 12/28/2022] Open
Abstract
In the present study, we determined the cellular regulators of ERK1/2 and Akt signaling pathways in response to human CRF1 receptor (CRF1R) activation in transfected COS-7 cells. We found that Pertussis Toxin (PTX) treatment or sequestering Gβγ reduced CRF1R-mediated activation of ERK1/2, suggesting the involvement of a Gi-linked cascade. Neither Gs/PKA nor Gq/PKC were associated with ERK1/2 activation. Besides, CRF induced EGF receptor (EGFR) phosphorylation at Tyr1068, and selective inhibition of EGFR kinase activity by AG1478 strongly inhibited the CRF1R-mediated phosphorylation of ERK1/2, indicating the participation of EGFR transactivation. Furthermore, CRF-induced ERK1/2 phosphorylation was not altered by pretreatment with batimastat, GM6001, or an HB-EGF antibody indicating that metalloproteinase processing of HB-EGF ligands is not required for the CRF-mediated EGFR transactivation. We also observed that CRF induced Src and PYK2 phosphorylation in a Gβγ-dependent manner. Additionally, using the specific Src kinase inhibitor PP2 and the dominant-negative-SrcYF-KM, it was revealed that CRF-stimulated ERK1/2 phosphorylation depends on Src activation. PP2 also blocked the effect of CRF on Src and EGFR (Tyr845) phosphorylation, further demonstrating the centrality of Src. We identified the formation of a protein complex consisting of CRF1R, Src, and EGFR facilitates EGFR transactivation and CRF1R-mediated signaling. CRF stimulated Akt phosphorylation, which was dependent on Gi/βγ subunits, and Src activation, however, was only slightly dependent on EGFR transactivation. Moreover, PI3K inhibitors were able to inhibit not only the CRF-induced phosphorylation of Akt, as expected, but also ERK1/2 activation by CRF suggesting a PI3K dependency in the CRF1R ERK signaling. Finally, CRF-stimulated ERK1/2 activation was similar in the wild-type CRF1R and the phosphorylation-deficient CRF1R-Δ386 mutant, which has impaired agonist-dependent β-arrestin-2 recruitment; however, this situation may have resulted from the low β-arrestin expression in the COS-7 cells. When β-arrestin-2 was overexpressed in COS-7 cells, CRF-stimulated ERK1/2 phosphorylation was markedly upregulated. These findings indicate that on the base of a constitutive CRF1R/EGFR interaction, the Gi/βγ subunits upstream activation of Src, PYK2, PI3K, and transactivation of the EGFR are required for CRF1R signaling via the ERK1/2-MAP kinase pathway. In contrast, Akt activation via CRF1R is mediated by the Src/PI3K pathway with little contribution of EGFR transactivation.
Collapse
Affiliation(s)
- G. Karina Parra-Mercado
- Laboratory of Signal Transduction, Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV-IPN, Mexico City, Mexico
| | - Alma M. Fuentes-Gonzalez
- Laboratory of Signal Transduction, Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV-IPN, Mexico City, Mexico
| | - Judith Hernandez-Aranda
- Laboratory of Signal Transduction, Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV-IPN, Mexico City, Mexico
| | - Monica Diaz-Coranguez
- Laboratory of Signal Transduction, Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV-IPN, Mexico City, Mexico
| | | | - Kevin J. Catt
- Section on Hormonal Regulation, Program on Developmental Endocrinology and Genetics, National Institute of Child Health and Human Development, Bethesda, MD, United States
| | - Richard L. Hauger
- Center of Excellence for Stress and Mental Health, VA Healthcare System, San Diego, CA, United States
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - J. Alberto Olivares-Reyes
- Laboratory of Signal Transduction, Department of Biochemistry, Center for Research and Advanced Studies of the National Polytechnic Institute, CINVESTAV-IPN, Mexico City, Mexico
- *Correspondence: J. Alberto Olivares-Reyes
| |
Collapse
|
105
|
Dai LX, Yang J, Liu JM, Huang S, Wang BN, Li H, Yang J, Zhao ZY, Cao K, Li MY. Adenovirus-Mediated CRM197 Sensitizes Human Glioma Cells to Gemcitabine by the Mitochondrial Pathway. Cancer Biother Radiopharm 2018; 34:171-180. [PMID: 30585767 DOI: 10.1089/cbr.2017.2363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
PURPOSE The cross-reacting material 197 (CRM197) is a mutation of the diphtheria toxin. The protein of CRM197 was used successfully for the therapy of various tumors in the recent studies. In this study, the recombinant adenoviruses containing the CRM197gene(AdCRM197) were used to enhance the cellar toxicity of gemcitabine in human glioma U87, U251, and H4 cells. PROCEDURES MTT assay and flow cytometric analysis were performed to test the apoptosis of the U87, U251 and H4 cells with the combined treatment of AdCRM197 plus gemcitabine. Western blotting analyses were carried out to detect the cell apoptosis of the mitochondrial pathway. And the xenograft nude mice were used to observe the enhanced antitumor effect of AdCRM197 in vivo. RESULTS AdCRM197 sensitizes human glioma cells to gemcitabine in vitro by the mitochondrial pathway. Tumor volume was inhibited and survival time was prolonged in the U251 or U87 xenografted nude mice with gemcitabine plus AdCRM197. The enhanced antitumor effect of AdCRM197 was also detected by the immunohistochemical analyses and TUNEL staining. CONCLUSION The authors found that AdCRM197 sensitized the human glioma to gemcitabine not only in vitro but also in vivo. They provide the first evidence that adenovirus-mediated CRM197 may be a potential chemosensitizing agent for the treatment of cancer. The diphtheria toxin is of great toxicity that even one molecule of diphtheria toxin is enough to kill one cell. However, because of the high toxicity, the diphtheria toxin would kill the packing cells when it is being packaged into the recombinant viruses. Therefore, the diphtheria toxin is hard to be used in the gene therapy for virus vectors. The cross-reacting material 197 (CRM197) is a mutation of the diphtheria toxin. Unlike DTA, CRM197 exhibit a weak toxicity. The week toxicity of CRM197 is a good feature for the virus packaging. In the present study, we used a recombinant adenovirus which carried a CRM197 gene (AdCRM197) to enhance the cellar toxicity of gemcitabine in human glioma cells.
Collapse
Affiliation(s)
- Lv-Xia Dai
- 1 Department of Microbiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University , Chengdu, China .,2 Department of Pathogen Biology, Chengdu Medical College , Chengdu, China
| | - Jing Yang
- 3 Department of Infectious Disease, Renmin Hospital, Hubei University of Medicine , Shiyan, China
| | - Jian-Min Liu
- 4 Department of Neurosurgery, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine , Guangzhou, China
| | - Sizhou Huang
- 2 Department of Pathogen Biology, Chengdu Medical College , Chengdu, China
| | - Bao-Ning Wang
- 1 Department of Microbiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University , Chengdu, China
| | - Hong Li
- 5 West China Second University Hospital, Sichuan University , Chengdu, China
| | - Jie Yang
- 6 Department of Neurology, The First Affiliated Hospital of Chengdu Medical College , Chengdu, China
| | - Zhong-Yi Zhao
- 1 Department of Microbiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University , Chengdu, China
| | - Kang Cao
- 2 Department of Pathogen Biology, Chengdu Medical College , Chengdu, China
| | - Ming-Yuan Li
- 1 Department of Microbiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University , Chengdu, China
| |
Collapse
|
106
|
Role of Epidermal Growth Factor Receptor (EGFR) and Its Ligands in Kidney Inflammation and Damage. Mediators Inflamm 2018; 2018:8739473. [PMID: 30670929 PMCID: PMC6323488 DOI: 10.1155/2018/8739473] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 10/29/2018] [Accepted: 11/07/2018] [Indexed: 12/29/2022] Open
Abstract
Chronic kidney disease (CKD) is characterized by persistent inflammation and progressive fibrosis, ultimately leading to end-stage renal disease. Although many studies have investigated the factors involved in the progressive deterioration of renal function, current therapeutic strategies only delay disease progression, leaving an unmet need for effective therapeutic interventions that target the cause behind the inflammatory process and could slow down or reverse the development and progression of CKD. Epidermal growth factor receptor (EGFR) (ERBB1), a membrane tyrosine kinase receptor expressed in the kidney, is activated after renal damage, and preclinical studies have evidenced its potential as a therapeutic target in CKD therapy. To date, seven official EGFR ligands have been described, including epidermal growth factor (EGF) (canonical ligand), transforming growth factor-α, heparin-binding epidermal growth factor, amphiregulin, betacellulin, epiregulin, and epigen. Recently, the connective tissue growth factor (CTGF/CCN2) has been described as a novel EGFR ligand. The direct activation of EGFR by its ligands can exert different cellular responses, depending on the specific ligand, tissue, and pathological condition. Among all EGFR ligands, CTGF/CCN2 is of special relevance in CKD. This growth factor, by binding to EGFR and downstream signaling pathway activation, regulates renal inflammation, cell growth, and fibrosis. EGFR can also be “transactivated” by extracellular stimuli, including several key factors involved in renal disease, such as angiotensin II, transforming growth factor beta (TGFB), and other cytokines, including members of the tumor necrosis factor superfamily, showing another important mechanism involved in renal pathology. The aim of this review is to summarize the contribution of EGFR pathway activation in experimental kidney damage, with special attention to the regulation of the inflammatory response and the role of some EGFR ligands in this process. Better insights in EGFR signaling in renal disease could improve our current knowledge of renal pathology contributing to therapeutic strategies for CKD development and progression.
Collapse
|
107
|
van Gastel J, Hendrickx JO, Leysen H, Santos-Otte P, Luttrell LM, Martin B, Maudsley S. β-Arrestin Based Receptor Signaling Paradigms: Potential Therapeutic Targets for Complex Age-Related Disorders. Front Pharmacol 2018; 9:1369. [PMID: 30546309 PMCID: PMC6280185 DOI: 10.3389/fphar.2018.01369] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/07/2018] [Indexed: 12/14/2022] Open
Abstract
G protein coupled receptors (GPCRs) were first characterized as signal transducers that elicit downstream effects through modulation of guanine (G) nucleotide-binding proteins. The pharmacotherapeutic exploitation of this signaling paradigm has created a drug-based field covering nearly 50% of the current pharmacopeia. Since the groundbreaking discoveries of the late 1990s to the present day, it is now clear however that GPCRs can also generate productive signaling cascades through the modulation of β-arrestin functionality. β-Arrestins were first thought to only regulate receptor desensitization and internalization - exemplified by the action of visual arrestin with respect to rhodopsin desensitization. Nearly 20 years ago, it was found that rather than controlling GPCR signal termination, productive β-arrestin dependent GPCR signaling paradigms were highly dependent on multi-protein complex formation and generated long-lasting cellular effects, in contrast to G protein signaling which is transient and functions through soluble second messenger systems. β-Arrestin signaling was then first shown to activate mitogen activated protein kinase signaling in a G protein-independent manner and eventually initiate protein transcription - thus controlling expression patterns of downstream proteins. While the possibility of developing β-arrestin biased or functionally selective ligands is now being investigated, no additional research has been performed on its possible contextual specificity in treating age-related disorders. The ability of β-arrestin-dependent signaling to control complex and multidimensional protein expression patterns makes this therapeutic strategy feasible, as treating complex age-related disorders will likely require therapeutics that can exert network-level efficacy profiles. It is our understanding that therapeutically targeting G protein-independent effectors such as β-arrestin will aid in the development of precision medicines with tailored efficacy profiles for disease/age-specific contextualities.
Collapse
Affiliation(s)
- Jaana van Gastel
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Translational Neurobiology Group, Centre for Molecular Neuroscience, VIB, Antwerp, Belgium
| | - Jhana O Hendrickx
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Translational Neurobiology Group, Centre for Molecular Neuroscience, VIB, Antwerp, Belgium
| | - Hanne Leysen
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Translational Neurobiology Group, Centre for Molecular Neuroscience, VIB, Antwerp, Belgium
| | - Paula Santos-Otte
- Institute of Biophysics, Humboldt University of Berlin, Berlin, Germany
| | - Louis M Luttrell
- Division of Endocrinology, Diabetes and Medical Genetics, Medical University of South Carolina, Charleston, SC, United States
| | - Bronwen Martin
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Stuart Maudsley
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Translational Neurobiology Group, Centre for Molecular Neuroscience, VIB, Antwerp, Belgium
| |
Collapse
|
108
|
Di Liberto V, Mudò G, Belluardo N. Crosstalk between receptor tyrosine kinases (RTKs) and G protein-coupled receptors (GPCR) in the brain: Focus on heteroreceptor complexes and related functional neurotrophic effects. Neuropharmacology 2018; 152:67-77. [PMID: 30445101 DOI: 10.1016/j.neuropharm.2018.11.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 11/01/2018] [Accepted: 11/12/2018] [Indexed: 01/11/2023]
Abstract
Neuronal events are regulated by the integration of several complex signaling networks in which G protein-coupled receptors (GPCRs) and receptor tyrosine kinases (RTKs) are considered key players of an intense bidirectional cross-communication in the cell, generating signaling mechanisms that, at the same time, connect and diversify the traditional signal transduction pathways activated by the single receptor. For this receptor-receptor crosstalk, the two classes of receptors form heteroreceptor complexes resulting in RTKs transactivation and in growth-promoting signals. In this review, we describe heteroreceptor complexes between GPCR and RTKs in the central nervous system (CNS) and their functional effects in controlling a variety of neuronal effects, ranging from development, proliferation, differentiation and migration, to survival, repair, synaptic transmission and plasticity. In this interaction, RTKs can also recruit components of the G protein signaling cascade, creating a bidirectional intricate interplay that provides complex control over multiple cellular events. These heteroreceptor complexes, by the integration of different signals, have recently attracted a growing interest as novel molecular target for depressive disorders. This article is part of the Special Issue entitled 'Receptor heteromers and their allosteric receptor-receptor interactions'.
Collapse
Affiliation(s)
- Valentina Di Liberto
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Giuseppa Mudò
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Natale Belluardo
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy.
| |
Collapse
|
109
|
Neural regulation of drug resistance in cancer treatment. Biochim Biophys Acta Rev Cancer 2018; 1871:20-28. [PMID: 30419311 DOI: 10.1016/j.bbcan.2018.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/16/2018] [Accepted: 10/16/2018] [Indexed: 12/18/2022]
Abstract
The treatment of cancer has made great progress. However, drug resistance remains problematic. Multiple physiologic processes of tumor development can be dominated by central and sympathetic nervous systems. The interactions between the nervous system, immune system, and tumor occur consistently and dynamically. Recent evidence suggests that nerves and neural signals are intimately involved in the development of resistance to cancer therapies. In this review, we will provide an overview of the recent progress in this rapidly growing area and discuss the potential new strategies for targeting the neural signaling pathway to improve the effectiveness of chemotherapies, targeted therapies, and immunotherapies.
Collapse
|
110
|
Dao DT, Anez-Bustillos L, Adam RM, Puder M, Bielenberg DR. Heparin-Binding Epidermal Growth Factor-Like Growth Factor as a Critical Mediator of Tissue Repair and Regeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2446-2456. [PMID: 30142332 PMCID: PMC6207098 DOI: 10.1016/j.ajpath.2018.07.016] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 06/21/2018] [Accepted: 07/13/2018] [Indexed: 11/20/2022]
Abstract
Heparin-binding epidermal growth factor-like growth factor (HB-EGF) is a member of the EGF family. It contains an EGF-like domain as well as a heparin-binding domain that allows for interactions with heparin and cell-surface heparan sulfate. Soluble mature HB-EGF, a ligand of human epidermal growth factor receptors 1 and 4, is cleaved from the membrane-associated pro-HB-EGF by matrix metalloproteinase or a disintegrin and metalloproteinase in a process called ectodomain shedding. Signaling through human epidermal growth factor receptors 1 and 4 results in a variety of effects, including cellular proliferation, migration, adhesion, and differentiation. HB-EGF levels increase in response to different forms of injuries as well as stimuli, such as lysophosphatidic acid, retinoic acid, and 17β-estradiol. Because it is widely expressed in many organs, HB-EGF plays a critical role in tissue repair and regeneration throughout the body. It promotes cutaneous wound healing, hepatocyte proliferation after partial hepatectomy, intestinal anastomosis strength, alveolar regeneration after pneumonectomy, neurogenesis after ischemic injury, bladder wall thickening in response to urinary tract obstruction, and protection against ischemia/reperfusion injury to many cell types. Additionally, innovative strategies to deliver HB-EGF to sites of organ injury or to increase the endogenous levels of shed HB-EGF have been attempted with promising results. Harnessing the reparatory properties of HB-EGF in the clinical setting, therefore, may produce therapies that augment the treatment of various organ injuries.
Collapse
Affiliation(s)
- Duy T Dao
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts; Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Lorenzo Anez-Bustillos
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts; Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rosalyn M Adam
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts; Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts; Urological Diseases Research Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mark Puder
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts; Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Diane R Bielenberg
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts; Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
111
|
Bian G, Yu C, Liu L, Fang C, Chen K, Ren P, Zhang Q, Liu F, Zhang K, Xue Q, Xiang J, Guo H, Song J, Zhao Y, Wu W, Chung SK, Sun R, Ju G, Wang J. Sphingosine 1-phosphate stimulates eyelid closure in the developing rat by stimulating EGFR signaling. Sci Signal 2018; 11:11/553/eaat1470. [DOI: 10.1126/scisignal.aat1470] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In many mammals, the eyelids migrate over the eye and fuse during embryogenesis to protect the cornea from damage during birth and early life. Loss-of-function mutations affecting the epidermal growth factor receptor (EGFR) signaling pathway cause an eyes-open-at-birth (EOB) phenotype in rodents. We identified an insertional mutation in Spinster homolog 2 (Spns2) in a strain of transgenic rats exhibiting the EOB phenotype. Spns2, a sphingosine 1-phosphate (S1P) transporter that releases S1P from cells, was enriched at the tip of developing eyelids in wild-type rat embryos. Spns2 expression or treatment with S1P or any one of several EGFR ligands rescued the EOB Spns2 mutant phenotype in vivo and in tissue explants in vitro and rescued the formation of stress fibers in primary keratinocytes from mutants. S1P signaled through the receptors S1PR1, S1PR2, and S1PR3 to activate extracellular signal–regulated kinase (ERK) and EGFR-dependent mitogen-activated protein kinase kinase kinase 1 (MEKK1)–c-Jun signaling. S1P also induced the nuclear translocation of the transcription factor MAL in a manner dependent on EGFR signaling. MAL and c-Jun stimulated the expression of the microRNAs miR-21 and miR-222, both of which target the metalloprotease inhibitor TIMP3, thus promoting metalloprotease activity. The metalloproteases ADAM10 and ADAM17 stimulated EGFR signaling by cleaving a membrane-anchored form of EGF to release the ligand. Our results outline a network by which S1P transactivates EGFR signaling through a complex mechanism involving feedback between several intra- and extracellular molecules to promote eyelid fusion in the developing rat.
Collapse
|
112
|
O'Brien SL, Johnstone EKM, Devost D, Conroy J, Reichelt ME, Purdue BW, Ayoub MA, Kawai T, Inoue A, Eguchi S, Hébert TE, Pfleger KDG, Thomas WG. BRET-based assay to monitor EGFR transactivation by the AT 1R reveals G q/11 protein-independent activation and AT 1R-EGFR complexes. Biochem Pharmacol 2018; 158:232-242. [PMID: 30347205 DOI: 10.1016/j.bcp.2018.10.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/17/2018] [Indexed: 01/09/2023]
Abstract
The type 1 angiotensin II (AngII) receptor (AT1R) transactivates the epidermal growth factor receptor (EGFR), which leads to pathological remodeling of heart, blood vessels and kidney. End-point assays are used as surrogates of EGFR activation, however these downstream readouts are not applicable to live cells, in real-time. Herein, we report the use of a bioluminescence resonance energy transfer (BRET)-based assay to assess recruitment of the EGFR adaptor protein, growth factor receptor-bound protein 2 (Grb2), to the EGFR. In a variety of cell lines, both epidermal growth factor (EGF) and AngII stimulated Grb2 recruitment to EGFR. The BRET assay was used to screen a panel of 9 G protein-coupled receptors (GPCRs) and further developed for other EGFR family members (HER2 and HER3); the AT1R was able to transactivate HER2, but not HER3. Mechanistically, AT1R-mediated ERK1/2 activation was dependent on Gq/11 and EGFR tyrosine kinase activity, whereas the recruitment of Grb2 to the EGFR was independent of Gq/11 and only partially dependent on EGFR tyrosine kinase activity. This Gq/11 independence of EGFR transactivation was confirmed using AT1R mutants and in CRISPR cell lines lacking Gq/11. EGFR transactivation was also apparently independent of β-arrestins. Finally, we used additional BRET-based assays and confocal microscopy to provide evidence that both AngII- and EGF-stimulation promoted AT1R-EGFR heteromerization. In summary, we report an alternative approach to monitoring AT1R-EGFR transactivation in live cells, which provides a more direct and proximal view of this process, including the potential for complexes between the AT1R and EGFR.
Collapse
Affiliation(s)
- Shannon L O'Brien
- Receptor Biology Group, The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia 4072, Queensland, Australia
| | - Elizabeth K M Johnstone
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia 6009, Australia
| | - Dominic Devost
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Jacinta Conroy
- Receptor Biology Group, The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia 4072, Queensland, Australia
| | - Melissa E Reichelt
- Receptor Biology Group, The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia 4072, Queensland, Australia
| | - Brooke W Purdue
- Receptor Biology Group, The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia 4072, Queensland, Australia
| | - Mohammed A Ayoub
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia 6009, Australia
| | - Tatsuo Kawai
- Cardiovascular Research Centre, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Satoru Eguchi
- Cardiovascular Research Centre, Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Kevin D G Pfleger
- Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Crawley, Western Australia 6009, Australia; Dimerix Limited, Nedlands, Western Australia 6009, Australia
| | - Walter G Thomas
- Receptor Biology Group, The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia 4072, Queensland, Australia; Centre for Cardiac and Vasculature Biology, The University of Queensland, St Lucia 4072, Queensland, Australia.
| |
Collapse
|
113
|
Rhomboids and regulation of receptor tyrosine kinase ligands shedding. EBioMedicine 2018; 37:19-20. [PMID: 30297146 PMCID: PMC6284349 DOI: 10.1016/j.ebiom.2018.09.053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 09/30/2018] [Indexed: 11/22/2022] Open
|
114
|
Sethuraman A, Brown M, Krutilina R, Wu ZH, Seagroves TN, Pfeffer LM, Fan M. BHLHE40 confers a pro-survival and pro-metastatic phenotype to breast cancer cells by modulating HBEGF secretion. Breast Cancer Res 2018; 20:117. [PMID: 30285805 PMCID: PMC6167787 DOI: 10.1186/s13058-018-1046-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 08/28/2018] [Indexed: 12/15/2022] Open
Abstract
Background Metastasis is responsible for a significant number of breast cancer-related deaths. Hypoxia, a primary driving force of cancer metastasis, induces the expression of BHLHE40, a transcription regulator. This study aimed to elucidate the function of BHLHE40 in the metastatic process of breast cancer cells. Methods To define the role of BHLHE40 in breast cancer, BHLHE40 expression was knocked down by a lentiviral construct expressing a short hairpin RNA against BHLHE40 or knocked out by the CRISPR/Cas9 editing system. Orthotopic xenograft and experimental metastasis (tail vein injection) mouse models were used to analyze the role of BHLHE40 in lung metastasis of breast cancer. Global gene expression analysis and public database mining were performed to identify signaling pathways regulated by BHLHE40 in breast cancer. The action mechanism of BHLHE40 was examined by chromatin immunoprecipitation (ChIP), co-immunoprecipitation (CoIP), exosome analysis, and cell-based assays for metastatic potential. Results BHLHE40 knockdown significantly reduced primary tumor growth and lung metastasis in orthotopic xenograft and experimental metastasis models of breast cancer. Gene expression analysis implicated a role of BHLHE40 in transcriptional activation of heparin-binding epidermal growth factor (HBEGF). ChIP and CoIP assays revealed that BHLHE40 induces HBEGF transcription by blocking DNA binding of histone deacetylases (HDAC)1 and HDAC2. Cell-based assays showed that HBEGF is secreted through exosomes and acts to promote cell survival and migration. Public databases provided evidence linking high expression of BHLHE40 and HBEGF to poor prognosis of triple-negative breast cancer. Conclusion This study reveals a novel role of BHLHE40 in promoting tumor cell survival and migration by regulating HBEGF secretion.
Collapse
Affiliation(s)
- Aarti Sethuraman
- Department of Pathology and Laboratory Medicine, and the Center for Cancer Research, University of Tennessee Health Science Center, 19 South Manassas Street, Memphis, TN, 38163, USA
| | - Martin Brown
- Department of Pathology and Laboratory Medicine, and the Center for Cancer Research, University of Tennessee Health Science Center, 19 South Manassas Street, Memphis, TN, 38163, USA
| | - Raya Krutilina
- Department of Pathology and Laboratory Medicine, and the Center for Cancer Research, University of Tennessee Health Science Center, 19 South Manassas Street, Memphis, TN, 38163, USA
| | - Zhao-Hui Wu
- Department of Pathology and Laboratory Medicine, and the Center for Cancer Research, University of Tennessee Health Science Center, 19 South Manassas Street, Memphis, TN, 38163, USA
| | - Tiffany N Seagroves
- Department of Pathology and Laboratory Medicine, and the Center for Cancer Research, University of Tennessee Health Science Center, 19 South Manassas Street, Memphis, TN, 38163, USA
| | - Lawrence M Pfeffer
- Department of Pathology and Laboratory Medicine, and the Center for Cancer Research, University of Tennessee Health Science Center, 19 South Manassas Street, Memphis, TN, 38163, USA
| | - Meiyun Fan
- Department of Pathology and Laboratory Medicine, and the Center for Cancer Research, University of Tennessee Health Science Center, 19 South Manassas Street, Memphis, TN, 38163, USA.
| |
Collapse
|
115
|
Natural molecules induce and synergize to boost expression of the human antimicrobial peptide β-defensin-3. Proc Natl Acad Sci U S A 2018; 115:E9869-E9878. [PMID: 30275324 DOI: 10.1073/pnas.1805298115] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Antimicrobial peptides (AMPs) are mucosal defense effectors of the human innate immune response. In the intestine, AMPs are produced and secreted by epithelial cells to protect the host against pathogens and to support homeostasis with commensals. The inducible nature of AMPs suggests that potent inducers could be used to increase their endogenous expression for the prevention or treatment of diseases. Here we aimed at identifying molecules from the natural pharmacopoeia that induce expression of human β-defensin-3 (HBD3), one of the most efficient AMPs, without modifying the production of proinflammatory cytokines. By screening, we identified three molecules isolated from medicinal plants, andrographolide, oridonin, and isoliquiritigenin, which induced HBD3 production in human colonic epithelial cells. This effect was observed without activation of the NF-κB pathway or the expression of associated proinflammatory cytokines. We identified the EGF receptor as the target of these compounds and characterized the downstream-activated MAPK pathways. At the chromatin level, molecules increased phosphorylation of histone H3 on serine S10 and recruitment of the c-Fos, c-Jun, and Elk1 or c-Myc transcription factors at the HBD3 promoter. Interestingly, stimulating cells with a combination of andrographolide and isoliquiritigenin synergistically enhanced HBD3 induction 10-fold more than observed with each molecule alone. Finally, we investigated the molecular basis governing the synergistic effect, confirmed our findings in human colonic primary cells, and demonstrated that synergism increased cellular antimicrobial activity. This work shows the capability of small molecules to achieve induction of epithelial antimicrobial defenses while simultaneously avoiding the deleterious risks of an inflammatory response.
Collapse
|
116
|
Harun-Or-Rashid M, Hallböök F. Alpha 2-Adrenergic Receptor Agonist Brimonidine Stimulates ERK1/2 and AKT Signaling via Transactivation of EGF Receptors in the Human MIO-M1 Müller Cell Line. Curr Eye Res 2018; 44:34-45. [DOI: 10.1080/02713683.2018.1516783] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Mohammad Harun-Or-Rashid
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Finn Hallböök
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| |
Collapse
|
117
|
Chen R, Jin G, Li W, McIntyre TM. Epidermal Growth Factor (EGF) Autocrine Activation of Human Platelets Promotes EGF Receptor-Dependent Oral Squamous Cell Carcinoma Invasion, Migration, and Epithelial Mesenchymal Transition. THE JOURNAL OF IMMUNOLOGY 2018; 201:2154-2164. [PMID: 30150285 DOI: 10.4049/jimmunol.1800124] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 07/25/2018] [Indexed: 12/11/2022]
Abstract
Activated platelets release functional, high m.w. epidermal growth factor (HMW-EGF). In this study, we show platelets also express epidermal growth factor (EGF) receptor (EGFR) protein, but not ErbB2 or ErbB4 coreceptors, and so might respond to HMW-EGF. We found HMW-EGF stimulated platelet EGFR autophosphorylation, PI3 kinase-dependent AKT phosphorylation, and a Ca2+ transient that were blocked by EGFR tyrosine kinase inhibition. Strong (thrombin) and weak (ADP, platelet-activating factor) G protein-coupled receptor agonists and non-G protein-coupled receptor collagen recruited EGFR tyrosine kinase activity that contributed to platelet activation because EGFR kinase inhibition reduced signal transduction and aggregation induced by each agonist. EGF stimulated ex vivo adhesion of platelets to collagen-coated microfluidic channels, whereas systemic EGF injection increased initial platelet deposition in FeCl3-damaged murine carotid arteries. EGFR signaling contributes to oral squamous cell carcinoma (OSCC) tumorigenesis, but the source of its ligand is not established. We find individual platelets were intercalated within OSCC tumors. A portion of these platelets expressed stimulation-dependent Bcl-3 and IL-1β and so had been activated. Stimulated platelets bound OSCC cells, and material released from stimulated platelets induced OSCC epithelial-mesenchymal transition and stimulated their migration and invasion through Matrigel barriers. Anti-EGF Ab or EGFR inhibitors abolished platelet-induced tumor cell phenotype transition, migration, and invasion; so the only factor released from activated platelets necessary for OSCC metastatic activity was HMW-EGF. These results establish HMW-EGF in platelet function and elucidate a previously unsuspected connection between activated platelets and tumorigenesis through rapid, and prolonged, autocrine-stimulated release of HMW-EGF by tumor-associated platelets.
Collapse
Affiliation(s)
- Rui Chen
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Ge Jin
- Case Western Reserve University School of Dental Medicine, Cleveland, OH 44106
| | - Wei Li
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195.,Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106; and.,Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195
| | - Thomas M McIntyre
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; .,Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106; and.,Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195
| |
Collapse
|
118
|
Qorri B, Kalaydina RV, Velickovic A, Kaplya Y, Decarlo A, Szewczuk MR. Agonist-Biased Signaling via Matrix Metalloproteinase-9 Promotes Extracellular Matrix Remodeling. Cells 2018; 7:cells7090117. [PMID: 30149671 PMCID: PMC6162445 DOI: 10.3390/cells7090117] [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: 06/30/2018] [Revised: 08/12/2018] [Accepted: 08/23/2018] [Indexed: 12/26/2022] Open
Abstract
The extracellular matrix (ECM) is a highly dynamic noncellular structure that is crucial for maintaining tissue architecture and homeostasis. The dynamic nature of the ECM undergoes constant remodeling in response to stressors, tissue needs, and biochemical signals that are mediated primarily by matrix metalloproteinases (MMPs), which work to degrade and build up the ECM. Research on MMP-9 has demonstrated that this proteinase exists on the cell surface of many cell types in complex with G protein-coupled receptors (GPCRs), and receptor tyrosine kinases (RTKs) or Toll-like receptors (TLRs). Through a novel yet ubiquitous signaling platform, MMP-9 is found to play a crucial role not only in the direct remodeling of the ECM but also in the transactivation of associated receptors to mediate and recruit additional remodeling proteins. Here, we summarize the role of MMP-9 as it exists in a tripartite complex on the cell surface and discuss how its association with each of the TrkA receptor, Toll-like receptors, epidermal growth factor receptor, and the insulin receptor contributes to various aspects of ECM remodeling.
Collapse
Affiliation(s)
- Bessi Qorri
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada.
| | | | - Aleksandra Velickovic
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada.
| | - Yekatrina Kaplya
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada.
| | - Alexandria Decarlo
- Department of Biology, Biosciences Complex, Queen's University, Kingston, ON K7L 3N6, Canada.
| | - Myron R Szewczuk
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada.
| |
Collapse
|
119
|
Mitchell RA, Luwor RB, Burgess AW. Epidermal growth factor receptor: Structure-function informing the design of anticancer therapeutics. Exp Cell Res 2018; 371:1-19. [PMID: 30098332 DOI: 10.1016/j.yexcr.2018.08.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/30/2018] [Accepted: 08/01/2018] [Indexed: 12/19/2022]
Abstract
Research on the epidermal growth factor (EGF) family and the family of receptors (EGFR) has progressed rapidly in recent times. New crystal structures of the ectodomains with different ligands, the activation of the kinase domain through oligomerisation and the use of fluorescence techniques have revealed profound conformational changes on ligand binding. The control of cell signaling from the EGFR-family is complex, with heterodimerisation, ligand affinity and signaling cross-talk influencing cellular outcomes. Analysis of tissue homeostasis indicates that the control of pro-ligand processing is likely to be as important as receptor activation events. Several members of the EGFR-family are overexpressed and/or mutated in cancer cells. The perturbation of EGFR-family signaling drives the malignant phenotype of many cancers and both inhibitors and antagonists of signaling from these receptors have already produced therapeutic benefits for patients. The design of affibodies, antibodies, small molecule inhibitors and even immunotherapeutic drugs targeting the EGFR-family has yielded promising new approaches to improving outcomes for cancer patients. In this review, we describe recent discoveries which have increased our understanding of the structure and dynamics of signaling from the EGFR-family, the roles of ligand processing and receptor cross-talk. We discuss the relevance of these studies to the development of strategies for designing more effective targeted treatments for cancer patients.
Collapse
Affiliation(s)
- Ruth A Mitchell
- Structural Biology Division, The Walter and Eliza Hall Institute of Medical Research, Victoria 3052, Australia; Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Rodney B Luwor
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Antony W Burgess
- Structural Biology Division, The Walter and Eliza Hall Institute of Medical Research, Victoria 3052, Australia; Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia.
| |
Collapse
|
120
|
Lichtenthaler SF, Lemberg MK, Fluhrer R. Proteolytic ectodomain shedding of membrane proteins in mammals-hardware, concepts, and recent developments. EMBO J 2018; 37:e99456. [PMID: 29976761 PMCID: PMC6068445 DOI: 10.15252/embj.201899456] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/05/2018] [Accepted: 06/18/2018] [Indexed: 12/14/2022] Open
Abstract
Proteolytic removal of membrane protein ectodomains (ectodomain shedding) is a post-translational modification that controls levels and function of hundreds of membrane proteins. The contributing proteases, referred to as sheddases, act as important molecular switches in processes ranging from signaling to cell adhesion. When deregulated, ectodomain shedding is linked to pathologies such as inflammation and Alzheimer's disease. While proteases of the "a disintegrin and metalloprotease" (ADAM) and "beta-site APP cleaving enzyme" (BACE) families are widely considered as sheddases, in recent years a much broader range of proteases, including intramembrane and soluble proteases, were shown to catalyze similar cleavage reactions. This review demonstrates that shedding is a fundamental process in cell biology and discusses the current understanding of sheddases and their substrates, molecular mechanisms and cellular localizations, as well as physiological functions of protein ectodomain shedding. Moreover, we provide an operational definition of shedding and highlight recent conceptual advances in the field. While new developments in proteomics facilitate substrate discovery, we expect that shedding is not a rare exception, but rather the rule for many membrane proteins, and that many more interesting shedding functions await discovery.
Collapse
Affiliation(s)
- Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, Klinikum rechts der Isar, School of Medicine, and Institute for Advanced Study, Technical University Munich, Munich, Germany
- Munich Center for Systems Neurology (SyNergy), Munich, Germany
| | - Marius K Lemberg
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Regina Fluhrer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Biomedizinisches Centrum (BMC), Ludwig-Maximilians University of Munich, Munich, Germany
| |
Collapse
|
121
|
Hugo de Almeida V, Guimarães IDS, Almendra LR, Rondon AMR, Tilli TM, de Melo AC, Sternberg C, Monteiro RQ. Positive crosstalk between EGFR and the TF-PAR2 pathway mediates resistance to cisplatin and poor survival in cervical cancer. Oncotarget 2018; 9:30594-30609. [PMID: 30093972 PMCID: PMC6078136 DOI: 10.18632/oncotarget.25748] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 06/25/2018] [Indexed: 01/09/2023] Open
Abstract
Cisplatin-based chemoradiation is the standard treatment for cervical cancer, but chemosensitizing strategies are needed to improve patient survival. EGFR (Epidermal Growth Factor Receptor) is an oncogene overexpressed in cervical cancer that is involved in chemoresistance. Recent studies showed that EGFR upregulates multiple elements of the coagulation cascade, including tissue factor (TF) and the protease-activated receptors (PAR) 1 and 2. Moreover, many G protein-coupled receptors, including PARs, have been implicated in EGFR transactivation. However, the role of coagulation proteins in the progression of cervical cancer has been poorly investigated. Herein we employed cervical cancer cell lines and The Cancer Genome Atlas (TCGA) database to evaluate the role of EGFR, TF and PAR2 in chemoresistance. The SLIGKL-NH2 peptide (PAR2-AP) and coagulation factor VIIa (FVIIa) were used as PAR2 agonists, while cetuximab was used to inhibit EGFR. The more aggressive cell line CASKI showed higher expression levels of EGFR, TF and PAR2 than that of C33A. PAR2 transactivated EGFR, which further upregulated cyclooxygenase-2 (COX2) expression. PAR2-AP decreased cisplatin-induced apoptosis through an EGFR- and COX2-dependent mechanism. Furthermore, treatment of CASKI cells with EGF upregulated TF expression, while treatment with cetuximab decreased the TF protein levels. The RNA-seq data from 309 TCGA samples showed a strong positive correlation between EGFR and TF expression (P = 0.0003). In addition, the increased expression of EGFR, PAR2 or COX2 in cervical cancer patients was significantly correlated with poor overall survival. Taken together, our results suggest that EGFR and COX2 are effectors of the TF/FVIIa/PAR2 signaling pathway, promoting chemoresistance.
Collapse
Affiliation(s)
- Vitor Hugo de Almeida
- Instituto de Bioquímica Médica Leopoldo De Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.,Divisão de Pesquisa Clínica e Desenvolvimento Tecnológico, Instituto Nacional de Câncer, Rio de Janeiro, RJ, Brazil
| | | | - Lucas R Almendra
- Instituto de Bioquímica Médica Leopoldo De Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Araci M R Rondon
- Instituto de Bioquímica Médica Leopoldo De Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Tatiana M Tilli
- Centro de Desenvolvimento Tecnológico em Saúde, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Andréia C de Melo
- Divisão de Pesquisa Clínica e Desenvolvimento Tecnológico, Instituto Nacional de Câncer, Rio de Janeiro, RJ, Brazil
| | - Cinthya Sternberg
- Divisão de Pesquisa Clínica e Desenvolvimento Tecnológico, Instituto Nacional de Câncer, Rio de Janeiro, RJ, Brazil.,Present address: Sociedade Brasileira de Oncologia Clínica (SBOC), Belo Horizonte, MG, Brazil
| | - Robson Q Monteiro
- Instituto de Bioquímica Médica Leopoldo De Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
122
|
Shen Z, Yang X, Chen Y, Shi L. CAPA periviscerokinin-mediated activation of MAPK/ERK signaling through Gq-PLC-PKC-dependent cascade and reciprocal ERK activation-dependent internalized kinetics of Bom-CAPA-PVK receptor 2. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2018; 98:1-15. [PMID: 29730398 DOI: 10.1016/j.ibmb.2018.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 04/16/2018] [Accepted: 04/28/2018] [Indexed: 06/08/2023]
Abstract
Bombyx mori neuropeptide G protein-coupled receptor (BNGR)-A27 is a specific receptor for B. mori capability (CAPA) periviscerokinin (PVK), that is, Bom-CAPA-PVK receptor 2. Upon stimulation of Bom-CAPA-PVK-1 or -PVK-2, Bom-CAPA-PVK receptor 2 significantly increases cAMP-response element-controlled luciferase activity and Ca2+ mobilization in a Gq inhibitor-sensitive manner. However, the underlying mechanism(s) for CAPA/CAPA receptor system mediation of extracellular signal-regulated kinases1/2 (ERK1/2) activation remains to be explained further. Here, we discovered that Bom-CAPA-PVK receptor 2 stimulated ERK1/2 phosphorylation in a dose- and time-dependent manner in response to Bom-CAPA-PVK-1 or -PVK-2 with similar potencies. Furthermore, ERK1/2 phosphorylation can be inhibited by Gq inhibitor UBO-QIC, PLC inhibitor U73122, protein kinase C (PKC) inhibitor Go 6983, phospholipase D (PLD) inhibitor FIPI and Ca2+ chelators EGTA and BAPTA-AM. Moreover, Bom-CAPA-PVK-R2-induced activation of ERK1/2 was significantly attenuated by treatment with the Gβγ-specific inhibitors, phosphatidylinositol 3-kinase (PI3K)-specific inhibitor Wortmannin and Src-specific inhibitor PP2. Our data also demonstrate that receptor tyrosine kinase (RTK) transactivation pathways are involved in the mechanisms of Bom-CAPA-PVK receptor to ERK1/2 phosphorylation. In addition, β-arrestin1/2 is not involved in Bom-CAPA-PVK-R2-mediated ERK1/2 activation but required for the agonist-independent, ERK1/2 activation-dependent internalization of the G protein-coupled receptor (GPCR).
Collapse
Affiliation(s)
- Zhangfei Shen
- Department of Economic Zoology, College of Animal Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Xiaoyuan Yang
- College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yu Chen
- College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Liangen Shi
- Department of Economic Zoology, College of Animal Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
123
|
Phosphorylation of iRhom2 Controls Stimulated Proteolytic Shedding by the Metalloprotease ADAM17/TACE. Cell Rep 2018; 21:745-757. [PMID: 29045841 PMCID: PMC5656746 DOI: 10.1016/j.celrep.2017.09.074] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 07/12/2017] [Accepted: 09/22/2017] [Indexed: 12/02/2022] Open
Abstract
Cell surface metalloproteases coordinate signaling during development, tissue homeostasis, and disease. TACE (TNF-α-converting enzyme), is responsible for cleavage (“shedding”) of membrane-tethered signaling molecules, including the cytokine TNF, and activating ligands of the EGFR. The trafficking of TACE within the secretory pathway requires its binding to iRhom2, which mediates the exit of TACE from the endoplasmic reticulum. An important, but mechanistically unclear, feature of TACE biology is its ability to be stimulated rapidly on the cell surface by numerous inflammatory and growth-promoting agents. Here, we report a role for iRhom2 in TACE stimulation on the cell surface. TACE shedding stimuli trigger MAP kinase-dependent phosphorylation of iRhom2 N-terminal cytoplasmic tail. This recruits 14-3-3 proteins, enforcing the dissociation of TACE from complexes with iRhom2, promoting the cleavage of TACE substrates. Our data reveal that iRhom2 controls multiple aspects of TACE biology, including stimulated shedding on the cell surface. iRhom2 is phosphorylated in response to stimuli that activate the sheddase TACE Blocking iRhom phosphorylation represses TACE stimulated shedding Phosphorylated iRhom2 recruits 14-3-3 and dissociates from TACE, enabling shedding iRhom2 is thus a signal integrator and transducer of stimulated TACE shedding
Collapse
|
124
|
Sun N, Zhang X, Guo S, Le HT, Zhang X, Kim KM. Molecular mechanisms involved in epidermal growth factor receptor-mediated inhibition of dopamine D 3 receptor signaling. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1187-1200. [PMID: 29885323 DOI: 10.1016/j.bbamcr.2018.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/28/2018] [Accepted: 06/05/2018] [Indexed: 01/22/2023]
Abstract
The phenomenon wherein the signaling by a given receptor is regulated by a different class of receptors is termed transactivation or crosstalk. Crosstalk between receptor tyrosine kinases (RTKs) and G protein-coupled receptors (GPCRs) is highly diverse and has unique functional implications because of the distinct structural features of the receptors and the signaling pathways involved. The present study used the epidermal growth factor receptor (EGFR) and dopamine D3 receptor (D3R), which are both associated with schizophrenia, as the model system to study crosstalk between RTKs and GPCRs. Loss-of-function approaches were used to identify the cellular components involved in the tyrosine phosphorylation of G protein-coupled receptor kinase 2 (GRK2), which is responsible for EGFR-induced regulation of the functions of D3R. SRC proto-oncogene (Src, non-receptor tyrosine kinase), heterotrimeric G protein Gβγ subunit, and endocytosis of EGFR were involved in the tyrosine phosphorylation of GRK2. In response to EGF treatment, Src interacted with EGFR in a Gβγ-dependent manner, resulting in the endocytosis of EGFR. Internalized EGFR in the cytosol mediated Src/Gβγ-dependent tyrosine phosphorylation of GRK2. The binding of tyrosine-phosphorylated GRK2 to the T142 residue of D3R resulted in uncoupling from G proteins, endocytosis, and lysosomal downregulation. This study identified the molecular mechanisms involved in the EGFR-mediated regulation of the functions of D3R, which can be extended to the crosstalk between other RTKs and GPCRs.
Collapse
Affiliation(s)
- Ningning Sun
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju, 61186, Republic of Korea
| | - Xiaowei Zhang
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju, 61186, Republic of Korea
| | - Shuohan Guo
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju, 61186, Republic of Korea
| | - Hang Thi Le
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju, 61186, Republic of Korea
| | - Xiaohan Zhang
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju, 61186, Republic of Korea
| | - Kyeong-Man Kim
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju, 61186, Republic of Korea.
| |
Collapse
|
125
|
Kreitman M, Noronha A, Yarden Y. Irreversible modifications of receptor tyrosine kinases. FEBS Lett 2018; 592:2199-2212. [PMID: 29790151 DOI: 10.1002/1873-3468.13095] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 04/12/2018] [Accepted: 05/08/2018] [Indexed: 01/18/2023]
Abstract
Each group of the 56 receptor tyrosine kinases (RTK) binds with one or more soluble growth factors and coordinates a vast array of cellular functions. These outcomes are tightly regulated by inducible post-translational events, such as tyrosine phosphorylation, ubiquitination, ectodomain shedding, and regulated intramembrane proteolysis. Because of the delicate balance required for appropriate RTK function, cells may become pathogenic upon dysregulation of RTKs themselves or their post-translational covalent modifications. For example, reduced ectodomain shedding and decreased ubiquitination of the cytoplasmic region, both of which enhance growth factor signals, characterize malignant cells. Whereas receptor phosphorylation and ubiquitination are reversible, proteolytic cleavage events are irreversible, and either modification might alter the subcellular localization of RTKs. Herein, we focus on ectodomain shedding by metalloproteinases (including ADAM family proteases), cleavage within the membrane or cytoplasmic regions of RTKs (by gamma-secretases and caspases, respectively), and complete receptor proteolysis in lysosomes and proteasomes. Roles of irreversible modifications in RTK signaling, pathogenesis, and pharmacology are highlighted.
Collapse
Affiliation(s)
- Matthew Kreitman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Ashish Noronha
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
126
|
Okusha Y, Eguchi T, Sogawa C, Okui T, Nakano K, Okamoto K, Kozaki K. The intranuclear PEX domain of MMP involves proliferation, migration, and metastasis of aggressive adenocarcinoma cells. J Cell Biochem 2018; 119:7363-7376. [DOI: 10.1002/jcb.27040] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 04/06/2018] [Indexed: 01/06/2023]
Affiliation(s)
- Yuka Okusha
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayama UniversityOkayamaJapan
| | - Takanori Eguchi
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayama UniversityOkayamaJapan
- Advanced Research Center for Oral and Craniofacial Sciences, Graduate School of Medicine, Dentistry andPharmaceutical SciencesOkayama UniversityOkayamaJapan
| | - Chiharu Sogawa
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayama UniversityOkayamaJapan
| | - Tatsuo Okui
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayama UniversityOkayamaJapan
| | - Keisuke Nakano
- Advanced Research Center for Oral and Craniofacial Sciences, Graduate School of Medicine, Dentistry andPharmaceutical SciencesOkayama UniversityOkayamaJapan
- Department of Pathology and Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayama UniversityOkayamaJapan
| | - Kuniaki Okamoto
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayama UniversityOkayamaJapan
| | - Ken‐Ichi Kozaki
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayama UniversityOkayamaJapan
| |
Collapse
|
127
|
Khedr MEMS, Abdelmotelb AM, Pender SLF, Zhou X, Walls AF. Neutrophilia, gelatinase release and microvascular leakage induced by human mast cell tryptase in a mouse model: Lack of a role of protease-activated receptor 2 (PAR2). Clin Exp Allergy 2018; 48:555-567. [PMID: 29383785 PMCID: PMC5969079 DOI: 10.1111/cea.13108] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 01/20/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND Tryptase, the most abundant protease of the human mast cell, has been implicated as a key mediator of allergic inflammation that acts through activation of PAR2. OBJECTIVES To investigate the contribution of PAR2 in the pro-inflammatory actions mediated by tryptase in a mice model. METHODS We have injected recombinant human βII-tryptase into the peritoneum of PAR2-deficient and wild-type C57BL/6 mice. After 6, 12 and 24 hours, mice were killed, peritoneal lavage performed and inflammatory changes investigated. RESULTS Tryptase stimulated an increase in neutrophil numbers in the peritoneum, but responses did not differ between PAR2-deficient and wild-type mice. Heat inactivation of tryptase or pre-incubation with a selective tryptase inhibitor reduced neutrophilia, but neutrophil accumulation was not elicited with a peptide agonist of PAR2 (SLIGRL-NH2 ). Zymography indicated that tryptase stimulated the release of matrix metalloproteinases (MMP) 2 and 9 in the peritoneum of both mouse strains. Studies involving immunomagnetic isolation of neutrophils suggested that neutrophils represent the major cellular source of tryptase-induced MMP2 and MMP9. At 24 hours after tryptase injection, there was increased microvascular leakage as indicated by high levels of albumin in peritoneal lavage fluid, and this appeared to be partially abolished by heat-inactivating tryptase or addition of a protease inhibitor. There was no corresponding increase in levels of histamine or total protein. The extent of tryptase-induced microvascular leakage or gelatinase release into the peritoneum did not differ between PAR2-deficient and wild-type mice. CONCLUSIONS Our findings indicate that tryptase is a potent stimulus for neutrophil accumulation, MMP release and microvascular leakage. Although these actions required an intact catalytic site, the primary mechanism of tryptase in vivo would appear to involve processes independent of PAR2.
Collapse
Affiliation(s)
- M. E. M. S. Khedr
- Clinical and Experimental Sciences Academic UnitFaculty of MedicineUniversity of SouthamptonSouthamptonUK
- Faculty of MedicineSuez Canal UniversityIsmailiaEgypt
| | - A. M. Abdelmotelb
- Clinical and Experimental Sciences Academic UnitFaculty of MedicineUniversity of SouthamptonSouthamptonUK
- Faculty of MedicineTanta UniversityTantaEgypt
| | - S. L. F. Pender
- Clinical and Experimental Sciences Academic UnitFaculty of MedicineUniversity of SouthamptonSouthamptonUK
| | - X. Zhou
- Clinical and Experimental Sciences Academic UnitFaculty of MedicineUniversity of SouthamptonSouthamptonUK
| | - A. F. Walls
- Clinical and Experimental Sciences Academic UnitFaculty of MedicineUniversity of SouthamptonSouthamptonUK
| |
Collapse
|
128
|
Scopinaro F, Varvarigou A, Ussof W, De Vincentis G, Archimandritis S, Evangelatos G, Corleto V, Pulcini A, Capoccetti F, Remediani S, Massa R. Breast Cancer Takes up 99mTc Bombesin. A Preliminary Report. TUMORI JOURNAL 2018; 88:S25-8. [PMID: 12365377 DOI: 10.1177/030089160208800331] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background Several tumors including lung, prostate, ovarian, colon, and exocrine pancreatic cancer show receptors for the amphibian neurotransmitter and growth factor bombesin (BN) and its mammalian counterparts gastrin-releasing peptide and neuromedin B. Also breast cancer has been reported to show such receptors: the presence of BN receptors in primary breast cancer has been demonstrated on cultured cells and by autoradiography on breast tissue samples. Authors who have studied BN receptors in breast cancer do not agree on their frequency in primary cancer, but indicate that 100% of metastatic breast cancers show such receptors. Methods We examined three primary breast cancer patients with 99mTc BN and 99mTc sestamibi one week before surgery. One of them showed axillary node invasion. The same acquisition technique was used for breast and chest imaging with both radiopharmaceuticals, whereas total body images were acquired only with 99mTc BN. Also the administered radioactivity was different: 20 mCi of 99mTc sestamibi and 5-8 mCi of 99mTc BN. Dynamic images were acquired for 20 mins after iv injection with the patient in ventral decubitus and the gamma camera positioned in lateral view, as is generally done in Khakhali's prone scintimammography. Anterior chest images were acquired for 30 mins. Prone scintimammography was performed one hour after administration of both tracers. ROIs were drawn on tumors and surrounding breast with the same technique in order to calculate the tumor to breast ratio (T/B). In addition, total body scan was performed one hour and three hours after 99mTc BN administration. All three patients underwent breast conserving surgery with lymphadenectomy. Postoperative pathologic assessment showed the following T and N stages in the three patients: T1bN0, T1cN0. and T1cN1. Results All three cancers were imaged with both tracers. The T/B of 99mTc BN was always higher than that of 99mTc sestamibi. Chest uptake was always much higher with 99mTc sestamibi than with 99mTc BN. Comparison between 99mTc BN and 99mTc sestamibi images gave other intriguing results: in the N1 patient both tracers clearly imaged the invaded node, but on the 99mTc BN image the primary tumor was larger than on the 99mTc sestamibi image and the node was smaller. It is known that 99mTc BN is not taken up by vessels and inflammatory tissue. The time activity curves of the two tracers were significantly different in all patients, with an increase in 99mTc BN uptake in the first three to five minutes, followed by a less sharp uprise of the curve, quite similar to a plateau. Conclusions Our first impression is that 99mTc BN is a useful breast cancer seeking agent and very promising for lymph node staging.
Collapse
|
129
|
Ozis SE, Akhayeva T, Guner S, Kilicoglu SS, Pampal A. Etanercept restores vasocontractile sensitivity affected by mesenteric ischemia reperfusion. J Surg Res 2018; 226:8-14. [PMID: 29661292 DOI: 10.1016/j.jss.2018.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/23/2017] [Accepted: 01/03/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND The aim of the study is to evaluate in vivo and in vitro effects of etanercept, a soluble tumor necrosis factor receptor, on the contractile responses of superior mesenteric artery in an experimental mesenteric ischemia and reperfusion model. MATERIAL AND METHODS After obtaining animal ethics committee approval, 24 Sprague-Dawley rats were allocated to three groups. Control group (Gr C, n = 6) underwent a sham operation, whereas ischemia/reperfusion and treatment groups underwent 90 min ischemia and 24-h reperfusion (Gr I/R, n = 12; Gr I/R+E, n = 6). The treatment group received 5 mg/kg etanercept intravenously at the beginning of reperfusion. At the end of reperfusion, all animals were sacrificed, and third branch of superior mesenteric artery was dissected for evaluation of contractile responses. In vitro effects of etanercept on vasocontractile responses were also evaluated. The excised ileums were analyzed under light microscope. Two-way analysis of variance following Bonferroni post hoc test was used for evaluation of contractile responses. RESULTS Endothelin-1 and phenylephrine-mediated vasocontractile sensitivity were found increased in Gr I/R when compared with Gr C. Both intravenous administration and organ bath incubation of etanercept decreased the sensitivity of contractile agents for Gr I/R. Mucosal injury, lamina propria disintegration, and denuded villous tips were observed in Gr I/R, whereas the epithelial injury and the subepithelial edema were found to be milder in Gr I/R+E. CONCLUSIONS Etanercept can be a promising agent in mesenteric ischemic reperfusion injury as it does not only inhibit inflammation by blocking tumor necrosis factor-α in circulation but also restores vascular contractility during reflow. These findings support an unexplained recuperative effect of drug beyond its anti-inflammatory effects.
Collapse
Affiliation(s)
- S Erpulat Ozis
- Department of General Surgery, Faculty of Medicine, TOBB-ETU University, Ankara, Turkey
| | - Tamila Akhayeva
- Department of Pharmacology, Astana Medical University, Astana, Kazakhstan
| | - Sahika Guner
- Department of Medical Pharmacology, Faculty of Medicine, Ufuk University, Ankara, Turkey
| | - Sibel S Kilicoglu
- Department of Histology and Embryology, Faculty of Medicine, Ufuk University, Ankara, Turkey
| | - Arzu Pampal
- Department of Pediatric Surgery, Faculty of Medicine, Ufuk University, Ankara, Turkey.
| |
Collapse
|
130
|
Abstract
G protein-coupled receptors (GPCRs) comprise the largest family of receptors in humans. Traditional activation of GPCRs involves binding of a ligand to the receptor, activation of heterotrimeric G proteins and induction of subsequent signaling molecules. It is now known that GPCR signaling occurs through G protein-independent pathways including signaling through β-arrestin and transactivation of other receptor types. Generally, transactivation occurs when activation of one receptor leads to the activation of another receptor(s). GPCR-mediated transactivation is an essential component of GPCR signaling, as activation of other receptor types, such as receptor tyrosine kinases, allows GPCRs to expand their signal transduction and affect various cellular responses. Several mechanisms have been identified for receptor transactivation downstream of GPCRs, one of which involves activation of extracellular proteases, such as a disintegrin and metalloprotease, and matrix metalloproteases . These proteases cleave and release ligands that are then able to activate their respective receptors. A disintegrin and metalloprotease, and matrix metalloproteases can be activated via various mechanisms downstream of GPCR activation, including activation via second messenger, direct phosphorylation, or direct G protein interaction. Additional understanding of the mechanisms involved in GPCR-mediated protease activation and subsequent receptor transactivation could lead to identification of new therapeutic targets.
Collapse
|
131
|
Jain R, Watson U, Vasudevan L, Saini DK. ERK Activation Pathways Downstream of GPCRs. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 338:79-109. [PMID: 29699693 DOI: 10.1016/bs.ircmb.2018.02.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
GPCRs, the 7-TM receptors, represent a class of cell surface receptors which modulate a variety of physiological responses. The serpentine structure in addition to contributing the diversity of stimuli these receptors can sense also provides flexibility to the extracellular and intracellular regions where other proteins can interact with and can form functionally active multimeric entities. The range in signaling and physiological responses generated by these receptors can be attributed to a large repertoire of the receptor subtypes as well as their differential coupling to various classes of G-protein subunits and other proteins which facilitate multistate activation. A multistate GPCR can engage diverse signaling molecules, thereby modulating not only the canonical cellular responses but also noncanonical responses typically associated with activation of other cascades such as RTK and MAPK/ERK signaling. Given the crucial involvement of MAP kinase/ERK signaling in cell fate determination specially with respect to regulating cell proliferation, cellular apoptosis, and survival, GPCR-mediated cross-activation of MAPK has been explored in various systems and shown to involve functional integration of multiple pathways. This review describes the present knowledge of the different mechanisms of ERK activation downstream of GPCRs and our present understanding of receptor-dependent and -independent MAPK activation cascades.
Collapse
Affiliation(s)
- Ruchi Jain
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Uchenna Watson
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India; Department of Studies in Zoology, University of Mysore, Manasagangothri, Mysore, India
| | - Lakshmi Vasudevan
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India; L-GEST-Laboratory of Toxicology, Faculty of Pharmaceutical Sciences, Ghent, Belgium
| | - Deepak K Saini
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India; Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India.
| |
Collapse
|
132
|
Dai L, Pan Q, Peng Y, Huang S, Liu J, Chen T, Wang X, Chen D, Wang J, Zhu Y, Wang H, Liu Y, Ou Y, Yu X, Cao K. p53 Plays a Key Role in the Apoptosis of Human Ovarian Cancer Cells Induced by Adenovirus-Mediated CRM197. Hum Gene Ther 2018; 29:916-926. [PMID: 29620944 DOI: 10.1089/hum.2017.186] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cross-reacting material 197 (CRM197) is a mutant form of the diphtheria toxin. Recent studies have found that CRM197 exerts an experimental antitumor effect on several types of tumors. This study applied a novel treatment of adenovirus-mediated CRM197 (AdCRM197) to human ovarian cancer cells. Interestingly, it was found that A2780 cells were sensitive to AdCRM197, but SKOV3 cells were resistant to it. Since SKOV3 cells are p53 deletion cells, while A2780 cells are p53 wild-type cells, it was postulated that p53 might play a key role in AdCRM197-induced apoptosis. This presumption was demonstrated by means of knockdown of p53 of the A2780 cells through lentivirus-mediated RNA interference. This knockdown resulted in the A2780 cells becoming resistant to AdCRM197. To verify this presumption further, the wild-type p53 gene in the SKOV3 cells was replaced with adenovirus-mediated p53 (Adp53). As expected, AdCRM197 plus Adp53 resulted in apoptosis of the SKOV3 cells. The combined treatment of AdCRM197 plus Adp53 also showed a good antitumor effect in the in vivo experiment on nude mice with xenograft tumors. Taking these results together, it is concluded that AdCRM197 induces apoptosis of human ovarian cancer cells via the p53 pathway. Moreover, it was found that Adp53 can reverse the resistance of p53-deletion human ovarian cancer cells to AdCRM197. The combination of AdCRM197 and Adp53 may be a potentially effective method for overcoming the resistance of p53-deficient human ovarian cancer to AdCRM197.
Collapse
Affiliation(s)
- Lvxia Dai
- 1 Experiment Teaching Center of Clinical Medicine, Chengdu Medical College , Chengdu, China
| | - Qu Pan
- 2 Department of Pathogen Biology, Chengdu Medical College , Chengdu, China
| | - Yanjuan Peng
- 3 Department of Pharmacology, Chengdu Medical College , Chengdu, China
| | - Sizhou Huang
- 4 Development and Regeneration Key Laboratory of Sichuan Province, Chengdu Medical College , Chengdu, China
| | - Jianmin Liu
- 5 Department of Neurosurgery, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine , Guangzhou, China
| | - Tian Chen
- 2 Department of Pathogen Biology, Chengdu Medical College , Chengdu, China
| | - Xin Wang
- 2 Department of Pathogen Biology, Chengdu Medical College , Chengdu, China
| | - Dengbang Chen
- 1 Experiment Teaching Center of Clinical Medicine, Chengdu Medical College , Chengdu, China
| | - Jiandong Wang
- 2 Department of Pathogen Biology, Chengdu Medical College , Chengdu, China
| | - Yanfeng Zhu
- 6 School of Public Health, Chengdu Medical College , Chengdu, China
| | - Hui Wang
- 2 Department of Pathogen Biology, Chengdu Medical College , Chengdu, China
| | - Yilun Liu
- 7 Center for Scientific Research , First Affiliated Hospital, Chengdu Medical College, Chengdu, China
| | - Yu Ou
- 6 School of Public Health, Chengdu Medical College , Chengdu, China
| | - Xiaoping Yu
- 6 School of Public Health, Chengdu Medical College , Chengdu, China
| | - Kang Cao
- 2 Department of Pathogen Biology, Chengdu Medical College , Chengdu, China
| |
Collapse
|
133
|
Wang W, Qiao Y, Li Z. New Insights into Modes of GPCR Activation. Trends Pharmacol Sci 2018; 39:367-386. [DOI: 10.1016/j.tips.2018.01.001] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/03/2018] [Accepted: 01/08/2018] [Indexed: 12/22/2022]
|
134
|
Bazzani L, Donnini S, Finetti F, Christofori G, Ziche M. PGE2/EP3/SRC signaling induces EGFR nuclear translocation and growth through EGFR ligands release in lung adenocarcinoma cells. Oncotarget 2018; 8:31270-31287. [PMID: 28415726 PMCID: PMC5458206 DOI: 10.18632/oncotarget.16116] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 03/01/2017] [Indexed: 12/12/2022] Open
Abstract
Prostaglandin E2 (PGE2) interacts with tyrosine kinases receptor signaling in both tumor and stromal cells supporting tumor progression. Here we demonstrate that in non-small cell lung carcinoma (NSCLC) cells, A549 and GLC82, PGE2 promotes nuclear translocation of epidermal growth factor receptor (nEGFR), affects gene expression and induces cell growth. Indeed, cyclin D1, COX-2, iNOS and c-Myc mRNA levels are upregulated following PGE2 treatment. The nuclear localization sequence (NLS) of EGFR as well as its tyrosine kinase activity are required for the effect of PGE2 on nEGFR and downstream signaling activities. PGE2 binds its bona fide receptor EP3 which by activating SRC family kinases, induces ADAMs activation which, in turn, releases EGFR-ligands from the cell membrane and promotes nEGFR. Amphiregulin (AREG) and Epiregulin (EREG) appear to be involved in nEGFR promoted by the PGE2/EP3-SRC axis. Pharmacological inhibition or silencing of the PGE2/EP3/SRC-ADAMs signaling axis or EGFR ligands i.e. AREG and EREG expression abolishes nEGFR induced by PGE2. In conclusion, PGE2 induces NSCLC cell proliferation by EP3 receptor, SRC-ADAMs activation, EGFR ligands shedding and finally, phosphorylation and nEGFR. Since nuclear EGFR is a hallmark of cancer aggressiveness, our findings reveal a novel mechanism for the contribution of PGE2 to tumor progression.
Collapse
Affiliation(s)
- Lorenzo Bazzani
- Department of Life Sciences, University of Siena, 53100, Siena, Italy.,Department of Biomedizin, University of Basel, 4058, Basel, Switzerland
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, 53100, Siena, Italy
| | - Federica Finetti
- Department of Life Sciences, University of Siena, 53100, Siena, Italy
| | | | - Marina Ziche
- Department of Life Sciences, University of Siena, 53100, Siena, Italy
| |
Collapse
|
135
|
Hedemann N, Rogmans C, Sebens S, Wesch D, Reichert M, Schmidt-Arras D, Oberg HH, Pecks U, van Mackelenbergh M, Weimer J, Arnold N, Maass N, Bauerschlag DO. ADAM17 inhibition enhances platinum efficiency in ovarian cancer. Oncotarget 2018; 9:16043-16058. [PMID: 29662625 PMCID: PMC5882316 DOI: 10.18632/oncotarget.24682] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 02/28/2018] [Indexed: 12/18/2022] Open
Abstract
Chemotherapeutic resistance evolves in about 70 % of ovarian cancer patients and is a major cause of death in this tumor entity. Novel approaches to overcome these therapeutic limitations are therefore highly warranted. A disintegrin and metalloprotease 17 (ADAM17) is highly expressed in ovarian cancer and required for releasing epidermal growth factor receptor (EGFR) ligands like amphiregulin (AREG). This factor has recently been detected in ascites of advanced stage ovarian cancer patients. However, it is not well understood, whether and how ADAM17 might contribute to chemo resistance of ovarian cancer. In this study, we identified ADAM17 as an essential upstream regulator of AREG release under chemotherapeutic treatment in ovarian cancer cell lines and patient derived cells. In the majority of ovarian cancer cells cisplatin treatment resulted in enhanced ADAM17 activity, as shown by an increased shedding of AREG. Moreover, both mRNA and the protein content of AREG were dose-dependently increased by cisplatin exposure. Consequently, cisplatin strongly induced phosphorylation of ADAM17-downstream mediators, the EGFR and extracellular signal-regulated kinases (ERK). Phorbol 12-myristate 13-acetate (PMA), similarly to cisplatin, mediated AREG shedding and membrane fading of surface ADAM17. Inhibition of ADAM17 with either GW280264X or the anti-ADAM17 antibody D1 (A12) as well as silencing of ADAM17 by siRNA selectively reduced AREG release. Thus, ADAM17 inhibition sensitized cancer cells to cisplatin-induced apoptosis, and significantly reduced cell viability. Based on these findings, we propose that targeting of ADAM17 in parallel to chemotherapeutic treatment suppresses survival pathways and potentially diminish evolving secondary chemo resistance mechanisms.
Collapse
Affiliation(s)
- Nina Hedemann
- Department of Gynecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Christoph Rogmans
- Department of Gynecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Susanne Sebens
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Daniela Wesch
- Institute of Immunology, Christian-Albrechts-University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Manuel Reichert
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Dirk Schmidt-Arras
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Hans-Heinrich Oberg
- Institute of Immunology, Christian-Albrechts-University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Ulrich Pecks
- Department of Gynecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Marion van Mackelenbergh
- Department of Gynecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Jörg Weimer
- Department of Gynecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Norbert Arnold
- Department of Gynecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Nicolai Maass
- Department of Gynecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Dirk O Bauerschlag
- Department of Gynecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| |
Collapse
|
136
|
Talati N, Kamato D, Piva TJ, Little PJ, Osman N. Thrombin promotes PAI-1 expression and migration in keratinocytes via ERK dependent Smad linker region phosphorylation. Cell Signal 2018; 47:37-43. [PMID: 29577978 DOI: 10.1016/j.cellsig.2018.03.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 02/19/2018] [Accepted: 03/20/2018] [Indexed: 01/18/2023]
Abstract
Keratinocyte proliferation and migration is essential during re-epithelialisation for the restoration of the epithelial barrier during skin wound healing. Numerous growth factors are involved in the stimulation of keratinocyte proliferation and migration. The signalling pathways that drive these processes during wound healing are not well defined. This study investigated thrombin-mediated signalling in keratinocytes. The thrombin receptor, protease-activated receptor 1 (PAR-1) is a seven transmembrane G-protein coupled receptor that is known to transactivate the epidermal growth factor receptor (EGFR). Immortalized human keratinocytes (HaCaT cells) were treated with thrombin and selective inhibitors to EGFR and MAP kinases. Whole cell lysates were separated on SDS-PAGE and analysed by Western blot using antibodies against transcription factor Smad2. Quantitative real-time polymerase chain reaction was used to measure the mRNA expression of PAI-1 while scratch wound assays were used to measure keratinocyte migration. Western blot data showed that thrombin mediates PAR-1 transactivation of EGFR and the downstream phosphorylation of the transcription factor Smad2 linker (Smad2L) region. ERK1/2 inhibition by UO126 caused a decrease in Smad2L phosphorylation while the p38 inhibitor SB202190 and JNK inhibitor SP600125 did not. Smad2L Ser250 was specifically phosphorylated by this thrombin mediated pathway while Ser245 and Ser255 were not. Thrombin increased PAI-1 mRNA expression and keratinocyte migration and this was reduced when either EGFR or ERK1/2 were blocked. Taken together these results show that thrombin mediated mRNA expression of PAI-1 in keratinocytes and migration occurs via EGFR transactivation and involves signalling intermediates ERK1/2 and Smad2 and may be a key pathway in skin wound healing.
Collapse
Affiliation(s)
- Nirali Talati
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - Danielle Kamato
- School of Pharmacy, The University of Queensland, Wooloongabba, QLD 4102, Australia
| | - Terrence J Piva
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Wooloongabba, QLD 4102, Australia; Department of Pharmacy, Xinhua College of Sun Yat-sen University, Guangzhou 510520, China
| | - Narin Osman
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia; School of Pharmacy, The University of Queensland, Wooloongabba, QLD 4102, Australia; Department of Immunology, Monash University, Melbourne, VIC 3004, Australia.
| |
Collapse
|
137
|
Szczepanska-Sadowska E, Czarzasta K, Cudnoch-Jedrzejewska A. Dysregulation of the Renin-Angiotensin System and the Vasopressinergic System Interactions in Cardiovascular Disorders. Curr Hypertens Rep 2018; 20:19. [PMID: 29556787 PMCID: PMC5859051 DOI: 10.1007/s11906-018-0823-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Purpose of Review In many instances, the renin-angiotensin system (RAS) and the vasopressinergic system (VPS) are jointly activated by the same stimuli and engaged in the regulation of the same processes. Recent Findings Angiotensin II (Ang II) and arginine vasopressin (AVP), which are the main active compounds of the RAS and the VPS, interact at several levels. Firstly, Ang II, acting on AT1 receptors (AT1R), plays a significant role in the release of AVP from vasopressinergic neurons and AVP, stimulating V1a receptors (V1aR), regulates the release of renin in the kidney. Secondly, Ang II and AVP, acting on AT1R and V1aR, respectively, exert vasoconstriction, increase cardiac contractility, stimulate the sympathoadrenal system, and elevate blood pressure. At the same time, they act antagonistically in the regulation of blood pressure by baroreflex. Thirdly, the cooperative action of Ang II acting on AT1R and AVP stimulating both V1aR and V2 receptors in the kidney is necessary for the appropriate regulation of renal blood flow and the efficient resorption of sodium and water. Furthermore, both peptides enhance the release of aldosterone and potentiate its action in the renal tubules. Summary In this review, we (1) point attention to the role of the cooperative action of Ang II and AVP for the regulation of blood pressure and the water-electrolyte balance under physiological conditions, (2) present the subcellular mechanisms underlying interactions of these two peptides, and (3) provide evidence that dysregulation of the cooperative action of Ang II and AVP significantly contributes to the development of disturbances in the regulation of blood pressure and the water-electrolyte balance in cardiovascular diseases.
Collapse
Affiliation(s)
- Ewa Szczepanska-Sadowska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland.
| | - Katarzyna Czarzasta
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| | - Agnieszka Cudnoch-Jedrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| |
Collapse
|
138
|
Schreier B, Hünerberg M, Mildenberger S, Rabe S, Bethmann D, Wickenhauser C, Gekle M. Deletion of the EGF receptor in vascular smooth muscle cells prevents chronic angiotensin II-induced arterial wall stiffening and media thickening. Acta Physiol (Oxf) 2018; 222. [PMID: 29152859 DOI: 10.1111/apha.12996] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 11/09/2017] [Accepted: 11/10/2017] [Indexed: 01/13/2023]
Abstract
AIM In vivo vascular smooth muscle cell (VSMC) EGF receptor (EGFR) contributes to acute angiotensin II (AII) effects on vascular tone and blood pressure. The ubiquitously expressed EGFR has been implicated in vascular remodelling preceding end-organ damage by pharmacological inhibition, and AII signalling in cultured vascular cells is partly EGFR-dependent. However, the role of VSMC-EGFR in vivo during AII-induced pathophysiological processes is not known. METHODS This study assesses the in vivo relevance of VSMC-EGFR during chronic AII challenge without further stressors, using a mouse model with inducible, VSMC-specific EGFR knock out (VSMC-EGFR-KO). In these mice functional and structural vascular, renal and cardiac effects or biomarkers were investigated in vivo and ex vivo. RESULTS Vascular smooth muscle cell-EGFR-KO prevented AII-induced media hypertrophy of mesenteric arteries, renal arterioles and the aorta, VSMC ERK1/2-phosphorylation as well as the impairment of vascular compliance. Furthermore, induction of vascular fibrosis, creatinineamia, renal interstitial fibrosis as well as the increase in fractional water excretion was prevented. AII-induced increase in systolic blood pressure was mitigated. By contrast, endothelial dysfunction, induction of vascular inflammatory marker mRNA and albuminuria were not inhibited. Cardiac and cardiomyocyte hypertrophy were also not prevented by VSMC-EGFR-KO. CONCLUSION Vascular smooth muscle cell-EGFRs are relevant for pathological AII action in vivo. Our data show in vivo and ex vivo the necessity of VSMC-EGFR for AII-induced structural and functional vascular remodelling, not including endothelial dysfunction. Hereby, VSMC-EGFR gains importance for complete AII-induced renal end-organ damage succeeding vascular remodelling.
Collapse
Affiliation(s)
- B. Schreier
- Julius-Bernstein-Institute of Physiology; Martin Luther University Halle-Wittenberg; Halle Germany
| | - M. Hünerberg
- Julius-Bernstein-Institute of Physiology; Martin Luther University Halle-Wittenberg; Halle Germany
| | - S. Mildenberger
- Julius-Bernstein-Institute of Physiology; Martin Luther University Halle-Wittenberg; Halle Germany
| | - S. Rabe
- Julius-Bernstein-Institute of Physiology; Martin Luther University Halle-Wittenberg; Halle Germany
| | - D. Bethmann
- Institute of Pathology; Martin Luther University Halle-Wittenberg; Halle Germany
| | - C. Wickenhauser
- Institute of Pathology; Martin Luther University Halle-Wittenberg; Halle Germany
| | - M. Gekle
- Julius-Bernstein-Institute of Physiology; Martin Luther University Halle-Wittenberg; Halle Germany
| |
Collapse
|
139
|
Schmidt S, Schumacher N, Schwarz J, Tangermann S, Kenner L, Schlederer M, Sibilia M, Linder M, Altendorf-Hofmann A, Knösel T, Gruber ES, Oberhuber G, Bolik J, Rehman A, Sinha A, Lokau J, Arnold P, Cabron AS, Zunke F, Becker-Pauly C, Preaudet A, Nguyen P, Huynh J, Afshar-Sterle S, Chand AL, Westermann J, Dempsey PJ, Garbers C, Schmidt-Arras D, Rosenstiel P, Putoczki T, Ernst M, Rose-John S. ADAM17 is required for EGF-R-induced intestinal tumors via IL-6 trans-signaling. J Exp Med 2018; 215:1205-1225. [PMID: 29472497 PMCID: PMC5881468 DOI: 10.1084/jem.20171696] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 12/22/2017] [Accepted: 01/22/2018] [Indexed: 02/06/2023] Open
Abstract
Schmidt et al. show that loss of the membrane-bound metalloprotease ADAM17 led to impaired intestinal cancer development in the murine APCmin/+ model, which also depended on IL-6 trans-signaling via the soluble IL-6R and could be blocked by the specific IL-6 trans-signaling inhibitor sgp130Fc. Colorectal cancer is treated with antibodies blocking epidermal growth factor receptor (EGF-R), but therapeutic success is limited. EGF-R is stimulated by soluble ligands, which are derived from transmembrane precursors by ADAM17-mediated proteolytic cleavage. In mouse intestinal cancer models in the absence of ADAM17, tumorigenesis was almost completely inhibited, and the few remaining tumors were of low-grade dysplasia. RNA sequencing analysis demonstrated down-regulation of STAT3 and Wnt pathway components. Because EGF-R on myeloid cells, but not on intestinal epithelial cells, is required for intestinal cancer and because IL-6 is induced via EGF-R stimulation, we analyzed the role of IL-6 signaling. Tumor formation was equally impaired in IL-6−/− mice and sgp130Fc transgenic mice, in which only trans-signaling via soluble IL-6R is abrogated. ADAM17 is needed for EGF-R–mediated induction of IL-6 synthesis, which via IL-6 trans-signaling induces β-catenin–dependent tumorigenesis. Our data reveal the possibility of a novel strategy for treatment of colorectal cancer that could circumvent intrinsic and acquired resistance to EGF-R blockade.
Collapse
Affiliation(s)
- Stefanie Schmidt
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Neele Schumacher
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Jeanette Schwarz
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Simone Tangermann
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine, Vienna, Austria
| | - Lukas Kenner
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine, Vienna, Austria.,Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Department of Experimental and Laboratory Animal Pathology, Medical University Vienna, Vienna, Austria
| | - Michaela Schlederer
- Department of Experimental and Laboratory Animal Pathology, Medical University Vienna, Vienna, Austria
| | - Maria Sibilia
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Comprehensive Cancer Center, Vienna, Austria
| | - Markus Linder
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Comprehensive Cancer Center, Vienna, Austria
| | | | - Thomas Knösel
- Institute of Pathology, Ludwig-Maximilians-University, Munich, Germany
| | - Elisabeth S Gruber
- Department of General Surgery, Division of Surgery and Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| | - Georg Oberhuber
- Department of Experimental and Laboratory Animal Pathology, Medical University Vienna, Vienna, Austria
| | - Julia Bolik
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Ateequr Rehman
- Institute of Clinical Molecular Biology, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Anupam Sinha
- Institute of Clinical Molecular Biology, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Juliane Lokau
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Philipp Arnold
- Anatomisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Anne-Sophie Cabron
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Friederike Zunke
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | | | - Adele Preaudet
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Paul Nguyen
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Jennifer Huynh
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Shoukat Afshar-Sterle
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Ashwini L Chand
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | | | - Peter J Dempsey
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Christoph Garbers
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Dirk Schmidt-Arras
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian Albrechts Universität Kiel, Kiel, Germany
| | - Tracy Putoczki
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Stefan Rose-John
- Biochemisches Institut, Christian Albrechts Universität Kiel, Kiel, Germany
| |
Collapse
|
140
|
Chenodeoxycholic acid activates NLRP3 inflammasome and contributes to cholestatic liver fibrosis. Oncotarget 2018; 7:83951-83963. [PMID: 27924062 PMCID: PMC5356637 DOI: 10.18632/oncotarget.13796] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 11/22/2016] [Indexed: 12/20/2022] Open
Abstract
Accumulation of hydrophobic bile acids in the liver contributes to cholestatic liver injury. Inflammation induced by excessive bile acids is believed to play a crucial role, however, the mechanisms of bile acids triggered inflammatory response remain unclear. Recent studies have highlighted the effect of NLRP3 inflammasome in mediating liver inflammation and fibrosis. In this study, we for the first time showed that chenodeoxycholic acid (CDCA), the major hydrophobic primary bile acid involved in cholestatic liver injury, could dose-dependently induce NLRP3 inflammasome activation and secretion of pro-inflammatory cytokine-IL-1β in macrophages by promoting ROS production and K+ efflux. Mechanistically, CDCA triggered ROS formation in part through TGR5/EGFR downstream signaling, including protein kinase B, extracellular regulated protein kinases and c-Jun N-terminal kinase pathways. Meanwhile, CDCA also induced ATP release from macrophages which subsequently causes K+ efflux via P2X7 receptor. Furthermore, in vivo inhibition of NLRP3 inflammasome with caspase-1 inhibitor dramatically decreased mature IL-1β level of liver tissue and ameliorated liver fibrosis in bile duct ligation (BDL) mouse model. In conclusion, excessive CDCA may represent an endogenous danger signal to activate NLRP3 inflammasome and initiate liver inflammation during cholestasis. Our finding offers a mechanistic basis to ameliorate cholestatic liver fibrosis by targeting inflammasome activation.
Collapse
|
141
|
Barton M, Filardo EJ, Lolait SJ, Thomas P, Maggiolini M, Prossnitz ER. Twenty years of the G protein-coupled estrogen receptor GPER: Historical and personal perspectives. J Steroid Biochem Mol Biol 2018; 176:4-15. [PMID: 28347854 PMCID: PMC5716468 DOI: 10.1016/j.jsbmb.2017.03.021] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/22/2017] [Accepted: 03/23/2017] [Indexed: 12/24/2022]
Abstract
Estrogens play a critical role in many aspects of physiology, particularly female reproductive function, but also in pathophysiology, and are associated with protection from numerous diseases in premenopausal women. Steroids and the effects of estrogen have been known for ∼90 years, with the first evidence for a receptor for estrogen presented ∼50 years ago. The original ancestral steroid receptor, extending back into evolution more than 500 million years, was likely an estrogen receptor, whereas G protein-coupled receptors (GPCRs) trace their origins back into history more than one billion years. The classical estrogen receptors (ERα and ERβ) are ligand-activated transcription factors that confer estrogen sensitivity upon many genes. It was soon apparent that these, or novel receptors may also be responsible for the "rapid"/"non-genomic" membrane-associated effects of estrogen. The identification of an orphan GPCR (GPR30, published in 1996) opened a new field of research with the description in 2000 that GPR30 expression is required for rapid estrogen signaling. In 2005-2006, the field was greatly stimulated by two studies that described the binding of estrogen to GPR30-expressing cell membranes, followed by the identification of a GPR30-selective agonist (that lacked binding and activity towards ERα and ERβ). Renamed GPER (G protein-coupled estrogen receptor) by IUPHAR in 2007, the total number of articles in PubMed related to this receptor recently surpassed 1000. In this article, the authors present personal perspectives on how they became involved in the discovery and/or advancement of GPER research. These areas include non-genomic effects on vascular tone, receptor cloning, molecular and cellular biology, signal transduction mechanisms and pharmacology of GPER, highlighting the roles of GPER and GPER-selective compounds in diseases such as obesity, diabetes, and cancer and the obligatory role of GPER in propagating cardiovascular aging, arterial hypertension and heart failure through the stimulation of Nox expression.
Collapse
Affiliation(s)
- Matthias Barton
- Molecular Internal Medicine, University of Zürich, 8057 Zürich, Switzerland.
| | - Edward J Filardo
- Rhode Island Hospital, Brown University, Providence, RI 02903, USA
| | - Stephen J Lolait
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Peter Thomas
- Marine Science Institute, University of Texas at Austin, Port Aransas, TX 78373, USA
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Eric R Prossnitz
- Department of Internal Medicine, University of New Mexico Health Sciences Center and University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
142
|
Abstract
Emerging evidence points to a strong association between the gut microbiota and the risk, development and progression of gastrointestinal cancers such as colorectal cancer (CRC) and hepatocellular carcinoma (HCC). Bile acids, produced in the liver, are metabolized by enzymes derived from intestinal bacteria and are critically important for maintaining a healthy gut microbiota, balanced lipid and carbohydrate metabolism, insulin sensitivity and innate immunity. Given the complexity of bile acid signalling and the direct biochemical interactions between the gut microbiota and the host, a systems biology perspective is required to understand the liver-bile acid-microbiota axis and its role in gastrointestinal carcinogenesis to reverse the microbiota-mediated alterations in bile acid metabolism that occur in disease states. An examination of recent research progress in this area is urgently needed. In this Review, we discuss the mechanistic links between bile acids and gastrointestinal carcinogenesis in CRC and HCC, which involve two major bile acid-sensing receptors, farnesoid X receptor (FXR) and G protein-coupled bile acid receptor 1 (TGR5). We also highlight the strategies and cutting-edge technologies to target gut-microbiota-dependent alterations in bile acid metabolism in the context of cancer therapy.
Collapse
Affiliation(s)
- Wei Jia
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology & Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
- University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, Hawaii 96813, USA
| | - Guoxiang Xie
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology & Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
- University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, Hawaii 96813, USA
| | - Weiping Jia
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology & Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
143
|
Prossnitz ER. GPER modulators: Opportunity Nox on the heels of a class Akt. J Steroid Biochem Mol Biol 2018; 176:73-81. [PMID: 28285016 PMCID: PMC5591048 DOI: 10.1016/j.jsbmb.2017.03.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 12/14/2022]
Abstract
The (patho)physiology of estrogen and its receptors is complex. It is therefore not surprising that therapeutic approaches targeting this hormone include stimulation of its activity through supplementation with either the hormone itself or natural or synthetic agonists, inhibition of its activity through the use of antagonists or inhibitors of its synthesis, and tissue-selective modulation of its activity with biased ligands. The physiology of this hormone is further complicated by the existence of at least three receptors, the classical nuclear estrogen receptors α and β (ERα and ERβ), and the 7-transmembrane G protein-coupled estrogen receptor (GPER/GPR30), with overlapping but distinct pharmacologic profiles, particularly of anti-estrogenic ligands. GPER-selective ligands, as well as GPER knockout mice, have greatly aided our understanding of the physiological roles of GPER. Such ligands have revealed that GPER activation mediates many of the rapid cellular signaling events (including Ca2+ mobilization, ERK and PI3K/Akt activation) associated with estrogen activity, as opposed to the nuclear ERs that are traditionally described to function as ligand-induced transcriptional factors. Many of the salutary effects of estrogen throughout the body are reproduced by the GPER-selective agonist G-1, which, owing to its minimal effects on reproductive tissues, can be considered a non-feminizing estrogenic compound, and thus of potential therapeutic use in both women and men. On the contrary, until recently GPER-selective antagonists had predominantly found preclinical application in cancer models where estrogen stimulates cell growth and survival. This viewpoint changed recently with the discovery that GPER is associated with aging, particularly that of the cardiovascular system, where the GPER antagonist G36 reduced hypertension and GPER deficiency prevented cardiac fibrosis and vascular dysfunction with age, through the downregulation of Nox1 and as a consequence superoxide production. Thus, similar to the classical ERs, both agonists and antagonists of GPER may be of therapeutic benefit depending on the disease or condition to be treated.
Collapse
Affiliation(s)
- Eric R Prossnitz
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, United States.
| |
Collapse
|
144
|
Yu X, Stallone JN, Heaps CL, Han G. The activation of G protein-coupled estrogen receptor induces relaxation via cAMP as well as potentiates contraction via EGFR transactivation in porcine coronary arteries. PLoS One 2018; 13:e0191418. [PMID: 29360846 PMCID: PMC5779678 DOI: 10.1371/journal.pone.0191418] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 01/04/2018] [Indexed: 01/09/2023] Open
Abstract
Estrogen exerts protective effects against cardiovascular diseases in premenopausal women, but is associated with an increased risk of both coronary heart disease and stroke in older postmenopausal women. Studies have shown that activation of the G-protein-coupled estrogen receptor 1 (GPER) can cause either relaxation or contraction of arteries. It is highly likely that these dual actions of GPER may contribute to the seemingly paradoxical effects of estrogen in regulating coronary artery function. The objective of this study was to test the hypothesis that activation of GPER enhances agonist-stimulated porcine coronary artery contraction via epidermal growth factor receptor (EGFR) transactivation and its downstream extracellular signal-regulated kinases (ERK1/2) pathway. Isometric tension studies and western blot were performed to determine the effect of GPER activation on coronary artery contraction. Our findings demonstrated that G-1 caused concentration-dependent relaxation of ET-1-induced contraction, while pretreatment of arterial rings with G-1 significantly enhanced ET-1-induced contraction. GPER antagonist, G-36, significantly inhibited both the G-1-induced relaxation effect and G-1-enhanced ET-1 contraction. Gallein, a Gβγ inhibitor, significantly increased G-1-induced relaxation, yet inhibited G-1-enhanced ET-1-mediated contraction. Similarly, inhibition of EGFR with AG1478 or inhibition of Src with phosphatase 2 further increased G-1-induced relaxation responses in coronary arteries, but decreased G-1-enhanced ET-1-induced contraction. Western blot experiments in porcine coronary artery smooth muscle cells (PCASMC) showed that G-1 increased tyrosine phosphorylation of EGFR, which was inhibited by AG-1478. Furthermore, enzyme-linked immunosorbent assays showed that the level of heparin-binding EGF (HB-EGF) released by ET-1 treatment increased two-fold; whereas pre-incubation with G-1 further increased ET-1-induced HB-EGF release to four-fold over control conditions. Lastly, the role of ERK1/2 was determined by applying the MEK inhibitor, PD98059, in isometric tension studies and detecting phospho-ERK1/2 in immunoblotting. PD98059 potentiated G-1-induced relaxation response, but blocked G-1-enhanced ET-1-induced contraction. By western blot, G-1 treatment decreased phospho-ERK1/2, however, in the presence of the adenylyl cyclase inhibitor, SQ22536, G-1 significantly increased ERK1/2 phosphorylation in PCASMC. These data demonstrate that activation of GPER induces relaxation via cAMP as well as contraction via a mechanism involving transactivation of EGFR and the phosphorylation of ERK1/2 in porcine coronary arteries.
Collapse
Affiliation(s)
- Xuan Yu
- Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| | - John N. Stallone
- Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
- Women's Health Division, Michael E. DeBakey Institute Texas A&M University, College Station, TX, United States of America
| | - Cristine L. Heaps
- Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
- Women's Health Division, Michael E. DeBakey Institute Texas A&M University, College Station, TX, United States of America
| | - Guichun Han
- Veterinary Physiology & Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
- Women's Health Division, Michael E. DeBakey Institute Texas A&M University, College Station, TX, United States of America
- * E-mail:
| |
Collapse
|
145
|
Haxho F, Neufeld RJ, Szewczuk MR. Neuraminidase-1: a novel therapeutic target in multistage tumorigenesis. Oncotarget 2018; 7:40860-40881. [PMID: 27029067 PMCID: PMC5130050 DOI: 10.18632/oncotarget.8396] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 03/18/2016] [Indexed: 12/15/2022] Open
Abstract
Several of the growth factors and their receptor tyrosine kinases (RTK) such as epidermal growth factor (EGF), platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), nerve growth factor (NGF) and insulin are promising candidate targets for cancer therapy. Indeed, tyrosine kinase inhibitors (TKI) have been developed to target these growth factors and their receptors, and have demonstrated dramatic initial responses in cancer therapy. Yet, most patients ultimately develop TKI drug resistance and relapse. It is essential in the clinical setting that the targeted therapies are to circumvent multistage tumorigenesis, including genetic mutations at the different growth factor receptors, tumor neovascularization, chemoresistance of tumors, immune-mediated tumorigenesis and the development of tissue invasion and metastasis. Here, we identify a novel receptor signaling platform linked to EGF, NGF, insulin and TOLL-like receptor (TLR) activations, all of which are known to play major roles in tumorigenesis. The importance of these findings signify an innovative and promising entirely new targeted therapy for cancer. The role of mammalian neuraminidase-1 (Neu1) in complex with matrix metalloproteinase-9 and G protein-coupled receptor tethered to RTKs and TLRs is identified as a major target in multistage tumorigenesis. Evidence exposing the link connecting growth factor-binding and immune-mediated tumorigenesis to this novel receptor-signaling paradigm will be reviewed in its current relationship to cancer.
Collapse
Affiliation(s)
- Fiona Haxho
- Departments of Biomedical and Molecular Sciences, Kingston, Ontario, Canada
| | - Ronald J Neufeld
- Department of Chemical Engineering, Queen's University, Kingston, Ontario, Canada
| | - Myron R Szewczuk
- Departments of Biomedical and Molecular Sciences, Kingston, Ontario, Canada
| |
Collapse
|
146
|
Oyanadel C, Holmes C, Pardo E, Retamal C, Shaughnessy R, Smith P, Cortés P, Bravo-Zehnder M, Metz C, Feuerhake T, Romero D, Roa JC, Montecinos V, Soza A, González A. Galectin-8 induces partial epithelial-mesenchymal transition with invasive tumorigenic capabilities involving a FAK/EGFR/proteasome pathway in Madin-Darby canine kidney cells. Mol Biol Cell 2018; 29:557-574. [PMID: 29298841 PMCID: PMC6004583 DOI: 10.1091/mbc.e16-05-0301] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 12/07/2017] [Accepted: 12/27/2017] [Indexed: 12/22/2022] Open
Abstract
Epithelial cells can acquire invasive and tumorigenic capabilities through epithelial–mesenchymal-transition (EMT). The glycan-binding protein galectin-8 (Gal-8) activates selective β1-integrins involved in EMT and is overexpressed by certain carcinomas. Here we show that Gal-8 overexpression or exogenous addition promotes proliferation, migration, and invasion in nontumoral Madin–Darby canine kidney (MDCK) cells, involving focal-adhesion kinase (FAK)-mediated transactivation of the epidermal growth factor receptor (EGFR), likely triggered by α5β1integrin binding. Under subconfluent conditions, Gal-8–overexpressing MDCK cells (MDCK-Gal-8H) display hallmarks of EMT, including decreased E-cadherin and up-regulated expression of vimentin, fibronectin, and Snail, as well as increased β-catenin activity. Changes related to migration/invasion included higher expression of α5β1 integrin, extracellular matrix-degrading MMP13 and urokinase plasminogen activator/urokinase plasminogen activator receptor (uPA/uPAR) protease systems. Gal-8–stimulated FAK/EGFR pathway leads to proteasome overactivity characteristic of cancer cells. Yet MDCK-Gal-8H cells still develop apical/basolateral polarity reverting EMT markers and proteasome activity under confluence. This is due to the opposite segregation of Gal-8 secretion (apical) and β1-integrins distribution (basolateral). Strikingly, MDCK-Gal-8H cells acquired tumorigenic potential, as reflected in anchorage-independent growth in soft agar and tumor generation in immunodeficient NSG mice. Therefore, Gal-8 can promote oncogenic-like transformation of epithelial cells through partial and reversible EMT, accompanied by higher proliferation, migration/invasion, and tumorigenic properties.
Collapse
Affiliation(s)
- Claudia Oyanadel
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina, Universidad San Sebastián, 7510156 Santiago, Chile.,Fundación Ciencia y Vida, 7780272 Santiago, Chile
| | - Christopher Holmes
- Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330023 Santiago, Chile
| | - Evelyn Pardo
- Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330023 Santiago, Chile
| | - Claudio Retamal
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina, Universidad San Sebastián, 7510156 Santiago, Chile.,Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330023 Santiago, Chile
| | - Ronan Shaughnessy
- Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330023 Santiago, Chile
| | - Patricio Smith
- Unidad de Odontología, Pontificia Universidad Católica de Chile, 8330023 Santiago, Chile
| | - Priscilla Cortés
- Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330023 Santiago, Chile
| | - Marcela Bravo-Zehnder
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina, Universidad San Sebastián, 7510156 Santiago, Chile.,Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330023 Santiago, Chile
| | - Claudia Metz
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina, Universidad San Sebastián, 7510156 Santiago, Chile.,Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330023 Santiago, Chile
| | - Teo Feuerhake
- Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330023 Santiago, Chile
| | - Diego Romero
- Departamento de Patología, Pontificia Universidad Católica de Chile, 8330023 Santiago, Chile
| | - Juan Carlos Roa
- Departamento de Patología, Pontificia Universidad Católica de Chile, 8330023 Santiago, Chile
| | - Viviana Montecinos
- Departamento de Hematología y Oncología, Facultad de Medicina, Pontificia Universidad Católica de Chile, 8330023 Santiago, Chile
| | - Andrea Soza
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina, Universidad San Sebastián, 7510156 Santiago, Chile .,Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330023 Santiago, Chile
| | - Alfonso González
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina, Universidad San Sebastián, 7510156 Santiago, Chile .,Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330023 Santiago, Chile
| |
Collapse
|
147
|
Girgert R, Emons G, Gründker C. Estrogen Signaling in ERα-Negative Breast Cancer: ERβ and GPER. Front Endocrinol (Lausanne) 2018; 9:781. [PMID: 30687231 PMCID: PMC6333678 DOI: 10.3389/fendo.2018.00781] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 12/12/2018] [Indexed: 01/22/2023] Open
Abstract
Estrogen receptors are important regulators of the growth of breast tumors. Three different receptors for estrogens have been identified in breast tumors, two nuclear receptors, ERα and ERβ, and a G-protein coupled estrogen receptor 1 (GPER) that initiates non-genomic effects of estrogens in the cytosol. Recent findings show that the stimulation of cytoplasmic ERα and ERβ also triggers non-genomic signaling pathways. The treatment of breast cancer with anti-estrogens depends on the presence of ERα. About 40% of all breast cancers, however, do not express ERα. One subgroup of these tumors overexpress Her-2, another important group is designated as triple-negative breast cancer, as they neither express ERα, nor progesterone receptors, nor do they overexpress Her-2. This review addresses the signaling of ERβ and GPER in ERα-negative breast tumors. In addition to the well-established EGF-receptor transactivation pathways of GPER, more recent findings of GPER-dependent activation of FOXO3a, the Hippo-pathway, and HOTAIR-activation are summarized.
Collapse
|
148
|
Inoue Y, Shimazawa M, Nakamura S, Takata S, Hashimoto Y, Izawa H, Masuda T, Tsuruma K, Sakaue T, Nakayama H, Higashiyama S, Hara H. Both Autocrine Signaling and Paracrine Signaling of HB-EGF Enhance Ocular Neovascularization. Arterioscler Thromb Vasc Biol 2017; 38:174-185. [PMID: 29191924 DOI: 10.1161/atvbaha.117.310337] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 11/17/2017] [Indexed: 12/27/2022]
Abstract
OBJECTIVE The incidence of blindness is increasing because of the increase in abnormal ocular neovascularization. Anti-VEGF (vascular endothelial growth factor) therapies have led to good results, although they are not a cure for the blindness. The purpose of this study was to determine what role HB-EGF (heparin-binding epidermal growth factor-like growth factor) plays in ocular angiogenesis. APPROACH AND RESULTS We examined the role played by HB-EGF in ocular neovascularization in 2 animal models of neovascularization: laser-induced choroidal neovascularization (CNV) and oxygen-induced retinopathy. We also studied human retinal microvascular endothelial cells in culture. Our results showed that the neovascularization was decreased in both the CNV and oxygen-induced retinopathy models in HB-EGF conditional knockout mice compared with that in wild-type mice. Moreover, the expressions of HB-EGF and VEGF were increased after laser-induced CNV and oxygen-induced retinopathy, and their expression sites were located around the neovascular areas. Exposure of human retinal microvascular endothelial cells to HB-EGF and VEGF increased their proliferation and migration, and CRM-197 (cross-reactive material-197), an HB-EGF inhibitor, decreased the HB-EGF-induced and VEGF-induced cell proliferation and migration. VEGF increased the expression of HB-EGF mRNA. VEGF-dependent activation of EGFR (epidermal growth factor receptor)/ERK1/2 (extracellular signal-regulated kinase 1/2) signaling and cell proliferation of endothelial cells required stimulation of the ADAM17 (a disintegrin and metalloprotease) and ADAM12. CRM-197 decreased the grades of the fluorescein angiograms and size of the CNV areas in marmoset monkeys. CONCLUSIONS These findings suggest that HB-EGF plays an important role in the development of CNV. Therefore, further investigations of HB-EGF are needed as a potential therapeutic target in the treatment of exudative age-related macular degeneration.
Collapse
Affiliation(s)
- Yuki Inoue
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Masamitsu Shimazawa
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Shinsuke Nakamura
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Shinsuke Takata
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Yuhei Hashimoto
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Hiroshi Izawa
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Tomomi Masuda
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Kazuhiro Tsuruma
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Tomohisa Sakaue
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Hironao Nakayama
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Shigeki Higashiyama
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.)
| | - Hideaki Hara
- From the Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Japan (Y.I., M.S., S.N., S.T., Y.H., H.I., T.M., K.T., H.H.); Proteo-Science Center, Division of Cell Growth and Tumor Regulation, Ehime University Shitsukawa, Toon, Japan (T.S., H.N., S.H.); and Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Japan (T.S., S.H.).
| |
Collapse
|
149
|
Decoding resistant hypertension signalling pathways. Clin Sci (Lond) 2017; 131:2813-2834. [PMID: 29184046 DOI: 10.1042/cs20171398] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/16/2017] [Accepted: 10/23/2017] [Indexed: 01/01/2023]
Abstract
Resistant hypertension (RH) is a clinical condition in which the hypertensive patient has become resistant to drug therapy and is often associated with increased cardiovascular morbidity and mortality. Several signalling pathways have been studied and related to the development and progression of RH: modulation of sympathetic activity by leptin and aldosterone, primary aldosteronism, arterial stiffness, endothelial dysfunction and variations in the renin-angiotensin-aldosterone system (RAAS). miRNAs comprise a family of small non-coding RNAs that participate in the regulation of gene expression at post-transcriptional level. miRNAs are involved in the development of both cardiovascular damage and hypertension. Little is known of the molecular mechanisms that lead to development and progression of this condition. This review aims to cover the potential roles of miRNAs in the mechanisms associated with the development and consequences of RH, and explore the current state of the art of diagnostic and therapeutic tools based on miRNA approaches.
Collapse
|
150
|
Ramírez-Reveco A, Villarroel-Espíndola F, Rodríguez-Gil JE, Concha II. Neuronal signaling repertoire in the mammalian sperm functionality. Biol Reprod 2017; 96:505-524. [PMID: 28339693 DOI: 10.1095/biolreprod.116.144154] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 01/24/2017] [Indexed: 12/14/2022] Open
Abstract
The common embryonic origin has been a recurrent explanation to understand the presence of "neural receptors" in sperm. However, this designation has conditioned a bias marked by the classical neurotransmission model, dismissing the possibility that neurotransmitters can play specific roles in the sperm function by themselves. For instance, the launching of acrosome reaction, a fundamental sperm function, includes several steps that recall the process of presynaptic secretion. Unlike of postsynaptic neuron, whose activation is mediated by molecular interaction between neurotransmitter and postsynaptic receptors, the oocyte activation is not mediated by receptors, but by cytosolic translocation of sperm phospholipase (PLCζ). Thus, the sperm has a cellular design to access and activate the oocyte and restore the ploidy of the species by an "allogenic pronuclear fusion." At subcellular level, the events controlling sperm function, particularly the capacitation process, are activated by chemical signals that trigger ion fluxes, sterol oxidation, synthesis of cyclic adenosine monophosphate, protein kinase A activation, tyrosine phosphorylations and calcium signaling, which correspond to second messengers similar to those associated with exocytosis and growth cone guidance in neurons. Classically, the sperm function associated with neural signals has been analyzed as a unidimensional approach (single ligand-receptor effect). However, the in vivo sperm are exposed to multidimensional signaling context, for example, the GABAergic, monoaminergic, purinergic, cholinergic, and melatoninergic, to name a few. The aim of this review is to present an overview of sperm functionality associated with "neuronal signaling" and possible cellular and molecular mechanisms involved in their regulation.
Collapse
Affiliation(s)
- Alfredo Ramírez-Reveco
- Instituto de Ciencia Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Franz Villarroel-Espíndola
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile.,Department of Pathology and Pediatric Pathology, Yale University, New Haven, Connecticut, USA
| | - Joan E Rodríguez-Gil
- Unitat de Reproducció Animal, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Ilona I Concha
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|