1
|
Lao-Peregrin C, Xiang G, Kim J, Srivastava I, Fall AB, Gerhard DM, Kohtala P, Kim D, Song M, Garcia-Marcos M, Levitz J, Lee FS. Synaptic plasticity via receptor tyrosine kinase/G-protein-coupled receptor crosstalk. Cell Rep 2024; 43:113595. [PMID: 38117654 PMCID: PMC10844890 DOI: 10.1016/j.celrep.2023.113595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/15/2023] [Accepted: 12/01/2023] [Indexed: 12/22/2023] Open
Abstract
Cellular signaling involves a large repertoire of membrane receptors operating in overlapping spatiotemporal regimes and targeting many common intracellular effectors. However, both the molecular mechanisms and the physiological roles of crosstalk between receptors, especially those from different superfamilies, are poorly understood. We find that the receptor tyrosine kinase (RTK) TrkB and the G-protein-coupled receptor (GPCR) metabotropic glutamate receptor 5 (mGluR5) together mediate hippocampal synaptic plasticity in response to brain-derived neurotrophic factor (BDNF). Activated TrkB enhances constitutive mGluR5 activity to initiate a mode switch that drives BDNF-dependent sustained, oscillatory Ca2+ signaling and enhanced MAP kinase activation. This crosstalk is mediated, in part, by synergy between Gβγ, released by TrkB, and Gαq-GTP, released by mGluR5, to enable physiologically relevant RTK/GPCR crosstalk.
Collapse
Affiliation(s)
| | - Guoqing Xiang
- Department of Psychiatry, Weill Cornell Medicine. New York, NY 10065, USA; Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Jihye Kim
- Department of Psychiatry, Weill Cornell Medicine. New York, NY 10065, USA
| | - Ipsit Srivastava
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Alexandra B Fall
- Department of Psychiatry, Weill Cornell Medicine. New York, NY 10065, USA
| | - Danielle M Gerhard
- Department of Psychiatry, Weill Cornell Medicine. New York, NY 10065, USA
| | - Piia Kohtala
- Department of Psychiatry, Weill Cornell Medicine. New York, NY 10065, USA
| | - Daegeon Kim
- Department of Life Sciences, Yeongnam University, Gyeongsan, Gyeongbuk 38451, South Korea
| | - Minseok Song
- Department of Life Sciences, Yeongnam University, Gyeongsan, Gyeongbuk 38451, South Korea
| | - Mikel Garcia-Marcos
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Joshua Levitz
- Department of Psychiatry, Weill Cornell Medicine. New York, NY 10065, USA; Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Francis S Lee
- Department of Psychiatry, Weill Cornell Medicine. New York, NY 10065, USA.
| |
Collapse
|
2
|
Lao-Peregrin C, Xiang G, Kim J, Srivastava I, Fall AB, Gerhard DM, Kohtala P, Kim D, Song M, Garcia-Marcos M, Levitz J, Lee FS. Synaptic plasticity via receptor tyrosine kinase/G protein-coupled receptor crosstalk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.28.555210. [PMID: 37693535 PMCID: PMC10491144 DOI: 10.1101/2023.08.28.555210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Cellular signaling involves a large repertoire of membrane receptors operating in overlapping spatiotemporal regimes and targeting many common intracellular effectors. However, both the molecular mechanisms and physiological roles of crosstalk between receptors, especially those from different superfamilies, are poorly understood. We find that the receptor tyrosine kinase (RTK), TrkB, and the G protein-coupled receptor (GPCR), metabotropic glutamate receptor 5 (mGluR5), together mediate a novel form of hippocampal synaptic plasticity in response to brain-derived neurotrophic factor (BDNF). Activated TrkB enhances constitutive mGluR5 activity to initiate a mode-switch that drives BDNF-dependent sustained, oscillatory Ca 2+ signaling and enhanced MAP kinase activation. This crosstalk is mediated, in part, by synergy between Gβγ, released by TrkB, and Gα q -GTP, released by mGluR5, to enable a previously unidentified form of physiologically relevant RTK/GPCR crosstalk.
Collapse
|
3
|
Khanmammadova N, Islam S, Sharma P, Amit M. Neuro-immune interactions and immuno-oncology. Trends Cancer 2023; 9:636-649. [PMID: 37258398 PMCID: PMC10524972 DOI: 10.1016/j.trecan.2023.05.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/24/2023] [Accepted: 05/08/2023] [Indexed: 06/02/2023]
Abstract
The nervous system is an important component of the tumor microenvironment (TME), driving tumorigenesis and tumor progression. Neuronal cues (e.g., neurotransmitters and neuropeptides) in the TME cause phenotypic changes in immune cells, such as increased exhaustion and inhibition of effector cells, which promote immune evasion and cancer progression. Two types of immune regulation by tumor-associated nerves are discussed in this review: regulation via neuronal stimuli (i.e., by neural transmission) and checkpoint-mediated neuronal immune regulation. The latter occurs via the expression of immune checkpoints on the membranes of intratumoral nerves and glial cells. Here, we summarize novel findings regarding the neuroimmune circuits in the tumor milieu, while emphasizing the potential targets of new and affordable anticancer therapeutic approaches.
Collapse
Affiliation(s)
- Narmina Khanmammadova
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shajedul Islam
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Padmanee Sharma
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Immunobiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Moran Amit
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The University of Texas Houston Health Science Center Graduate School of Biomedical Sciences, Department of Neuroscience, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
4
|
Aronowitz AL, Ali SR, Glaun MDE, Amit M. Acetylcholine in Carcinogenesis and Targeting Cholinergic Receptors in Oncology. Adv Biol (Weinh) 2022; 6:e2200053. [PMID: 35858206 DOI: 10.1002/adbi.202200053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/23/2022] [Indexed: 01/28/2023]
Abstract
Tumor cells modulate and are modulated by their microenvironments, which include the nervous system. Accumulating evidence links the overexpression and activity of nicotinic and muscarinic cholinergic receptor subtypes to tumorigenesis in breast, ovarian, prostate, gastric, pancreatic, and head and neck cancers. Nicotinic and muscarinic receptors have downstream factors are associated with angiogenesis, cell proliferation and migration, antiapoptotic signaling, and survival. Clinical trials analyzing the efficacy of various therapies targeting cholinergic signaling or downstream pathways of acetylcholine have shed promising light on novel cancer therapeutics. Although the evidence for cholinergic signaling involvement in tumor development is substantial, a more detailed understanding of the acetylcholine-induced mechanisms of tumorigenesis remains to be unlocked. Such an understanding would enable the development of clinical applications ranging from the identification of novel biomarkers to the utilization of existing drugs to modulate cholinergic signaling to the development of novel cancer therapies, as discussed in this review.
Collapse
Affiliation(s)
- Alexandra L Aronowitz
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,McGovern Medical School at UTHealth, Houston, TX, 77555, USA
| | - Shahrukh R Ali
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,The University of Texas Medical Branch, Galveston, TX, 77030, USA
| | - Mica D E Glaun
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,Department of Otolaryngology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Moran Amit
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
5
|
Dong L, Zhang RH, Zhou WD, Li YF, Li HY, Wu HT, Shi XH, Jonas JB, Wei WB. Epiregulin, epigen and betacellulin antibodies and axial elongation in young guinea pigs with lens-induced myopization. BMC Ophthalmol 2022; 22:193. [PMID: 35477375 PMCID: PMC9044769 DOI: 10.1186/s12886-022-02417-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/21/2022] [Indexed: 11/28/2022] Open
Abstract
Background To examine an effect of intravitreally applied antibodies against epidermal growth factor family members, namely epiregulin, epigen and betacellulin, on ocular axial elongation. Methods The experimental study included 30 guinea pigs (age:3–4 weeks) which underwent bilateral lens-induced myopization and received three intraocular injections of 20 µg of epiregulin antibody, epigen antibody and betacellulin antibody in weekly intervals into their right eyes, and of phosphate-buffered saline into their left eyes. Seven days after the last injection, the animals were sacrificed. Axial length was measured by sonographic biometry. Results At baseline, right eyes and left eyes did not differ (all P > 0.10) in axial length in neither group, nor did the interocular difference in axial length vary between the groups (P = 0.19). During the study period, right and left eyes elongated (P < 0.001) from 8.08 ± 0.07 mm to 8.59 ± 0.06 mm and from 8.08 ± 0.07 mm to 8.66 ± 0.07 mm, respectively. The interocular difference (left eye minus right eye) in axial elongation increased significantly in all three groups (epiregulin-antibody:from 0.03 ± 0.06 mm at one week after baseline to 0.16 ± 0.08 mm at three weeks after baseline;P = 0.001); epigen-antibody group:from -0.01 ± 0.06 mm to 0.06 ± 0.08 mm;P = 0.02; betacellulin antibody group:from -0.05 ± 0.05 mm to 0.02 ± 0.04 mm;P = 0.004). Correspondingly, interocular difference in axial length increased from -0.02 ± 0.04 mm to 0.13 ± 0.06 mm in the epiregulin-antibody group (P < 0.001), and from 0.01 ± 0.05 mm to 0.07 ± 0.05 mm in the epigen-antibody group (P = 0.045). In the betacellulin-antibody group the increase (0.01 ± 0.04 mm to 0.03 ± 0.03 mm) was not significant (P = 0.24). Conclusions The EGF family members epiregulin, epigen and betacellulin may be associated with axial elongation in young guinea pigs, with the effect decreasing from epiregulin to epigen and to betacellulin.
Collapse
Affiliation(s)
- Li Dong
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Rui-Heng Zhang
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Wen-Da Zhou
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yi-Fan Li
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - He-Yan Li
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hao-Tian Wu
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xu-Han Shi
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jost B Jonas
- Beijing Institute of Ophthalmology and Beijing Ophthalmology and Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China.,Department of Ophthalmology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland.,Privatpraxis Prof Jonas Und Dr Panda-Jonas, Heidelberg, Germany
| | - Wen-Bin Wei
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
6
|
Modeling Intestinal Stem Cell Function with Organoids. Int J Mol Sci 2021; 22:ijms222010912. [PMID: 34681571 PMCID: PMC8535974 DOI: 10.3390/ijms222010912] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/11/2022] Open
Abstract
Intestinal epithelial cells (IECs) are crucial for the digestive process and nutrient absorption. The intestinal epithelium is composed of the different cell types of the small intestine (mainly, enterocytes, goblet cells, Paneth cells, enteroendocrine cells, and tuft cells). The small intestine is characterized by the presence of crypt-villus units that are in a state of homeostatic cell turnover. Organoid technology enables an efficient expansion of intestinal epithelial tissue in vitro. Thus, organoids hold great promise for use in medical research and in the development of new treatments. At present, the cholinergic system involved in IECs and intestinal stem cells (ISCs) are attracting a great deal of attention. Thus, understanding the biological processes triggered by epithelial cholinergic activation by acetylcholine (ACh), which is produced and released from neuronal and/or non-neuronal tissue, is of key importance. Cholinergic signaling via ACh receptors plays a pivotal role in IEC growth and differentiation. Here, we discuss current views on neuronal innervation and non-neuronal control of the small intestinal crypts and their impact on ISC proliferation, differentiation, and maintenance. Since technology using intestinal organoid culture systems is advancing, we also outline an organoid-based organ replacement approach for intestinal diseases.
Collapse
|
7
|
Multiple Roles for Cholinergic Signaling from the Perspective of Stem Cell Function. Int J Mol Sci 2021; 22:ijms22020666. [PMID: 33440882 PMCID: PMC7827396 DOI: 10.3390/ijms22020666] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 01/11/2023] Open
Abstract
Stem cells have extensive proliferative potential and the ability to differentiate into one or more mature cell types. The mechanisms by which stem cells accomplish self-renewal provide fundamental insight into the origin and design of multicellular organisms. These pathways allow the repair of damage and extend organismal life beyond that of component cells, and they probably preceded the evolution of complex metazoans. Understanding the true nature of stem cells can only come from discovering how they are regulated. The concept that stem cells are controlled by particular microenvironments, also known as niches, has been widely accepted. Technical advances now allow characterization of the zones that maintain and control stem cell activity in several organs, including the brain, skin, and gut. Cholinergic neurons release acetylcholine (ACh) that mediates chemical transmission via ACh receptors such as nicotinic and muscarinic receptors. Although the cholinergic system is composed of organized nerve cells, the system is also involved in mammalian non-neuronal cells, including stem cells, embryonic stem cells, epithelial cells, and endothelial cells. Thus, cholinergic signaling plays a pivotal role in controlling their behaviors. Studies regarding this signal are beginning to unify our understanding of stem cell regulation at the cellular and molecular levels, and they are expected to advance efforts to control stem cells therapeutically. The present article reviews recent findings about cholinergic signaling that is essential to control stem cell function in a cholinergic niche.
Collapse
|
8
|
Louhivuori LM, Turunen PM, Louhivuori V, Al Rayyes I, Nordström T, Uhlén P, Åkerman KE. Neurotransmitters and Endothelins Acting on Radial Glial G-Protein-Coupled Receptors Are, Through Proteolytic NRG/ErbB4 Activation, Able to Modify the Migratory Behavior of Neocortical Cells and Mediate Bipolar-to-Multipolar Transition. Stem Cells Dev 2020; 29:1160-1177. [PMID: 31941419 DOI: 10.1089/scd.2019.0133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cell-cell communication plays a central role in the guidance of migrating neurons during the development of the cerebral cortex. Neuregulins (NRGs) are essential mediators for migration and maintenance of the radial glial scaffold. We show, in this study that soluble NRG reduces neuronal motility, causes transition of bipolar cells to multipolar ones, and induces neuronal mitosis. Blocking the NRG receptor, ErbB4, results in reduction of neuron-neuron and neuron-radial glial contacts and causes an increase in neuronal motility. Blocking the radial glial metabotropic glutamate receptor 5 (mGluR5), the nonselective cation channel transient receptor potential 3 (TRPC3), or matrix metalloproteinases (MMPs) results in similar effects as ErbB4 blockade. Soluble NRG counteract the changes in motility pattern. Stimulation of other radial glial G-protein-coupled receptors (GPCRs), such as muscarinic acetylcholine receptors or endothelin receptors counteract all the effect of mGluR5 blockade, but not that of ErbB4, TRPC3, and MMP blockade. The results indicate that neurotransmitters and endothelins acting on radial glial GPCRs are, through proteolytic NRG/ErbB4 activation, able to modify the migratory behavior of neurons.
Collapse
Affiliation(s)
- Lauri M Louhivuori
- Department of Physiology, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland.,Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Pauli M Turunen
- Department of Physiology, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Verna Louhivuori
- Department of Physiology, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Ibrahim Al Rayyes
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Tommy Nordström
- Department of Physiology, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Per Uhlén
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Karl E Åkerman
- Department of Physiology, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| |
Collapse
|
9
|
Dong L, Shi XH, Li YF, Jiang X, Wang YX, Lan YJ, Wu HT, Jonas JB, Wei WB. Blockade of epidermal growth factor and its receptor and axial elongation in experimental myopia. FASEB J 2020; 34:13654-13670. [PMID: 32799354 DOI: 10.1096/fj.202001095rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/18/2020] [Accepted: 07/29/2020] [Indexed: 01/10/2023]
Abstract
To examine the influence of epidermal growth factor (EGF) and its receptor (EGFR) on axial ocular elongation, we intraocularly injected an EGF antibody and an EGFR antibody into young guinea pigs with lens-induced axial elongation (myopization). Mean axial elongation was reduced in the eyes injected with the EGF/EGFR-antibody compared with the contralateral control eyes injected with PBS (phosphate-buffered solution) (0.43 ± 0.13 mm vs 0.53 ± 0.13 mm; P < .001). The intereye difference in axial length increased (P = .005) as the doses of the EGF antibody and EGFR antibody increased. As a corollary, the thickness of the retina at the posterior pole was dose-dependently increased in the injected eyes compared to the contralateral control eyes. Immunohistochemical staining for EGF and the relative mRNA expression of EGF and EGFR were the highest in eyes not injected with the EGF antibody or EGFR antibody and decreased (P < .05) as the dose of EGF antibody or EGFR antibody increased. In an in vitro study, EGF had a stimulating effect and the EGF antibody had an inhibitory effect on the proliferation and migration of RPE cells. The findings showed that the intravitreal application of an EGF antibody and EGFR antibody is associated with a dose-dependent reduction in lens-induced axial elongation in young guinea pigs. The EGFR family may play a role in axial elongation of the eye and in the development of myopia.
Collapse
Affiliation(s)
- Li Dong
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xu Han Shi
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yi Fan Li
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xue Jiang
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ya Xing Wang
- Beijing Institute of Ophthalmology and Beijing Ophthalmology and Visual Science Key Lab, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yin Jun Lan
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hao Tian Wu
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jost B Jonas
- Department of Ophthalmology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Wen Bin Wei
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Ophthalmology & Visual Sciences Key Lab, Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Estadella I, Pedrós-Gámez O, Colomer-Molera M, Bosch M, Sorkin A, Felipe A. Endocytosis: A Turnover Mechanism Controlling Ion Channel Function. Cells 2020; 9:E1833. [PMID: 32759790 PMCID: PMC7463639 DOI: 10.3390/cells9081833] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/30/2020] [Accepted: 08/02/2020] [Indexed: 01/08/2023] Open
Abstract
Ion channels (IChs) are transmembrane proteins that selectively drive ions across membranes. The function of IChs partially relies on their abundance and proper location in the cell, fine-tuned by the delicate balance between secretory, endocytic, and degradative pathways. The disruption of this balance is associated with several diseases, such as Liddle's and long QT syndromes. Because of the vital role of these proteins in human health and disease, knowledge of ICh turnover is essential. Clathrin-dependent and -independent mechanisms have been the primary mechanisms identified with ICh endocytosis and degradation. Several molecular determinants recognized by the cellular internalization machinery have been discovered. Moreover, specific conditions can trigger the endocytosis of many IChs, such as the activation of certain receptors, hypokalemia, and some drugs. Ligand-dependent receptor activation primarily results in the posttranslational modification of IChs and the recruitment of important mediators, such as β-arrestins and ubiquitin ligases. However, endocytosis is not a final fate. Once internalized into endosomes, IChs are either sorted to lysosomes for degradation or recycled back to the plasma membrane. Rab proteins are crucial participants during these turnover steps. In this review, we describe the major ICh endocytic pathways, the signaling inputs triggering ICh internalization, and the key mediators of this essential cellular process.
Collapse
Affiliation(s)
- Irene Estadella
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain; (I.E.); (O.P.-G.); (M.C.-M.); (M.B.)
| | - Oriol Pedrós-Gámez
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain; (I.E.); (O.P.-G.); (M.C.-M.); (M.B.)
| | - Magalí Colomer-Molera
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain; (I.E.); (O.P.-G.); (M.C.-M.); (M.B.)
| | - Manel Bosch
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain; (I.E.); (O.P.-G.); (M.C.-M.); (M.B.)
- Centres Científics i Tecnològics de la Universitat de Barcelona (CCiTUB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - Alexander Sorkin
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Antonio Felipe
- Molecular Physiology Laboratory, Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, 08028 Barcelona, Spain; (I.E.); (O.P.-G.); (M.C.-M.); (M.B.)
| |
Collapse
|
11
|
Dong L, Shi XH, Kang YK, Wei WB, Wang YX, Xu XL, Gao F, Yuan LH, Zhen J, Jiang WJ, Jonas JB. Amphiregulin and ocular axial length. Acta Ophthalmol 2019; 97:e460-e470. [PMID: 30860674 DOI: 10.1111/aos.14080] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 02/08/2019] [Accepted: 02/10/2019] [Indexed: 12/15/2022]
Abstract
PURPOSE To assess the potential role of amphiregulin as messenger molecule in ocular axial elongation. METHODS The experimental study included guinea pigs (total n = 78) (age: 3-4 weeks) which underwent bilateral lens-induced myopization and received 15 days later three intraocular injections in weekly intervals of amphiregulin antibody (doses:5 μg, 10 μg, 20 μg) into their right eyes, and three phosphate-buffered saline injections into their left eyes; and guinea pigs without lens-induced myopization and which received three unilateral intraocular injections of amphiregulin antibody (dose: 20 μg) or amphiregulin (doses: 1 ng; 10 ng; 20 ng) into their right eyes, and three phosphate-buffered saline injections into their left eyes. Seven days later, the animals were sacrificed. Intravitally, we performed biometry, and histology and immunohistochemistry post-mortem. RESULTS In animals with bilateral lens-induced myopization, the right eyes receiving amphiregulin antibody showed reduced axial elongation in a dose-dependent manner (dose: 5 μg: side difference: 0.14 ± 0.05 mm;10 μg: 0.22 ± 0.06 mm; 20 μg: 0.32 ± 0.06 mm; p < 0.001), thicker sclera (all p < 0.05) and higher cell density in the retinal nuclear layers and retinal pigment epithelium (RPE) (all p < 0.05). In animals without lens-induced myopia, the right eyes with amphiregulin antibody application (20 μg) showed reduced axial elongation (p = 0.04), and the right eyes with amphiregulin injections experienced increased (p = 0.02) axial elongation in a dose-dependent manner (1 ng: 0.04 ± 0.06 mm; 10 ng: 0.10 ± 0.05 mm; 20 ng: 0.11 ± 0.06 mm). Eyes with lens-induced axial elongation as compared to eyes without lens-induced axial elongation revealed an increased visualization of amphiregulin upon immunohistochemistry and higher expression of mRNA of endogenous amphiregulin and epidermal growth factor receptor, in particular in the outer part of the retinal inner nuclear layer and in the RPE. CONCLUSION Amphiregulin may be associated with axial elongation in young guinea pigs.
Collapse
Affiliation(s)
- Li Dong
- Beijing Tongren Eye Center Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment Beijing Ophthalmology & Visual Sciences Key Lab Beijing Tongren Hospital Capital Medical University Beijing China
| | - Xu Han Shi
- Beijing Tongren Eye Center Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment Beijing Ophthalmology & Visual Sciences Key Lab Beijing Tongren Hospital Capital Medical University Beijing China
| | - Yi Kun Kang
- Department of Oncology Beijing Chao‐Yang Hospital Capital Medical University Beijing China
| | - Wen Bin Wei
- Beijing Tongren Eye Center Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment Beijing Ophthalmology & Visual Sciences Key Lab Beijing Tongren Hospital Capital Medical University Beijing China
| | - Ya Xing Wang
- Beijing Institute of Ophthalmology and Beijing Ophthalmology and Visual Science Key Lab Beijing Tongren Eye Center Beijing Tongren Hospital Capital Medical University Beijing China
| | - Xiao Lin Xu
- Beijing Tongren Eye Center Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment Beijing Ophthalmology & Visual Sciences Key Lab Beijing Tongren Hospital Capital Medical University Beijing China
- Beijing Institute of Ophthalmology and Beijing Ophthalmology and Visual Science Key Lab Beijing Tongren Eye Center Beijing Tongren Hospital Capital Medical University Beijing China
| | - Fei Gao
- Beijing Institute of Ophthalmology and Beijing Ophthalmology and Visual Science Key Lab Beijing Tongren Eye Center Beijing Tongren Hospital Capital Medical University Beijing China
| | - Lin Hong Yuan
- Department of Nutrition and Food Hygiene School of Public Health Capital Medical University Beijing China
| | - Jie Zhen
- Department of Nutrition and Food Hygiene School of Public Health Capital Medical University Beijing China
| | - Wen Jun Jiang
- Eye Institute of Shandong University of Traditional Chinese Medicine Jinan Shandong China
| | - Jost B. Jonas
- Department of Ophthalmology Medical Faculty Mannheim of the Ruprecht‐Karls‐University Heidelberg Mannheim Germany
| |
Collapse
|
12
|
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: 88] [Impact Index Per Article: 12.6] [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
|
13
|
Sajda T, Sinha AA. Autoantibody Signaling in Pemphigus Vulgaris: Development of an Integrated Model. Front Immunol 2018; 9:692. [PMID: 29755451 PMCID: PMC5932349 DOI: 10.3389/fimmu.2018.00692] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/21/2018] [Indexed: 01/10/2023] Open
Abstract
Pemphigus vulgaris (PV) is an autoimmune skin blistering disease effecting both cutaneous and mucosal epithelia. Blister formation in PV is known to result from the binding of autoantibodies (autoAbs) to keratinocyte antigens. The primary antigenic targets of pathogenic autoAbs are known to be desmoglein 3, and to a lesser extent, desmoglein 1, cadherin family proteins that partially comprise the desmosome, a protein structure responsible for maintaining cell adhesion, although additional autoAbs, whose role in blister formation is still unclear, are also known to be present in PV patients. Nevertheless, there remain large gaps in knowledge concerning the precise mechanisms through which autoAb binding induces blister formation. Consequently, the primary therapeutic interventions for PV focus on systemic immunosuppression, whose side effects represent a significant health risk to patients. In an effort to identify novel, disease-specific therapeutic targets, a multitude of studies attempting to elucidate the pathogenic mechanisms downstream of autoAb binding, have led to significant advancements in the understanding of autoAb-mediated blister formation. Despite this enhanced characterization of disease processes, a satisfactory explanation of autoAb-induced acantholysis still does not exist. Here, we carefully review the literature investigating the pathogenic disease mechanisms in PV and, taking into account the full scope of results from these studies, provide a novel, comprehensive theory of blister formation in PV.
Collapse
Affiliation(s)
- Thomas Sajda
- Department of Dermatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Animesh A Sinha
- Department of Dermatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
14
|
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: 45] [Impact Index Per Article: 6.4] [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
|
15
|
Louhivuori LM, Turunen PM, Louhivuori V, Yellapragada V, Nordström T, Uhlén P, Åkerman KE. Regulation of radial glial process growth by glutamate via mGluR5/TRPC3 and neuregulin/ErbB4. Glia 2017; 66:94-107. [PMID: 28887860 DOI: 10.1002/glia.23230] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/18/2017] [Accepted: 08/24/2017] [Indexed: 11/09/2022]
Abstract
Radial glial cells play an essential role through their function as guides for neuronal migration during development. Disruption of metabotropic glutamate receptor 5 (mGluR5) function retards the growth of radial glial processes in vitro. Neuregulins (NRG) are activated by proteolytic cleavage and regulate (radial) glial maintenance via ErbB3/ErbB4 receptors. We show here that blocking ErbB4 disrupts radial process extension. Soluble NRG acting on ErbB4 receptors is able to promote radial process extension in particular where process elongation has been impeded by blockade of mGluR5, the nonselective cation channel canonical transient receptor potential 3 (TRPC3), or matrix metalloproteases (MMP). NRG does not restore retarded process growth caused by ErbB4 blockade. Stimulation of muscarinic receptors restores process elongation due to mGluR5 blockade but not that caused by TRPC3, MMP or ErbB4 blockade suggesting that muscarinic receptors can replace mGluR5 with respect to radial process extension. Additionally, NRG/ErbB4 causes Ca2+ mobilization in a population of cells through cooperation with ErbB1 receptors. Our results indicate that mGluR5 promotes radial process growth via NRG activation by a mechanism involving TRPC3 channels and MMPs. Thus neurotransmitters acting on G-protein coupled receptors could play a central role in the maintenance of the radial glial scaffold through activation of NRG/ErbB4 signaling.
Collapse
Affiliation(s)
- Lauri M Louhivuori
- University of Helsinki, Biomedicum, Medicum/Physiology, Helsinki, FIN-00014, Finland.,Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Pauli M Turunen
- University of Helsinki, Biomedicum, Medicum/Physiology, Helsinki, FIN-00014, Finland
| | - Verna Louhivuori
- University of Helsinki, Biomedicum, Medicum/Physiology, Helsinki, FIN-00014, Finland
| | | | - Tommy Nordström
- University of Helsinki, Biomedicum, Medicum/Physiology, Helsinki, FIN-00014, Finland
| | - Per Uhlén
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Karl E Åkerman
- University of Helsinki, Biomedicum, Medicum/Physiology, Helsinki, FIN-00014, Finland
| |
Collapse
|
16
|
Baronas VA, Yang RY, Kurata HT. Extracellular redox sensitivity of Kv1.2 potassium channels. Sci Rep 2017; 7:9142. [PMID: 28831076 PMCID: PMC5567313 DOI: 10.1038/s41598-017-08718-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/17/2017] [Indexed: 12/03/2022] Open
Abstract
Kv1.2 is a prominent potassium channel subtype in the nervous system and serves as an important structural template for investigation of ion channel function. However, Kv1.2 voltage-dependence exhibits dramatic cell-to-cell variability due to a gating mode shift that is regulated by an unknown mechanism. We report that this variable behavior is regulated by the extracellular redox environment. Exposure to reducing agents promotes a shift in gating properties towards an 'inhibited' gating mode that resists opening, and causes channels to exhibit pronounced use-dependent activation during trains of repetitive depolarizations. This sensitivity to extracellular redox potential is absent in other Kv1 channels, but is apparent in heteromeric channels containing Kv1.2 subunits, and overlaps with the reported physiological range of extracellular redox couples. Mutagenesis of candidate cysteine residues fails to abolish redox sensitivity. Therefore, we suggest that an extrinsic, redox-sensitive binding partner imparts these properties.
Collapse
Affiliation(s)
- Victoria A Baronas
- Department of Pharmacology, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Runying Y Yang
- Department of Pharmacology, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Harley T Kurata
- Department of Pharmacology, Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
17
|
He Y, Li H, Chen Y, Li P, Gao L, Zheng Y, Sun Y, Chen J, Qian X. Expression of anoctamin 1 is associated with advanced tumor stage in patients with non-small cell lung cancer and predicts recurrence after surgery. Clin Transl Oncol 2017; 19:1091-1098. [PMID: 28299581 DOI: 10.1007/s12094-017-1643-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 03/02/2017] [Indexed: 02/07/2023]
|
18
|
Jain R, Watson U, Saini DK. ERK activated by Histamine H1 receptor is anti-proliferative through spatial restriction in the cytosol. Eur J Cell Biol 2016; 95:623-634. [DOI: 10.1016/j.ejcb.2016.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/04/2016] [Accepted: 10/24/2016] [Indexed: 10/20/2022] Open
|
19
|
Di Liberto V, Borroto-Escuela DO, Frinchi M, Verdi V, Fuxe K, Belluardo N, Mudò G. Existence of muscarinic acetylcholine receptor (mAChR) and fibroblast growth factor receptor (FGFR) heteroreceptor complexes and their enhancement of neurite outgrowth in neural hippocampal cultures. Biochim Biophys Acta Gen Subj 2016; 1861:235-245. [PMID: 27815219 DOI: 10.1016/j.bbagen.2016.10.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 10/15/2016] [Accepted: 10/31/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Recently, it was demonstrated that G-protein-coupled receptors (GPCRs) can transactivate tyrosine kinase receptors in absence of their ligands. In this work, driven by the observation that mAChRs and fibroblast growth factor receptors (FGFRs) share signalling pathways and regulation of brain functions, it was decided to explore whether mAChRs activation may transactivate FGFRs and, if so, to characterize the related trophic effects in cultured hippocampal neurons. METHODS Oxotremorine-M transactivation of FGFRs and related trophic effects were tested in primary hippocampal neurons. Western blotting and in situ proximity ligation assay (PLA) were used to detect FGFR phosphorylation (pFGFR) levels and M1R-FGFR1 heteroreceptor complexes, respectively. RESULTS Oxotremorine-M, a non-selective mAChRs agonist, was able to transactivate FGFR and this transactivation was blocked by Src inhibitors. Oxotremorine-M treatment produced a significant increase in the primary neurite outgrowth that was blocked by pre-treatment with the pFGFR inhibitor SU5402 and Src inhibitors. This trophic effect was almost similar to that induced by fibroblast growth factor-2 (FGF-2). By using atropine as nonselective mAChRs or pirenzepine as selective antagonist for M1 receptor (M1R) we could show that mAChRs are involved in modulating the pFGFRs. Using PLA, M1R-FGFR1 heteroreceptor complexes were identified in the hippocampus and cerebral cortex. CONCLUSION The current findings, by showing functional mAChR-FGFR interactions, will contribute to advance the understanding of the mechanisms involved in the actions of cholinergic drugs on neuronal plasticity. GENERAL SIGNIFICANT Data may help to develop novel therapeutic strategies not only for neurodegenerative diseases but also for depression-induced atrophy of hippocampal neurons.
Collapse
Affiliation(s)
- V Di Liberto
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Corso Tukory 129, 90134 Palermo, Italy.
| | - D O Borroto-Escuela
- Karolinska Instituet, Department of Neuroscience, Retzius väg 8, 17177 Stockholm, Sweden; Department of Biomolecular Science, Section of Physiology, University of Urbino, Campus Scientifico Enrico Mattei, via Ca' le Suore 2, I-61029 Urbino, Italy; Observatorio Cubano de Neurociencias, Grupo Bohío-Estudio, Zayas 50, 62100 Yaguajay, Cuba.
| | - M Frinchi
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Corso Tukory 129, 90134 Palermo, Italy.
| | - V Verdi
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Corso Tukory 129, 90134 Palermo, Italy.
| | - K Fuxe
- Karolinska Instituet, Department of Neuroscience, Retzius väg 8, 17177 Stockholm, Sweden.
| | - N Belluardo
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Corso Tukory 129, 90134 Palermo, Italy.
| | - G Mudò
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Corso Tukory 129, 90134 Palermo, Italy.
| |
Collapse
|
20
|
Rafikova O, Rafikov R, Kangath A, Qu N, Aggarwal S, Sharma S, Desai J, Fields T, Ludewig B, Yuan JXY, Jonigk D, Black SM. Redox regulation of epidermal growth factor receptor signaling during the development of pulmonary hypertension. Free Radic Biol Med 2016; 95:96-111. [PMID: 26928584 PMCID: PMC5929487 DOI: 10.1016/j.freeradbiomed.2016.02.029] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 02/24/2016] [Accepted: 02/25/2016] [Indexed: 11/19/2022]
Abstract
The development of pulmonary hypertension (PH) involves the uncontrolled proliferation of pulmonary smooth muscle cells via increased growth factor receptor signaling. However, the role of epidermal growth factor receptor (EGFR) signaling is controversial, as humans with advanced PH exhibit no changes in EGFR protein levels and purpose of the present study was to determine whether there are post-translational mechanisms that enhance EGFR signaling in PH. The EGFR inhibitor, gefinitib, significantly attenuated EGFR signaling and prevented the development of PH in monocrotaline (MCT)-exposed rats, confirming the contribution of EGFR activation in MCT induced PH. There was an early MCT-mediated increase in hydrogen peroxide, which correlated with the binding of the active metabolite of MCT, monocrotaline pyrrole, to catalase Cys377, disrupting its multimeric structure. This early oxidative stress was responsible for the oxidation of EGFR and the formation of sodium dodecyl sulfate (SDS) stable EGFR dimers through dityrosine cross-linking. These cross-linked dimers exhibited increased EGFR autophosphorylation and signaling. The activation of EGFR signaling did not correlate with pp60(src) dependent Y845 phosphorylation or EGFR ligand expression. Importantly, the analysis of patients with advanced PH revealed the same enhancement of EGFR autophosphorylation and covalent dimer formation in pulmonary arteries, while total EGFR protein levels were unchanged. As in the MCT exposed rat model, the activation of EGFR in human samples was independent of pp60(src) phosphorylation site and ligand expression. This study provides a novel molecular mechanism of oxidative stress stimulated covalent EGFR dimerization via tyrosine dimerization that contributes into development of PH.
Collapse
Affiliation(s)
- Olga Rafikova
- Department of Medicine, University of Arizona, Tucson, AZ, United States
| | - Ruslan Rafikov
- Department of Medicine, University of Arizona, Tucson, AZ, United States
| | - Archana Kangath
- Department of Medicine, University of Arizona, Tucson, AZ, United States
| | - Ning Qu
- Department of Medicine, University of Arizona, Tucson, AZ, United States
| | - Saurabh Aggarwal
- Department of Anesthesiology, University of Alabama, Birmingham, AL, United States
| | - Shruti Sharma
- Center For Biotechnology & Genomic Medicine, Georgia Regents University, Augusta, GA, United States
| | - Julin Desai
- Vascular Biology Center, Georgia Regents University, Augusta, GA, United States
| | - Taylor Fields
- Vascular Biology Center, Georgia Regents University, Augusta, GA, United States
| | - Britta Ludewig
- Institute of Pathology, Hannover Medical School, Hanover, Germany
| | - Jason X-Y Yuan
- Department of Medicine, University of Arizona, Tucson, AZ, United States
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, Hanover, Germany
| | - Stephen M Black
- Department of Medicine, University of Arizona, Tucson, AZ, United States.
| |
Collapse
|
21
|
Fan Q, Verhoeven VJM, Wojciechowski R, Barathi VA, Hysi PG, Guggenheim JA, Höhn R, Vitart V, Khawaja AP, Yamashiro K, Hosseini SM, Lehtimäki T, Lu Y, Haller T, Xie J, Delcourt C, Pirastu M, Wedenoja J, Gharahkhani P, Venturini C, Miyake M, Hewitt AW, Guo X, Mazur J, Huffman JE, Williams KM, Polasek O, Campbell H, Rudan I, Vatavuk Z, Wilson JF, Joshi PK, McMahon G, St Pourcain B, Evans DM, Simpson CL, Schwantes-An TH, Igo RP, Mirshahi A, Cougnard-Gregoire A, Bellenguez C, Blettner M, Raitakari O, Kähönen M, Seppala I, Zeller T, Meitinger T, Ried JS, Gieger C, Portas L, van Leeuwen EM, Amin N, Uitterlinden AG, Rivadeneira F, Hofman A, Vingerling JR, Wang YX, Wang X, Tai-Hui Boh E, Ikram MK, Sabanayagam C, Gupta P, Tan V, Zhou L, Ho CEH, Lim W, Beuerman RW, Siantar R, Tai ES, Vithana E, Mihailov E, Khor CC, Hayward C, Luben RN, Foster PJ, Klein BEK, Klein R, Wong HS, Mitchell P, Metspalu A, Aung T, Young TL, He M, Pärssinen O, van Duijn CM, Jin Wang J, Williams C, Jonas JB, Teo YY, Mackey DA, Oexle K, Yoshimura N, Paterson AD, Pfeiffer N, Wong TY, Baird PN, Stambolian D, Wilson JEB, Cheng CY, Hammond CJ, et alFan Q, Verhoeven VJM, Wojciechowski R, Barathi VA, Hysi PG, Guggenheim JA, Höhn R, Vitart V, Khawaja AP, Yamashiro K, Hosseini SM, Lehtimäki T, Lu Y, Haller T, Xie J, Delcourt C, Pirastu M, Wedenoja J, Gharahkhani P, Venturini C, Miyake M, Hewitt AW, Guo X, Mazur J, Huffman JE, Williams KM, Polasek O, Campbell H, Rudan I, Vatavuk Z, Wilson JF, Joshi PK, McMahon G, St Pourcain B, Evans DM, Simpson CL, Schwantes-An TH, Igo RP, Mirshahi A, Cougnard-Gregoire A, Bellenguez C, Blettner M, Raitakari O, Kähönen M, Seppala I, Zeller T, Meitinger T, Ried JS, Gieger C, Portas L, van Leeuwen EM, Amin N, Uitterlinden AG, Rivadeneira F, Hofman A, Vingerling JR, Wang YX, Wang X, Tai-Hui Boh E, Ikram MK, Sabanayagam C, Gupta P, Tan V, Zhou L, Ho CEH, Lim W, Beuerman RW, Siantar R, Tai ES, Vithana E, Mihailov E, Khor CC, Hayward C, Luben RN, Foster PJ, Klein BEK, Klein R, Wong HS, Mitchell P, Metspalu A, Aung T, Young TL, He M, Pärssinen O, van Duijn CM, Jin Wang J, Williams C, Jonas JB, Teo YY, Mackey DA, Oexle K, Yoshimura N, Paterson AD, Pfeiffer N, Wong TY, Baird PN, Stambolian D, Wilson JEB, Cheng CY, Hammond CJ, Klaver CCW, Saw SM, Rahi JS, Korobelnik JF, Kemp JP, Timpson NJ, Smith GD, Craig JE, Burdon KP, Fogarty RD, Iyengar SK, Chew E, Janmahasatian S, Martin NG, MacGregor S, Xu L, Schache M, Nangia V, Panda-Jonas S, Wright AF, Fondran JR, Lass JH, Feng S, Zhao JH, Khaw KT, Wareham NJ, Rantanen T, Kaprio J, Pang CP, Chen LJ, Tam PO, Jhanji V, Young AL, Döring A, Raffel LJ, Cotch MF, Li X, Yip SP, Yap MK, Biino G, Vaccargiu S, Fossarello M, Fleck B, Yazar S, Tideman JWL, Tedja M, Deangelis MM, Morrison M, Farrer L, Zhou X, Chen W, Mizuki N, Meguro A, Mäkelä KM. Meta-analysis of gene-environment-wide association scans accounting for education level identifies additional loci for refractive error. Nat Commun 2016; 7:11008. [PMID: 27020472 PMCID: PMC4820539 DOI: 10.1038/ncomms11008] [Show More Authors] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 02/10/2016] [Indexed: 02/07/2023] Open
Abstract
Myopia is the most common human eye disorder and it results from complex genetic and environmental causes. The rapidly increasing prevalence of myopia poses a major public health challenge. Here, the CREAM consortium performs a joint meta-analysis to test single-nucleotide polymorphism (SNP) main effects and SNP × education interaction effects on refractive error in 40,036 adults from 25 studies of European ancestry and 10,315 adults from 9 studies of Asian ancestry. In European ancestry individuals, we identify six novel loci (FAM150B-ACP1, LINC00340, FBN1, DIS3L-MAP2K1, ARID2-SNAT1 and SLC14A2) associated with refractive error. In Asian populations, three genome-wide significant loci AREG, GABRR1 and PDE10A also exhibit strong interactions with education (P<8.5 × 10(-5)), whereas the interactions are less evident in Europeans. The discovery of these loci represents an important advance in understanding how gene and environment interactions contribute to the heterogeneity of myopia.
Collapse
Affiliation(s)
- Qiao Fan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Virginie J. M. Verhoeven
- Department of Ophthalmology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Robert Wojciechowski
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, Maryland 21224, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 20205, USA
| | - Veluchamy A. Barathi
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Ophthalmology, National University Health Systems, National University of Singapore Singapore 119228, Singapore
| | - Pirro G. Hysi
- Department of Twin Research and Genetic Epidemiology, King's College London School of Medicine, London SE1 7EH, UK
| | - Jeremy A. Guggenheim
- School of Optometry and Vision Sciences, Cardiff University, Cardiff CF24 4HQ, UK
| | - René Höhn
- Department of Ophthalmology, University Medical Center Mainz, 55131 Mainz, Germany
- Department of Ophthalmology, Inselspital, University Hospital Bern, CH-3010 Bern, Switzerland
| | - Veronique Vitart
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, Scotland
| | - Anthony P. Khawaja
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge School of Clinical Medicine, Cambridge CB2 0SR, UK
| | - Kenji Yamashiro
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - S Mohsen Hosseini
- Program in Genetics and Genome Biology, The Hospital for Sick Children and Institute for Medical Sciences, University of Toronto, Toronto Ontario, Canada M5G 1X8
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, Tampere 33520, Finland
| | - Yi Lu
- Statistical Genetics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland 4029, Australia
| | - Toomas Haller
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Jing Xie
- Centre for Eye Research Australia (CERA), Royal Victorian Eye and Ear Hospital, University of Melbourne, Melbourne, Victoria 3002, Australia
| | - Cécile Delcourt
- Université de Bordeaux, ISPED (Institut de Santé Publique d'Épidémiologie et de Développement), Bordeaux 33000, France
- INSERM, U1219-Bordeaux Population Health Research Center, Bordeaux 33000, France
| | - Mario Pirastu
- Institute of Population Genetics, National Research Council, Sassari 07100, Italy
| | - Juho Wedenoja
- Department of Public Health, University of Helsinki, Helsinki 00014, Finland
- Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Helsinki 00014, Finland
| | - Puya Gharahkhani
- Statistical Genetics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland 4029, Australia
| | - Cristina Venturini
- Department of Twin Research and Genetic Epidemiology, King's College London School of Medicine, London SE1 7EH, UK
- UCL Institute of Ophthalmology, London SE1 7EH, UK
| | - Masahiro Miyake
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Alex W. Hewitt
- Centre for Eye Research Australia (CERA), Royal Victorian Eye and Ear Hospital, University of Melbourne, Melbourne, Victoria 3002, Australia
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Xiaobo Guo
- Department of Statistical Science, School of Mathematics and Computational Science, Sun Yat-Sen University, Guangzhou 510275, China
| | - Johanna Mazur
- Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center Mainz, 55131 Mainz, Germany
| | - Jenifer E. Huffman
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, Scotland
| | - Katie M. Williams
- Department of Twin Research and Genetic Epidemiology, King's College London School of Medicine, London SE1 7EH, UK
- Department of Ophthalmology, King's College London, London SE1 7EH, UK
| | - Ozren Polasek
- Faculty of Medicine, University of Split, Split 21000, Croatia
| | - Harry Campbell
- Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, Scotland
| | - Igor Rudan
- Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, Scotland
| | - Zoran Vatavuk
- Department of Ophthalmology, Sisters of Mercy University Hospital, Zagreb 10000, Croatia
| | - James F. Wilson
- Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, Scotland
| | - Peter K. Joshi
- Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, Scotland
| | - George McMahon
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol BS8 2BN, UK
- School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Beate St Pourcain
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol BS8 2BN, UK
- School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
- Max Planck Institute for Psycholinguistics, Wundtlaan 1, 6525 XD Nijmegen, The Netherlands
| | - David M. Evans
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol BS8 2BN, UK
- School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland 4102, Australia
| | - Claire L. Simpson
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, Maryland 21224, USA
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Tae-Hwi Schwantes-An
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Robert P. Igo
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Alireza Mirshahi
- Department of Ophthalmology, University Medical Center Mainz, 55131 Mainz, Germany
- Dardenne Eye Hospital, Bonn-Bad Godesberg, 53177 Bonn, Germany
| | - Audrey Cougnard-Gregoire
- Université de Bordeaux, ISPED (Institut de Santé Publique d'Épidémiologie et de Développement), Bordeaux 33000, France
- INSERM, U1219-Bordeaux Population Health Research Center, Bordeaux 33000, France
| | - Céline Bellenguez
- Inserm, U1167, Lille 59000, France
- Univ. Lille, U1167, Lille 59000, France
- Université Lille 2, Lille 59000, France
| | - Maria Blettner
- Institute of Medical Biostatistics, Epidemiology and Informatics, University Medical Center Mainz, 55131 Mainz, Germany
| | - Olli Raitakari
- Research Centre of Applied and Preventive Medicine, University of Turku, Turku 20520, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku 20520, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital and School of Medicine, University of Tampere, Tampere 33520, Finland
| | - Ilkka Seppala
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, Tampere 33520, Finland
| | - Tanja Zeller
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, 20246 Hamburg, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | | | - Janina S. Ried
- Institute of Genetic Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Laura Portas
- Institute of Population Genetics, National Research Council, Sassari 07100, Italy
| | | | - Najaf Amin
- Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
| | - André G. Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
- Netherlands Consortium for Healthy Ageing, Netherlands Genomics Initiative, 2518 AD Hague, The Netherlands
| | - Fernando Rivadeneira
- Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
- Netherlands Consortium for Healthy Ageing, Netherlands Genomics Initiative, 2518 AD Hague, The Netherlands
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
- Netherlands Consortium for Healthy Ageing, Netherlands Genomics Initiative, 2518 AD Hague, The Netherlands
| | | | - Ya Xing Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing 100044, China
| | - Xu Wang
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health Systems, Singapore 117549, Singapore
| | - Eileen Tai-Hui Boh
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health Systems, Singapore 117549, Singapore
| | - M. Kamran Ikram
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Charumathi Sabanayagam
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Preeti Gupta
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
| | - Vincent Tan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
| | - Lei Zhou
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
| | - Candice E. H. Ho
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
| | - Wan'e Lim
- Department of Ophthalmology, National University Health Systems, National University of Singapore Singapore 119228, Singapore
| | - Roger W. Beuerman
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Ophthalmology, National University Health Systems, National University of Singapore Singapore 119228, Singapore
| | - Rosalynn Siantar
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | - E-Shyong Tai
- Duke-NUS Medical School, Singapore 169857, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health Systems, Singapore 117549, Singapore
- Department of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Eranga Vithana
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Ophthalmology, National University Health Systems, National University of Singapore Singapore 119228, Singapore
| | - Evelin Mihailov
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Chiea-Chuen Khor
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health Systems, Singapore 117549, Singapore
- Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, Scotland
| | - Robert N. Luben
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge School of Clinical Medicine, Cambridge CB2 0SR, UK
| | - Paul J. Foster
- Division of Genetics and Epidemiology, UCL Institute of Ophthalmology, London EC1V 9EL, UK
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 2PD, UK
| | - Barbara E. K. Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53726, USA
| | - Ronald Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53726, USA
| | - Hoi-Suen Wong
- Program in Genetics and Genome Biology, The Hospital for Sick Children and Institute for Medical Sciences, University of Toronto, Toronto Ontario, Canada M5G 1X8
| | - Paul Mitchell
- Department of Ophthalmology, Centre for Vision Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales 2145, Australia
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Tin Aung
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Department of Ophthalmology, National University Health Systems, National University of Singapore Singapore 119228, Singapore
| | - Terri L. Young
- Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53705, USA
| | - Mingguang He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Olavi Pärssinen
- Department of Ophthalmology, Central Hospital of Central Finland, Jyväskylä 40620, Finland
- Gerontology Research Center and Department of Health Sciences, University of Jyväskylä, Jyväskylä 40014, Finland
| | - Cornelia M. van Duijn
- Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Jie Jin Wang
- Department of Ophthalmology, Centre for Vision Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales 2145, Australia
| | - Cathy Williams
- School of Social and Community Medicine, University of Bristol, Bristol BS8 2BN, UK
| | - Jost B. Jonas
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing 100044, China
- Medical Faculty Mannheim, Department of Ophthalmology, Ruprecht-Karls-University Heidelberg, 69115 Mannheim, Germany
| | - Yik-Ying Teo
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health Systems, Singapore 117549, Singapore
- Division of Human Genetics, Genome Institute of Singapore, Singapore 138672, Singapore
- Department of Statistics and Applied Probability, National University of Singapore, Singapore 117546, Singapore
| | - David A. Mackey
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania 7000, Australia
- Centre for Ophthalmology and Visual Science, Lions Eye Institute, University of Western Australia, Perth, Western Australia 6009, Australia
| | - Konrad Oexle
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Nagahisa Yoshimura
- Department of Ophthalmology and Visual Sciences, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
| | - Andrew D. Paterson
- Program in Genetics and Genome Biology, The Hospital for Sick Children and Institute for Medical Sciences, University of Toronto, Toronto Ontario, Canada M5G 1X8
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Tien-Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Ophthalmology, National University Health Systems, National University of Singapore Singapore 119228, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health Systems, Singapore 117549, Singapore
| | - Paul N. Baird
- Centre for Eye Research Australia (CERA), Royal Victorian Eye and Ear Hospital, University of Melbourne, Melbourne, Victoria 3002, Australia
| | - Dwight Stambolian
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Joan E. Bailey Wilson
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Ophthalmology, National University Health Systems, National University of Singapore Singapore 119228, Singapore
| | - Christopher J. Hammond
- Department of Twin Research and Genetic Epidemiology, King's College London School of Medicine, London SE1 7EH, UK
- Department of Ophthalmology, King's College London, London SE1 7EH, UK
| | - Caroline C. W. Klaver
- Department of Ophthalmology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Seang-Mei Saw
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore 169856, Singapore
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Ophthalmology, National University Health Systems, National University of Singapore Singapore 119228, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health Systems, Singapore 117549, Singapore
| | - Jugnoo S. Rahi
- Medical Research Council Centre of Epidemiology for Child Health, Institute of Child Health, University College London, London WC1E 6BT, UK
- Institute of Ophthalmology, Moorfields Eye Hospital, London EC1V 2PD, UK
- Ulverscroft Vision Research Group, University College London, London WC1E 6BT, UK
| | - Jean-François Korobelnik
- Université de Bordeaux, 33400 Talence, France
- INSERM (Institut National de la Santé Et de la Recherche Médicale), ISPED (Institut de Santé Publique d'épidémiologie et de Développement), Centre INSERM U897-Epidemiologie-Biostatistique, 33076 Bordeaux, France
| | - John P. Kemp
- MRC Integrative Epidemiology Unit (IEU), The University of Bristol, Bristol BS8 2BN, UK
| | - Nicholas J. Timpson
- MRC Integrative Epidemiology Unit (IEU), The University of Bristol, Bristol BS8 2BN, UK
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU), The University of Bristol, Bristol BS8 2BN, UK
| | - Jamie E. Craig
- Department of Ophthalmology, Flinders University, Adelaide, South Australia 5001, Australia
| | - Kathryn P. Burdon
- Department of Ophthalmology, Flinders University, Adelaide, South Australia 5001, Australia
| | - Rhys D. Fogarty
- Department of Ophthalmology, Flinders University, Adelaide, South Australia 5001, Australia
| | - Sudha K. Iyengar
- Department of Epidemiology and Biostatistics, CaseWestern Reserve University, Cleveland, Ohio 44106, USA
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University and University Hospitals Eye Institute, Cleveland, Ohio 44106, USA
- Department of Genetics, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Emily Chew
- National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Sarayut Janmahasatian
- Department of Epidemiology and Biostatistics, CaseWestern Reserve University, Cleveland, Ohio 44106, USA
| | - Nicholas G. Martin
- Genetic Epidemiology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland 4029, Australia
| | - Stuart MacGregor
- Statistical Genetics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland 4029, Australia
| | - Liang Xu
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing 100044, China
| | - Maria Schache
- Centre for Eye Research Australia (CERA), Royal Victorian Eye and Ear Hospital, University of Melbourne, Melbourne, Victoria 3002, Australia
| | - Vinay Nangia
- Suraj Eye Institute, Nagpur, Maharashtra 440001, India
| | | | - Alan F. Wright
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, Scotland
| | - Jeremy R. Fondran
- Department of Epidemiology and Biostatistics, CaseWestern Reserve University, Cleveland, Ohio 44106, USA
| | - Jonathan H. Lass
- Department of Epidemiology and Biostatistics, CaseWestern Reserve University, Cleveland, Ohio 44106, USA
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University and University Hospitals Eye Institute, Cleveland, Ohio 44106, USA
| | - Sheng Feng
- Department of Pediatric Ophthalmology, Duke Eye Center For Human Genetics, Durham, North Carolina 27710, USA
| | - Jing Hua Zhao
- MRC Epidemiology Unit, Institute of Metabolic Sciences, University of Cambridge, Cambridge CB2 1TN, UK
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge School of Clinical Medicine, Cambridge CB2 0SR, UK
| | - Nick J. Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Taina Rantanen
- Gerontology Research Center, University of Jyväskylä, Jyväskylä Finland
| | - Jaakko Kaprio
- Department of Public Health, University of Helsinki, Helsinki 00014, Finland
- Institute for Molecular Medicine, University of Helsinki, Helsinki 00014, Finland
- Department of Mental Health and Alcohol Abuse Services, National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, Hong Kong Eye Hospital, The Chinese University of Hong Kong, Kowloon, Hong Kong
| | - Li Jia Chen
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Pancy O. Tam
- Department of Ophthalmology and Visual Sciences, Hong Kong Eye Hospital, The Chinese University of Hong Kong, Kowloon, Hong Kong
| | - Vishal Jhanji
- Department of Ophthalmology and Visual Sciences, Hong Kong Eye Hospital, The Chinese University of Hong Kong, Kowloon, Hong Kong
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Alvin L. Young
- Department of Ophthalmology and Visual Sciences, Hong Kong Eye Hospital, The Chinese University of Hong Kong, Kowloon, Hong Kong
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Angela Döring
- Institute of Epidemiology I, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Leslie J. Raffel
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | - Mary-Frances Cotch
- Division of Epidemiology and Clinical Applications, National Eye Institute, Bethesda, Maryland 20892, USA
| | - Xiaohui Li
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Los Angeles, California 90502, USA
| | - Shea Ping Yip
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong
| | - Maurice K.H. Yap
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong
| | - Ginevra Biino
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - Simona Vaccargiu
- Institute of Population Genetics, National Research Council, Sassari 07100, Italy
| | - Maurizio Fossarello
- Institute of Population Genetics, National Research Council, Sassari 07100, Italy
| | - Brian Fleck
- Princess Alexandra Eye Pavilion, Edinburgh EH3 9HA, UK
| | - Seyhan Yazar
- Centre for Eye Research Australia (CERA), Royal Victorian Eye and Ear Hospital, University of Melbourne, Melbourne, Victoria 3002, Australia
- Centre for Ophthalmology and Visual Science, Lions Eye Institute, University of Western Australia, Perth, Western Australia 6009, Australia
| | - Jan Willem L. Tideman
- Department of Ophthalmology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Milly Tedja
- Department of Ophthalmology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, 3000 CA Rotterdam, The Netherlands
| | - Margaret M. Deangelis
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah, Salt Lake City, Utah 84132, USA
| | - Margaux Morrison
- Department of Ophthalmology and Visual Sciences, John Moran Eye Center, University of Utah, Salt Lake City, Utah 84132, USA
| | - Lindsay Farrer
- Departments of Medicine (Biomedical Genetics), Ophthalmology, Neurology, Epidemiology and Biostatistics, Boston University Schools of Medicine and Public Health, Boston, Massachusetts 02118, USA
| | - Xiangtian Zhou
- School of ophthalmology and optometry, Wenzhou Medical University, Wenzhou 325035, China
| | - Wei Chen
- School of ophthalmology and optometry, Wenzhou Medical University, Wenzhou 325035, China
| | - Nobuhisa Mizuki
- Department of Ophthalmology, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0027, Japan
| | - Akira Meguro
- Department of Ophthalmology, Yokohama City University School of Medicine, Yokohama, Kanagawa 236-0027, Japan
| | - Kari Matti Mäkelä
- Department of Clinical Chemistry, Fimlab Laboratories and School of Medicine, University of Tampere, Tampere 33014, Finland
| |
Collapse
|
22
|
Abstract
In excitable cells, ion channels are frequently challenged by repetitive stimuli, and their responses shape cellular behavior by regulating the duration and termination of bursts of action potentials. We have investigated the behavior of Shaker family voltage-gated potassium (Kv) channels subjected to repetitive stimuli, with a particular focus on Kv1.2. Genetic deletion of this subunit results in complete mortality within 2 weeks of birth in mice, highlighting a critical physiological role for Kv1.2. Kv1.2 channels exhibit a unique property described previously as "prepulse potentiation," in which activation by a depolarizing step facilitates activation in a subsequent pulse. In this study, we demonstrate that this property enables Kv1.2 channels to exhibit use-dependent activation during trains of very brief depolarizations. Also, Kv subunits usually assemble into heteromeric channels in the central nervous system, generating diversity of function and sensitivity to signaling mechanisms. We demonstrate that other Kv1 channel types do not exhibit use-dependent activation, but this property is conferred in heteromeric channel complexes containing even a single Kv1.2 subunit. This regulatory mechanism is observed in mammalian cell lines as well as primary cultures of hippocampal neurons. Our findings illustrate that use-dependent activation is a unique property of Kv1.2 that persists in heteromeric channel complexes and may influence function of hippocampal neurons.
Collapse
|
23
|
Miao Y, Bi XY, Zhao M, Jiang HK, Liu JJ, Li DL, Yu XJ, Yang YH, Huang N, Zang WJ. Acetylcholine inhibits tumor necrosis factor α activated endoplasmic reticulum apoptotic pathway via EGFR-PI3K signaling in cardiomyocytes. J Cell Physiol 2015; 230:767-74. [PMID: 25201632 DOI: 10.1002/jcp.24800] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 09/05/2014] [Indexed: 12/11/2022]
Abstract
Previous findings have shown that acetylcholine (ACh) decreased hypoxia-induced tumor necrosis factor alpha (TNF α) production, thus protected against cardiomyocyte injury. However, whether and how ACh affects TNF α-induced endoplasmic reticulum (ER) stress and cell apoptosis remain poorly defined. This study was aimed at determining the effect of ACh in H9c2 cells after TNF α stimulation. Presence of ER stress was verified using the ER stress protein markers glucose regulatory protein 78 (GRP78) and C/EBP homologous protein (CHOP). Cell apoptosis was shown by caspase-3 activation and terminal deoxynucleotidyl transferase mediated dUTP-biotin nick end labeling. Exogenously administered ACh significantly decreased these TNF α-induced changes. Moreover, when the cells were exposed to nonspecific muscarinic receptor (M AChR) inhibitor atropine, methoctramine (M2 AChR inhibitor) or the epidermal growth factor receptor (EGFR) inhibitor AG1478, the cardioprotection elicited by ACh was diminished. Furthermore, the above effects were also blocked by M2 AChR or EGFR siRNA, indicating that EGFR transactivation by M2 AChR may be the major pathway responsible for the benefits of ACh. In addition, LY294002, a phosphatidylinositol-3-kinase (PI3K) inhibitor, displayed the similar trends as AG1478, suggesting that PI3K/Akt signaling may be the downstream of EGFR in ACh-elicited anti-apoptotic property. Together, these data indicate that EGFR-PI3K/Akt signaling is involved in M2 AChR-mediated ER apoptotic pathway suppression and the subsequent survival of H9c2 cardiomyocytes. We have identified a novel pathway underlying the cardioprotection afforded by ACh.
Collapse
Affiliation(s)
- Yi Miao
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, P. R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
El-Daly M, Saifeddine M, Mihara K, Ramachandran R, Triggle CR, Hollenberg MD. Proteinase-activated receptors 1 and 2 and the regulation of porcine coronary artery contractility: a role for distinct tyrosine kinase pathways. Br J Pharmacol 2014; 171:2413-25. [PMID: 24506284 DOI: 10.1111/bph.12593] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 12/23/2013] [Accepted: 01/17/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE Because angiotensin-II-mediated porcine coronary artery (PCA) vasoconstriction is blocked by protein tyrosine kinase (PYK) inhibitors, we hypothesized that proteinase-activated receptors (PARs), known to regulate vascular tension, like angiotensin-II, would also cause PCA contractions via PYK-dependent signalling pathways. EXPERIMENTAL APPROACH Contractions of intact and endothelium-free isolated PCA rings, stimulated by PAR1 /PAR2 -activating peptides, angiotensin-II, PGF2α , EGF, PDGF and KCl, were monitored with/without multiple signalling pathway inhibitors, including AG-tyrphostins AG18 (non-specific PYKs), AG1478 (EGF-receptor kinase), AG1296 (PDGF receptor kinase), PP1 (Src kinase), U0126 and PD98059 (MEK/MAPKinase kinase), indomethacin/SC-560/NS-398 (COX-1/2) and L-NAME (NOS). KEY RESULTS AG18 inhibited the contractions induced by all the agonists except KCl, whereas U0126 attenuated contractions induced by PAR1 /PAR2 agonists, EGF and angiotensin-II, but not by PGF2α , the COX-produced metabolites of arachidonate and KCl. PP1 only affected the responses to PAR1 /PAR2 -activating peptides and angiotensin-II. The EGF-kinase inhibitor, AG1478, attenuated contractions initiated by the PARs (PAR2 >> PAR1 ) and EGF itself, but not by angiotensin-II, PGF2α or KCl. COX-1/2 inhibitors blocked the contractions induced by all the agonists, except KCl and PGF2α . CONCLUSION AND IMPLICATIONS PAR1/2 -mediated contractions of the PCA are dependent on Src and MAPKinase and, in part, involve EGF-receptor-kinase transactivation and the generation of a COX-derived contractile agonist. However, the PYK signalling pathways used by PARs are distinct from each other and from those triggered by angiotensin-II and EGF. These signalling pathways may be therapeutic targets for managing coagulation-proteinase-induced coronary vasospasm.
Collapse
Affiliation(s)
- Mahmoud El-Daly
- Libin Cardiovascular Institute of Alberta and the Snyder Institute for Chronic Diseases, Calgary, AB, Canada; Department of Physiology and Pharmacology, The University of Calgary Faculty of Medicine, Calgary, AB, Canada
| | | | | | | | | | | |
Collapse
|
25
|
Ockenga W, Kühne S, Bocksberger S, Banning A, Tikkanen R. Epidermal growth factor receptor transactivation is required for mitogen-activated protein kinase activation by muscarinic acetylcholine receptors in HaCaT keratinocytes. Int J Mol Sci 2014; 15:21433-54. [PMID: 25421240 PMCID: PMC4264234 DOI: 10.3390/ijms151121433] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 11/13/2014] [Accepted: 11/17/2014] [Indexed: 01/03/2023] Open
Abstract
Non-neuronal acetylcholine plays a substantial role in the human skin by influencing adhesion, migration, proliferation and differentiation of keratinocytes. These processes are regulated by the Mitogen-Activated Protein (MAP) kinase cascade. Here we show that in HaCaT keratinocytes all five muscarinic receptor subtypes are expressed, but M1 and M3 are the subtypes involved in mitogenic signaling. Stimulation with the cholinergic agonist carbachol leads to activation of the MAP kinase extracellular signal regulated kinase, together with the protein kinase Akt. The activation is fully dependent on the transactivation of the epidermal growth factor receptor (EGFR), which even appears to be the sole pathway for the muscarinic receptors to facilitate MAP kinase activation in HaCaT cells. The transactivation pathway involves a triple-membrane-passing process, based on activation of matrix metalloproteases, and extracellular ligand release; whereas phosphatidylinositol 3-kinase, Src family kinases or protein kinase C do not appear to be involved in MAP kinase activation. Furthermore, phosphorylation, ubiquitination and endocytosis of the EGF receptor after cholinergic transactivation are different from that induced by a direct stimulation with EGF, suggesting that ligands other than EGF itself mediate the cholinergic transactivation.
Collapse
Affiliation(s)
- Wymke Ockenga
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, D-35392 Giessen, Germany.
| | - Sina Kühne
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, D-35392 Giessen, Germany.
| | - Simone Bocksberger
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, D-35392 Giessen, Germany.
| | - Antje Banning
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, D-35392 Giessen, Germany.
| | - Ritva Tikkanen
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, D-35392 Giessen, Germany.
| |
Collapse
|
26
|
Takahashi T, Ohnishi H, Sugiura Y, Honda K, Suematsu M, Kawasaki T, Deguchi T, Fujii T, Orihashi K, Hippo Y, Watanabe T, Yamagaki T, Yuba S. Non‐neuronal acetylcholine as an endogenous regulator of proliferation and differentiation of Lgr5‐positive stem cells in mice. FEBS J 2014; 281:4672-90. [DOI: 10.1111/febs.12974] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 07/22/2014] [Accepted: 08/14/2014] [Indexed: 12/13/2022]
Affiliation(s)
- Toshio Takahashi
- Suntory Foundation for Life Sciences Bioorganic Research Institute Osaka Japan
| | - Hiroe Ohnishi
- National Institute of Advanced Industrial Science and Technology Hyogo Japan
| | - Yuki Sugiura
- Department of Biochemistry School of Medicine Keio University Tokyo Japan
- Precursory Research for Embryonic Science and Technology Tokyo Japan
| | - Kurara Honda
- Department of Biochemistry School of Medicine Keio University Tokyo Japan
- Precursory Research for Embryonic Science and Technology Tokyo Japan
| | - Makoto Suematsu
- Department of Biochemistry School of Medicine Keio University Tokyo Japan
- Japan Science Technology Agency Exploratory Research for Advanced Technology Suematsu Gas Biology Project Tokyo Japan
| | - Takashi Kawasaki
- National Institute of Advanced Industrial Science and Technology Hyogo Japan
| | - Tomonori Deguchi
- National Institute of Advanced Industrial Science and Technology Hyogo Japan
| | - Takeshi Fujii
- Department of Pharmacology Faculty of Pharmaceutical Sciences Doshisha Women's College of Liberal Arts Kyoto Japan
| | - Kaoru Orihashi
- Division of Cancer Development System National Cancer Research Institute Tokyo Japan
| | - Yoshitaka Hippo
- Division of Cancer Development System National Cancer Research Institute Tokyo Japan
| | - Takehiro Watanabe
- Suntory Foundation for Life Sciences Bioorganic Research Institute Osaka Japan
| | - Tohru Yamagaki
- Suntory Foundation for Life Sciences Bioorganic Research Institute Osaka Japan
| | - Shunsuke Yuba
- National Institute of Advanced Industrial Science and Technology Hyogo Japan
| |
Collapse
|
27
|
Rayego-Mateos S, Morgado-Pascual JL, Sanz AB, Ramos AM, Eguchi S, Batlle D, Pato J, Keri G, Egido J, Ortiz A, Ruiz-Ortega M. TWEAK transactivation of the epidermal growth factor receptor mediates renal inflammation. J Pathol 2014; 231:480-94. [PMID: 24037740 DOI: 10.1002/path.4250] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 08/13/2013] [Accepted: 08/21/2013] [Indexed: 11/08/2022]
Abstract
TWEAK, a member of the TNF superfamily, binds to the Fn14 receptor, eliciting biological responses. EGFR signalling is involved in experimental renal injury. Our aim was to investigate the relationship between TWEAK and EGFR in the kidney. Systemic TWEAK administration into C57BL/6 mice increased renal EGFR phosphorylation, mainly in tubular epithelial cells. In vitro, in these cells TWEAK phosphorylated EGFR via Fn14 binding, ADAM17 activation and subsequent release of the EGFR ligands HB-EGF and TGFα. In vivo the EGFR kinase inhibitor Erlotinib inhibited TWEAK-induced renal EGFR activation and downstream signalling, including ERK activation, up-regulation of proinflammatory factors and inflammatory cell infiltration. Moreover, the ADAM17 inhibitor WTACE-2 also prevented those TWEAK-induced renal effects. In vitro TWEAK induction of proinflammatory factors was prevented by EGFR, ERK or ADAM17 inhibition. In contrast, EGFR transactivation did not modify TWEAK-mediated NF-κB activation. Our data suggest that TWEAK transactivates EGFR in the kidney, leading to modulation of downstream effects, including ERK activation and inflammation, and suggest that inhibition of EGFR signalling could be a novel therapeutic tool for renal inflammation.
Collapse
Affiliation(s)
- Sandra Rayego-Mateos
- Cellular Biology in Renal Diseases Laboratory, Universidad Autónoma, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Zhang C, Bosch MA, Rønnekleiv OK, Kelly MJ. Kisspeptin activation of TRPC4 channels in female GnRH neurons requires PIP2 depletion and cSrc kinase activation. Endocrinology 2013; 154:2772-83. [PMID: 23744639 PMCID: PMC3713215 DOI: 10.1210/en.2013-1180] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Kisspeptin signaling via its Gαq-coupled receptor GPR54 plays a crucial role in modulating GnRH neuronal excitability, which controls pituitary gonadotropins secretion and ultimately reproduction. Kisspeptin potently depolarizes GnRH neurons primarily through the activation of canonical transient receptor potential (TRPC) channels, but the intracellular signaling cascade has not been elucidated. Presently, we have established that kisspeptin activation of TRPC channels requires multiple membrane and intracellular signaling molecules. First, phosphatidylinositol-4,5-bisphosphate (PIP(2)) hydrolysis by phospholipase Cβ is required because whole-cell dialysis of Dioctanoylglycerol-PIP(2) (DiC8-PIP(2)) inhibited the kisspeptin activation of TRPC channels, and the phosphatidylinositol 4-kinase inhibitor wortmannin, which attenuates PIP(2) synthesis, prolonged TRPC channel activation. Using single cell RT-PCR, we identified that the mRNA for the PIP(2)-interacting TRPC channel subunit, TRPC4α, is expressed in GnRH neurons. Depletion of intracellular Ca(2+) stores by thapsigargin and inositol 1,4,5-trisphosphate had no effect, indicating that the TRPC channels are not store-operated. Neither removing extracellular Ca(2+) nor buffering intracellular Ca(2+) with EGTA or BAPTA had any effect on the kisspeptin activation of the TRPC channels. However, the Ca(2+) channel blocker Ni(2+) inhibited the kisspeptin-induced inward current. Moreover, inhibition of protein kinase C by bisindolylmaleimide-I or calphostin C had no effect, but activation of protein kinase C by phorbol 12,13-dibutyrate occluded the kisspeptin-activated current. Finally, inhibition of the cytoplasmic tyrosine kinase cSrc by genistein or the pyrazolo-pyrimidine PP2 blocked the activation of TRPC channels by kisspeptin. Therefore, TRPC channels in GnRH neurons are receptor-operated, and kisspeptin activates TRPC channels through PIP(2) depletion and cSrc tyrosine kinase activation, which is a novel signaling pathway for peptidergic excitation of GnRH neurons.
Collapse
Affiliation(s)
- Chunguang Zhang
- Departments of Physiology and Pharmacology, Oregon National Primate Research Center, Oregon Health and Science University, Portland, OR 97239, USA
| | | | | | | |
Collapse
|
29
|
Ockenga W, Kühne S, Bocksberger S, Banning A, Tikkanen R. Non-neuronal functions of the m2 muscarinic acetylcholine receptor. Genes (Basel) 2013; 4:171-97. [PMID: 24705159 PMCID: PMC3899973 DOI: 10.3390/genes4020171] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 03/10/2013] [Accepted: 03/25/2013] [Indexed: 12/26/2022] Open
Abstract
Acetylcholine is an important neurotransmitter whose effects are mediated by two classes of receptors. The nicotinic acetylcholine receptors are ion channels, whereas the muscarinic receptors belong to the large family of G protein coupled seven transmembrane helix receptors. Beyond its function in neuronal systems, it has become evident that acetylcholine also plays an important role in non-neuronal cells such as epithelial and immune cells. Furthermore, many cell types in the periphery are capable of synthesizing acetylcholine and express at least some of the receptors. In this review, we summarize the non-neuronal functions of the muscarinic acetylcholine receptors, especially those of the M2 muscarinic receptor in epithelial cells. We will review the mechanisms of signaling by the M2 receptor but also the cellular trafficking and ARF6 mediated endocytosis of this receptor, which play an important role in the regulation of signaling events. In addition, we provide an overview of the M2 receptor in human pathological conditions such as autoimmune diseases and cancer.
Collapse
Affiliation(s)
- Wymke Ockenga
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany.
| | - Sina Kühne
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany.
| | - Simone Bocksberger
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany.
| | - Antje Banning
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany.
| | - Ritva Tikkanen
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany.
| |
Collapse
|
30
|
Calcium-activated chloride channel ANO1 promotes breast cancer progression by activating EGFR and CAMK signaling. Proc Natl Acad Sci U S A 2013; 110:E1026-34. [PMID: 23431153 DOI: 10.1073/pnas.1217072110] [Citation(s) in RCA: 250] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The calcium-activated chloride channel anoctamin 1 (ANO1) is located within the 11q13 amplicon, one of the most frequently amplified chromosomal regions in human cancer, but its functional role in tumorigenesis has remained unclear. The 11q13 region is amplified in ∼15% of breast cancers. Whether ANO1 is amplified in breast tumors, the extent to which gene amplification contributes to ANO1 overexpression, and whether overexpression of ANO1 is important for tumor maintenance have remained unknown. We have found that ANO1 is amplified and highly expressed in breast cancer cell lines and primary tumors. Amplification of ANO1 correlated with disease grade and poor prognosis. Knockdown of ANO1 in ANO1-amplified breast cancer cell lines and other cancers bearing 11q13 amplification inhibited proliferation, induced apoptosis, and reduced tumor growth in established cancer xenografts. Moreover, ANO1 chloride channel activity was important for cell viability. Mechanistically, ANO1 knockdown or pharmacological inhibition of its chloride-channel activity reduced EGF receptor (EGFR) and calmodulin-dependent protein kinase II (CAMKII) signaling, which subsequently attenuated AKT, v-src sarcoma viral oncogene homolog (SRC), and extracellular signal-regulated kinase (ERK) activation in vitro and in vivo. Our results highlight the involvement of the ANO1 chloride channel in tumor progression and provide insights into oncogenic signaling in human cancers with 11q13 amplification, thereby establishing ANO1 as a promising target for therapy in these highly prevalent tumor types.
Collapse
|
31
|
Bektas M, Jolly PS, Berkowitz P, Amagai M, Rubenstein DS. A pathophysiologic role for epidermal growth factor receptor in pemphigus acantholysis. J Biol Chem 2013; 288:9447-56. [PMID: 23404504 DOI: 10.1074/jbc.m112.438010] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The pemphigus family of autoimmune bullous disorders is characterized by autoantibody binding to desmoglein 1 and/or 3 (dsg1/dsg3). In this study we show that EGF receptor (EGFR) is activated following pemphigus vulgaris (PV) IgG treatment of primary human keratinocytes and that EGFR activation is downstream of p38 mitogen-activated protein kinase (p38). Inhibition of EGFR blocked PV IgG-triggered dsg3 endocytosis, keratin intermediate filament retraction, and loss of cell-cell adhesion in vitro. Significantly, inhibiting EGFR prevented PV IgG-induced blister formation in the passive transfer mouse model of pemphigus. These data demonstrate cross-talk between dsg3 and EGFR, that this cross-talk is regulated by p38, and that EGFR is a potential therapeutic target for pemphigus. Small-molecule inhibitors and monoclonal antibodies directed against EGFR are currently used to treat several types of solid tumors. This study provides the experimental rationale for investigating the use of EGFR inhibitors in pemphigus.
Collapse
Affiliation(s)
- Meryem Bektas
- Department of Dermatology, University of North Carolina, Chapel Hill, NC 27599-7287, USA
| | | | | | | | | |
Collapse
|
32
|
Kimura R, Okouchi M, Kato T, Imaeda K, Okayama N, Asai K, Joh T. Epidermal growth factor receptor transactivation is necessary for glucagon-like peptide-1 to protect PC12 cells from apoptosis. Neuroendocrinology 2013; 97:300-8. [PMID: 23147408 DOI: 10.1159/000345529] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 10/10/2012] [Indexed: 11/19/2022]
Abstract
AIM Patients with long-standing diabetes commonly develop diabetic encephalopathy, which is characterized by cognitive impairment and dementia. To identify potential treatments for diabetic encephalopathy, we focused on the protective action of glucagon-like peptide-1 (GLP-1) against neural cell apoptosis. In this study, we evaluated whether exposure of cells to GLP-1 leads to epidermal growth factor receptor (EGFR) transactivation and signaling through the PI3K/Akt/mTOR/GCLc/redox pathway, which we previously reported. METHODS We monitored the phosphorylation of EGFR and Akt in PC12 cells exposed to MG and GLP-1 that had been first incubated in the presence or absence of various inhibitors of EGFR transactivation. RESULTS DAPI staining revealed that pretreatment of cells with BiPS, HB-EGF and anti-TGF-α neutralization antibodies or AG1478 abrogated the ability of GLP-1 to rescue cells from MG-induced apoptosis. We show that exposure of PC12 cells to GLP-1 induces EGFR phosphorylation and that this effect was inhibited by prior exposure of the cells to BiPS, HB-EGF and anti-TGF-α neutralization antibodies or AG1478. Interestingly, these agents also diminished the capacity of GLP-1 to protect cells from MG-induced apoptosis. Moreover, these agents reduced GLP-1-induced phosphorylation of Akt. EGF itself also protected the cells from MG-induced apoptosis and induced phosphorylation of Akt, which was inhibited by LY294002. CONCLUSION The neuroprotective effects of GLP-1 against MG-induced apoptosis are mediated by EGFR transactivation, which signals through the PI3K/Akt/mTOR/GCLc/redox pathway in PC12 cells.
Collapse
Affiliation(s)
- Ryosuke Kimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan. CQR00501 @ nifty.com
| | | | | | | | | | | | | |
Collapse
|
33
|
Effect of Bronchodilators on Bronchial Gland Cell Proliferation After Inhalation and Burn Injury in Sheep. J Burn Care Res 2013; 34:386-93. [DOI: 10.1097/bcr.0b013e31826fc51e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
34
|
G Protein-Coupled Receptors in cancer: biochemical interactions and drug design. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 115:143-73. [PMID: 23415094 DOI: 10.1016/b978-0-12-394587-7.00004-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
G Protein-Coupled Receptors (GPCRs) share the same topology made of seven-transmembrane segments and represent the largest family of membrane receptors. Initially associated with signal transduction in differentiated cells, GPCRs and heterotrimeric G proteins were shown to behave as proto-oncogenes whose overexpression or activating mutations confer transforming properties. The first part of this review focuses on the link between biochemical interactions of a GPCR with other receptors, such as dimerization or multiprotein complexes, and their oncogenic properties. Alteration of these interactions or deregulation of transduction cascades can promote uncontrolled cell proliferation or cell transformation that leads to tumorigenicity and malignancy. The second part concerns the design of drugs specifically targeting these complex interactions and their promise in cancer therapy.
Collapse
|
35
|
3′,4′-Dimethoxythioflavone induces endothelium-dependent vasorelaxation through activation of epidermal growth factor receptor. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2012; 386:339-50. [DOI: 10.1007/s00210-012-0818-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 11/27/2012] [Indexed: 11/26/2022]
|
36
|
Lanzafame AA, Christopoulos A, Mitchelson F. Cellular Signaling Mechanisms for Muscarinic Acetylcholine Receptors. ACTA ACUST UNITED AC 2011. [DOI: 10.3109/10606820308263] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
37
|
Regulation of PMA-induced MUC5AC expression by heparin in human bronchial epithelial cells. Mol Cell Biochem 2011; 360:383-91. [PMID: 21984036 DOI: 10.1007/s11010-011-1078-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 09/16/2011] [Indexed: 10/17/2022]
Abstract
Mucus hypersecretion is a major pathophysiologic feature in chronic inflammatory airway diseases. Oxidative stress plays a pivotal role in this process. Recent studies have found that heparin has antioxidant effects which can reduce free radical damage. Here, we hypothesized that heparin has some influence on the expression of mucin 5AC (MUC5AC) induced by phorbol myristate acetate (PMA) in a bronchial epithelial cell line (HBE16), also we have investigated the potential mechanism involved in the process. We found that ROS, the mRNA of Duox1, EGFR and MUC5AC, as well as the protein levels of Duox1, p-EGFR, EGFR, and MUC5AC in the PMA group were significantly increased when compared with the control group (all P < 0.01). After pretreatment with heparin however, there was a significant decrease in ROS levels, the mRNA of Duox1, EGFR, and MUC5AC, and the protein levels of Duox1, p-EGFR, EGFR, and MUC5AC, when compared with the PMA group (all P < 0.01). MUC5AC protein in the supernatant was inhibited in a dose-dependent manner by heparin. Pretreatment with DMTU resulted in a significant decrease in ROS content, the mRNA of Duox1, EGFR, and MUC5AC as well as the protein levels of Duox1, p-EGFR, EGFR, and MUC5AC when compared with the PMA group (all P < 0.01). When cells were pretreated with both heparin and DMTU, there was a further reduction in ROS content, the mRNA of Duox1, EGFR, and MUC5AC as well as the protein levels of Duox1, p-EGFR, EGFR, and MUC5AC, when compared with either the PMA group, heparin group, or DMTU group (all P < 0.01). Our results show that PMA can induce MUC5AC expression by activation of the Duox1-ROS-TACE-TGF-α-EGFR signaling pathway. Heparin can decrease the level of Duox1, ROS production and block the PMA-induced activation of EGFR, thus inhibiting the overexpression of mucin MUC5AC in a dose-dependent manner. In addition to reducing ROS production, heparin may also inhibit the expression of MUC5AC through other signal mechanisms.
Collapse
|
38
|
Ulu N, Gurdal H, Landheer SW, Duin M, Guc MO, Buikema H, Henning RH. α1-Adrenoceptor-mediated contraction of rat aorta is partly mediated via transactivation of the epidermal growth factor receptor. Br J Pharmacol 2011; 161:1301-10. [PMID: 20977469 DOI: 10.1111/j.1476-5381.2010.00829.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE High level of plasma catecholamines is a risk factor for vascular diseases such as hypertension and atherosclerosis. Catecholamines induce hypertrophy of vascular smooth muscle through α(1) -adrenoceptors, which in cell culture involves the transactivation of epidermal growth factor receptor (EGFR). We hypothesized that EGFR transactivation was also involved in contractions of rat aorta mediated by α(1) -adrenoceptors. EXPERIMENTAL APPROACH Thoracic aorta was isolated from 12-14 week old male Wistar rats. In vitro aortic contractile responses to cumulative doses of phenylephrine were characterized in the absence and presence of the EGFR kinase inhibitors, AG1478 and DAPH, in intact and endothelium-denuded rings. Involvement of signal transduction pathways was investigated by using heparin and inhibitors of Src, matrix metalloproteinase (MMP), extracellular signal-regulated kinase (ERK)1/2 and phosphatidyl inositol 3-kinase (PI3K). Phosphorylation of EGFR and ERK1/2 was measured after short-term phenylephrine or EGF stimulation in aorta segments in the presence of AG1478 and the PI3K inhibitor, wortmannin. KEY RESULTS AG1478 and DAPH concentration dependently attenuated phenylephrine-induced contractile responses in intact or endothelium-denuded aortic rings. Inhibition of PI3K (wortmannin and LY294002) but not heparin or inhibitors of Src or MMP, prevented the effect of AG1478 on the responses to phenylephrine. Phenylephrine induced phosphorylation of EGFR, which was partially blocked by AG1478. Phenylephrine also increased phosphorylation of ERK1/2, time-dependently and was blocked by AG1478 and wortmannin. CONCLUSIONS AND IMPLICATIONS Contractions of rat thoracic aorta mediated by α(1) -adrenoceptors involved transactivation of EGFR, mediated via a PI3K and ERK1/2 dependent pathway.
Collapse
Affiliation(s)
- N Ulu
- Department of Clinical Pharmacology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands.
| | | | | | | | | | | | | |
Collapse
|
39
|
Gu C, Barry J. Function and mechanism of axonal targeting of voltage-sensitive potassium channels. Prog Neurobiol 2011; 94:115-32. [PMID: 21530607 DOI: 10.1016/j.pneurobio.2011.04.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Revised: 03/22/2011] [Accepted: 04/01/2011] [Indexed: 12/20/2022]
Abstract
Precise localization of various ion channels into proper subcellular compartments is crucial for neuronal excitability and synaptic transmission. Axonal K(+) channels that are activated by depolarization of the membrane potential participate in the repolarizing phase of the action potential, and hence regulate action potential firing patterns, which encode output signals. Moreover, some of these channels can directly control neurotransmitter release at axonal terminals by constraining local membrane excitability and limiting Ca(2+) influx. K(+) channels differ not only in biophysical and pharmacological properties, but in expression and subcellular distribution as well. Importantly, proper targeting of channel proteins is a prerequisite for electrical and chemical functions of axons. In this review, we first highlight recent studies that demonstrate different roles of axonal K(+) channels in the local regulation of axonal excitability. Next, we focus on research progress in identifying axonal targeting motifs and machinery of several different types of K(+) channels present in axons. Regulation of K(+) channel targeting and activity may underlie a novel form of neuronal plasticity. This research field can contribute to generating novel therapeutic strategies through manipulating neuronal excitability in treating neurological diseases, such as multiple sclerosis, neuropathic pain, and Alzheimer's disease.
Collapse
Affiliation(s)
- Chen Gu
- Department of Neuroscience and Center for Molecular Neurobiology, The Ohio State University, Columbus, USA.
| | | |
Collapse
|
40
|
Jung C, Gené GG, Tomás M, Plata C, Selent J, Pastor M, Fandos C, Senti M, Lucas G, Elosua R, Valverde MA. A gain-of-function SNP in TRPC4 cation channel protects against myocardial infarction. Cardiovasc Res 2011; 91:465-71. [PMID: 21427121 DOI: 10.1093/cvr/cvr083] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS The TRPC4 non-selective cation channel is widely expressed in the endothelium, where it generates Ca(2+) signals that participate in the endothelium-mediated vasodilatory response. This study sought to identify single-nucleotide polymorphisms (SNPs) in the TRPC4 gene that are associated with myocardial infarction (MI). METHODS AND RESULTS Our candidate-gene association studies identified a missense SNP (TRPC4-I957V) associated with a reduced risk of MI in diabetic patients [odds ratio (OR) = 0.61; confidence interval (CI), 0.40-0.95, P= 0.02]. TRPC4 was also associated with MI in the Wellcome Trust Case-Control Consortium's genome-wide data: an intronic SNP (rs7319926) within the same linkage disequilibrium block as TRPC4-I957V showed an OR of 0.86 (CI, 0.81-0.94; P =10(-4)). Functional studies of the missense SNP were carried out in HEK293 and CHO cells expressing wild-type or mutant channels. Patch-clamp studies and measurement of intracellular [Ca(2+)] in response to muscarinic agonists and direct G-protein activation showed increased channel activity in TRPC4-I957V-transfected cells compared with TRPC4-WT. Site-directed mutagenesis and molecular modelling of TRPC4-I957V suggested that the gain of function was due to the presence of a less bulky Val-957. This permits a firmer interaction between the TRPC4 and the catalytic site of the tyrosine kinase that phosphorylates TRPC4 at Tyr-959 and facilitates channel insertion into the plasma membrane. CONCLUSION We provide evidence for the association of a TRPC4 SNP with MI in population-based genetic studies. The higher Ca(2+) signals generated by TRPC4-I957V may ultimately facilitate the generation of endothelium- and nitric oxide-dependent vasorelaxation, thereby explaining its protective effect at the vasculature.
Collapse
Affiliation(s)
- Carole Jung
- Laboratory of Molecular Physiology and Channelopathies, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona, C/ Dr. Aiguader 88, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Tamaishi N, Tsukimoto M, Kitami A, Kojima S. P2Y6 receptors and ADAM17 mediate low-dose gamma-ray-induced focus formation (activation) of EGF receptor. Radiat Res 2010; 175:193-200. [PMID: 21268712 DOI: 10.1667/rr2191.1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The EGF receptor (EGFR) is frequently expressed in tumors of epithelial origin. Although it is well known that ionizing radiation induces activation of EGFR, the mechanism remains unknown. Recently, we reported that activation of P2Y receptors is involved in γ-radiation-induced activation of extracellular signal-regulated kinase1/2 (ERK1/2), which is dependent on activation of EGFR. Here we focused on the mechanism of activation of EGFR in response to low-dose γ radiation, mainly in terms of the activation-associated formation of EGFR foci in A549 cells. Irradiation of cells with 0.1 Gy γ rays induced biphasic phosphorylation of EGFR and ERK1/2 as well as biphasic formation of EGFR foci. The radiation-induced focus formation of EGFR was abolished by ecto-nucleotidase, P2Y receptor antagonists and knockdown of P2Y6 receptor, suggesting the involvement of extracellular nucleotides and activation of P2Y6 receptors in this process. Further, a disintegrin and metalloprotease 17 (ADAM17) is expressed in A549 cells and an ADAM17 inhibitor significantly blocked the radiation-induced focus formation of EGFR. We conclude that activation of both P2Y6 receptors and ADAM17 mediates the low-dose γ-radiation-induced activation of EGFR, as evaluated in terms of focus formation, in A549 cells.
Collapse
Affiliation(s)
- Nana Tamaishi
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda-shi, Chiba, Japan
| | | | | | | |
Collapse
|
42
|
Paredes-Gamero EJ, Medeiros VP, Farias EHC, Justo GZ, Trindade ES, Andrade-Lopes AL, Godinho RO, de Miranda A, Ferreira AT, Tersariol ILS, Nader HB. Heparin induces rat aorta relaxation via integrin-dependent activation of muscarinic M3 receptors. Hypertension 2010; 56:713-21. [PMID: 20805441 DOI: 10.1161/hypertensionaha.110.156877] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Previous reports have shown that heparin may promote human hypotension and vascular relaxation by elevation of NO levels through unclear mechanisms. We hypothesized that endothelial muscarinic M(3) receptor activation mediates the heparin-induced vasodilation of rat aortic rings. The experiments were carried out using unfractionated heparin extracted from bovine intestinal mucosa, which elicited an endothelium and NO-dependent relaxation of aortic segments with maximal potency and efficacy (EC(50): 100±10 μmol/L; E(max): 41±3%). Atropine and 1,1-dimethyl-4-diphenylacetoxypiperidinium iodide inhibitors reduced the heparin-dependent relaxation, indicating that M(3) muscarinic receptor is involved in this phenomenon. However, no direct binding of heparin to muscarinic receptors was observed. More importantly, studies performed using the arginine-glycine-aspartic acid peptide and 1-(1,1-dimethylethyl)-3-(1-naphthalenyl)-1H-pyrazolo[3,4-day]pyrimidin-4-amine, an Src family inhibitor, reduced by 51% and 73% the heparin-dependent relaxation, respectively, suggesting the coupling of heparin and M(3) receptor through extracellular matrix molecules and integrin. Furthermore, unfractionated heparin induced activation of focal adhesion protein kinase, Src, and paxillin. Finally, fluorescence resonance energy transfer approach confirmed the interaction of the M(3) receptor to integrin. Taken together, these data demonstrate the participation of M(3) receptor and integrin in heparin-dependent relaxation of vascular smooth muscle. These results provide new insights into the molecular mechanism and potential pharmacological action of heparin in vascular physiology.
Collapse
|
43
|
Chi SS, Vetiska SM, Gill RS, Hsiung MS, Liu F, Van Tol HH. Transactivation of PDGFRbeta by dopamine D4 receptor does not require PDGFRbeta dimerization. Mol Brain 2010; 3:22. [PMID: 20659339 PMCID: PMC2919524 DOI: 10.1186/1756-6606-3-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Accepted: 07/26/2010] [Indexed: 11/25/2022] Open
Abstract
Growth factor-induced receptor dimerization and cross-phosphorylation are hallmarks of signal transduction via receptor tyrosine kinases (RTKs). G protein-coupled receptors (GPCRs) can activate RTKs through a process known as transactivation. The prototypical model of RTK transactivation involves ligand-mediated RTK dimerization and cross-phosphorylation. Here, we show that the platelet-derived growth factor receptor β (PDGFRβ) transactivation by the dopamine receptor D4 (DRD4) is not dependent on ligands for PDGFRβ. Furthermore, when PDGFRβ dimerization is inhibited and receptor phosphorylation is suppressed to near basal levels, the receptor maintains its ability to be transactivated and is still effective in signaling to ERK1/2. Hence, the DRD4-PDGFRβ-ERK1/2 pathway can occur independently of a PDGF-like ligand, PDGFRβ cross-phosphorylation and dimerization, which is distinct from other known forms of transactivation of RTKs by GPCRs.
Collapse
Affiliation(s)
- Sum Shing Chi
- Department of Neuroscience, Centre for Addiction and Mental Health, University of Toronto, Toronto M5T 1R8, Canada
| | | | | | | | | | | |
Collapse
|
44
|
Sharif A, Prevot V. ErbB receptor signaling in astrocytes: a mediator of neuron-glia communication in the mature central nervous system. Neurochem Int 2010; 57:344-58. [PMID: 20685225 DOI: 10.1016/j.neuint.2010.05.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 03/29/2010] [Accepted: 05/18/2010] [Indexed: 10/19/2022]
Abstract
Astrocytes are now recognized as active players in the developing and mature central nervous system. Each astrocyte contacts vascular structures and thousands of synapses within discrete territories. These cells receive a myriad of inputs and generate appropriate responses to regulate the function of brain microdomains. Emerging evidence has implicated receptors of the ErbB tyrosine kinase family in the integration and processing of neuronal inputs by astrocytes: ErbB receptors can be activated by a wide range of neuronal stimuli; they control critical steps of glutamate-glutamine metabolism; and they regulate the biosynthesis and release of various glial-derived neurotrophic factors, gliomediators and gliotransmitters. These key properties of astrocytic ErbB signaling in neuron-glia interactions have significance for the physiology of the mature central nervous system, as exemplified by the central control of reproduction within the hypothalamus, and are also likely to contribute to pathological situations, since both dysregulation of ErbB signaling and glial dysfunction occur in many neurological disorders.
Collapse
Affiliation(s)
- Ariane Sharif
- Inserm, Jean-Pierre Aubert Research Center, U837, Development and Plasticity of the postnatal Brain, Lille, France.
| | | |
Collapse
|
45
|
Ratchford AM, Baker OJ, Camden JM, Rikka S, Petris MJ, Seye CI, Erb L, Weisman GA. P2Y2 nucleotide receptors mediate metalloprotease-dependent phosphorylation of epidermal growth factor receptor and ErbB3 in human salivary gland cells. J Biol Chem 2010; 285:7545-55. [PMID: 20064929 DOI: 10.1074/jbc.m109.078170] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The G protein-coupled receptor P2Y(2) nucleotide receptor (P2Y(2)R) has been shown to be up-regulated in a variety of tissues in response to stress or injury. Recent studies have suggested that P2Y(2)Rs may play a role in immune responses, wound healing, and tissue regeneration via their ability to activate multiple signaling pathways, including activation of growth factor receptors. Here, we demonstrate that in human salivary gland (HSG) cells, activation of the P2Y(2)R by its agonist induces phosphorylation of ERK1/2 via two distinct mechanisms, a rapid, protein kinase C-dependent pathway and a slower and prolonged, epidermal growth factor receptor (EGFR)-dependent pathway. The EGFR-dependent stimulation of UTP-induced ERK1/2 phosphorylation in HSG cells is inhibited by the adamalysin inhibitor tumor necrosis factor-alpha protease inhibitor or by small interfering RNA that selectively silences ADAM10 and ADAM17 expression, suggesting that ADAM metalloproteases are required for P2Y(2)R-mediated activation of the EGFR. G protein-coupled receptors have been shown to promote proteolytic release of EGFR ligands; however, neutralizing antibodies to known ligands of the EGFR did not inhibit UTP-induced EGFR phosphorylation. Immunoprecipitation experiments indicated that UTP causes association of the EGFR with another member of the EGF receptor family, ErbB3. Furthermore, stimulation of HSG cells with UTP induced phosphorylation of ErbB3, and silencing of ErbB3 expression inhibited UTP-induced phosphorylation of both ErbB3 and EGFR. UTP-induced phosphorylation of ErbB3 and EGFR was also inhibited by silencing the expression of the ErbB3 ligand neuregulin 1 (NRG1). These results suggest that P2Y(2)R activation in salivary gland cells promotes the formation of EGFR/ErbB3 heterodimers and metalloprotease-dependent neuregulin 1 release, resulting in the activation of both EGFR and ErbB3.
Collapse
Affiliation(s)
- Ann M Ratchford
- Department of Biochemistry, University of Missouri, Christopher S. Bond Life Sciences Center, Columbia, Missouri 65211, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Kataoka H. EGFR ligands and their signaling scissors, ADAMs, as new molecular targets for anticancer treatments. J Dermatol Sci 2009; 56:148-53. [PMID: 19896805 DOI: 10.1016/j.jdermsci.2009.10.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Accepted: 10/01/2009] [Indexed: 12/17/2022]
Abstract
Members of the human epidermal growth factor receptor (HER) family of receptor tyrosine kinases and their ligands (EGFR ligands) are known to play crucial roles in the regulation of cell proliferation and differentiation, and in the survival of many types of cancer. HER family members are activated in cancer cells and are now considered to be useful molecular targets for cancer therapy. Recently, several new drugs, including monoclonal antibodies and small-molecule inhibitors that target HER members, have been developed and clinically used to treat solid tumors. Members of a disintegrin and metalloproteinase (ADAM) family are thought to mediate the shedding of EGFR ligands and this event is critical for the production of soluble functional EGFR ligands. In melanoma cells, UV irradiation activates some ADAM members and induces melanoma cell growth through EGFR ligand shedding by activated ADAMs. These findings suggest that ADAM inhibitors are also candidate anticancer drugs acting via the blockade of HER family signaling pathways. After shedding of EGFR ligands by ADAMs, the carboxy-terminal fragments (CTFs) of EGFR ligands in the cytoplasm are translocated to the nucleus and induce cell proliferation by binding and exporting repressors and activating cyclin A and c-Myc. Based on these findings, the present molecular targeting therapy against HER members, EGFR and HER2, may not be sufficient, while ADAMs and nuclear translocation of the CTF of EGFR ligands are potential targets for the treatment of cancer, particularly malignancies that are dependent on the EGF family.
Collapse
Affiliation(s)
- Hiromi Kataoka
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.
| |
Collapse
|
47
|
Fedor-Chaiken M, Hein PW, Stewart JC, Brackenbury R, Kinch MS. E-Cadherin Binding Modulates EGF Receptor Activation. ACTA ACUST UNITED AC 2009. [DOI: 10.1080/cac.10.2.105.118] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
48
|
Shin MS, Shinghirunnusorn P, Sugishima Y, Nishimura M, Suzuki S, Koizumi K, Saiki I, Sakurai H. Cross interference with TNF-alpha-induced TAK1 activation via EGFR-mediated p38 phosphorylation of TAK1-binding protein 1. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:1156-64. [PMID: 19393267 DOI: 10.1016/j.bbamcr.2009.04.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2008] [Revised: 03/31/2009] [Accepted: 04/13/2009] [Indexed: 12/17/2022]
Abstract
Transforming growth factor-alpha-activated kinase 1 (TAK1) has been widely recognized as a kinase that regulates multiple intracellular signaling pathways evoked by cytokines and immune receptor activation. We have recently reported that tumor necrosis factor-alpha (TNF-alpha) triggers internalization of epidermal growth factor receptor (EGFR) through a TAK1-p38alpha signaling pathway, which results in a transient suppression of the EGFR. In the present study, we investigated the pathway of intracellular signaling in the opposite direction. Ligand-induced activation of EGFR caused phosphorylation of the TAK1-binding proteins TAB1 and TAB2 in a TAK1-independent manner. EGFR-mediated phosphorylation of TAB1 was completely inhibited by a chemical inhibitor and siRNA of p38alpha. The phosphorylation of TAB1 was occurred at Ser-423 and Thr-431, the residues underlying the p38-mediated feedback inhibition of TAK1. In contrast, phosphorylation of TAB2 was sustained, and largely resistant to p38 inhibition. The inducible phosphorylation of TAB1 interfered with a response of EGF-treated cells to TNF-alpha-induced TAK1 activation, which led to the reduction of NF-kappaB activation. Collectively, these results demonstrated that EGFR activation interfered with TNF-alpha-induced TAK1 activation via p38-mediated phosphorylation of TAB1.
Collapse
Affiliation(s)
- Myoung-Sook Shin
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Signaling through the cyclic adenosine monophosphate-dependent protein kinase [protein kinase A (PKA)] is an important and widely studied area of signal transduction research. This signaling pathway is commonly investigated through the use of the pharmacological PKA inhibitors H89 and KT 5720. Both of these compounds are thought to block PKA actions through competitive inhibition of the adenosine triphosphate site on the PKA catalytic subunit. Recently, a number of studies have identified actions of H89 and KT 5720 that are independent of their effects on PKA. These nonspecific effects are widespread; they include actions on other protein kinases and signaling molecules and also on basic cellular functions, such as transcription. Here, I summarize the nonspecific effects of these two compounds and compare their actions with those of other PKA inhibitors.
Collapse
Affiliation(s)
- Andrew J Murray
- School of Medical Sciences, College of Life Sciences and Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.
| |
Collapse
|
50
|
Buvinic S, Bravo-Zehnder M, Boyer JL, Huidobro-Toro JP, González A. Nucleotide P2Y1 receptor regulates EGF receptor mitogenic signaling and expression in epithelial cells. J Cell Sci 2008; 120:4289-301. [PMID: 18057028 DOI: 10.1242/jcs.03490] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) function is transregulated by a variety of stimuli, including agonists of certain G-protein-coupled receptors (GPCRs). One of the most ubiquitous GPCRs is the P2Y(1) receptor (P2RY1, hereafter referred to as P2Y(1)R) for extracellular nucleotides, mainly ADP. Here, we show in tumoral HeLa cells and normal FRT epithelial cells that P2Y(1)R broadcasts mitogenic signals by transactivating the EGFR. The pathway involves PKC, Src and cell surface metalloproteases. Stimulation of P2Y(1)R for as little as 15-60 minutes triggers mitogenesis, mirroring the half-life of extracellular ADP. Apyrase degradation of extracellular nucleotides and drug inhibition of P2Y(1)R, both reduced basal cell proliferation of HeLa and FRT cells, but not MDCK cells, which do not express P2Y(1)R. Thus, cell-released nucleotides constitute strong mitogenic stimuli, which act via P2Y(1)R. Strikingly, MDCK cells ectopically expressing P2Y(1)R display a highly proliferative phenotype that depends on EGFR activity associated with an increased level of EGFR, thus disclosing a novel aspect of GPCR-mediated regulation of EGFR function. These results highlight a role of P2Y(1)R in EGFR-dependent epithelial cell proliferation. P2Y(1)R could potentially mediate both trophic stimuli of basally released nucleotides and first-line mitogenic stimulation upon tissue damage. It could also contribute to carcinogenesis and serve as target for antitumor therapies.
Collapse
Affiliation(s)
- Sonja Buvinic
- Centro de Regulación Celular y Patología JV Luco, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8330033, Santiago, Chile
| | | | | | | | | |
Collapse
|