1
|
Bunch H, Kim D, Naganuma M, Nakagawa R, Cong A, Jeong J, Ehara H, Vu H, Chang JH, Schellenberg MJ, Sekine SI. ERK2-topoisomerase II regulatory axis is important for gene activation in immediate early genes. Nat Commun 2023; 14:8341. [PMID: 38097570 PMCID: PMC10721843 DOI: 10.1038/s41467-023-44089-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/29/2023] [Indexed: 12/17/2023] Open
Abstract
The function of the mitogen-activated protein kinase signaling pathway is required for the activation of immediate early genes (IEGs), including EGR1 and FOS, for cell growth and proliferation. Recent studies have identified topoisomerase II (TOP2) as one of the important regulators of the transcriptional activation of IEGs. However, the mechanism underlying transcriptional regulation involving TOP2 in IEG activation has remained unknown. Here, we demonstrate that ERK2, but not ERK1, is important for IEG transcriptional activation and report a critical ELK1 binding sequence for ERK2 function at the EGR1 gene. Our data indicate that both ERK1 and ERK2 extensively phosphorylate the C-terminal domain of TOP2B at mutual and distinctive residues. Although both ERK1 and ERK2 enhance the catalytic rate of TOP2B required to relax positive DNA supercoiling, ERK2 delays TOP2B catalysis of negative DNA supercoiling. In addition, ERK1 may relax DNA supercoiling by itself. ERK2 catalytic inhibition or knock-down interferes with transcription and deregulates TOP2B in IEGs. Furthermore, we present the first cryo-EM structure of the human cell-purified TOP2B and etoposide together with the EGR1 transcriptional start site (-30 to +20) that has the strongest affinity to TOP2B within -423 to +332. The structure shows TOP2B-mediated breakage and dramatic bending of the DNA. Transcription is activated by etoposide, while it is inhibited by ICRF193 at EGR1 and FOS, suggesting that TOP2B-mediated DNA break to favor transcriptional activation. Taken together, this study suggests that activated ERK2 phosphorylates TOP2B to regulate TOP2-DNA interactions and favor transcriptional activation in IEGs. We propose that TOP2B association, catalysis, and dissociation on its substrate DNA are important processes for regulating transcription and that ERK2-mediated TOP2B phosphorylation may be key for the catalysis and dissociation steps.
Collapse
Affiliation(s)
- Heeyoun Bunch
- Department of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea.
- School of Applied Biosciences, College of Agriculture & Life Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea.
| | - Deukyeong Kim
- School of Applied Biosciences, College of Agriculture & Life Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Masahiro Naganuma
- Laboratory for Transcription Structural Biology, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Reiko Nakagawa
- RIKEN BDR Laboratory for Phyloinformatics, Hyogo, 650-0047, Japan
| | - Anh Cong
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jaehyeon Jeong
- Department of Applied Biosciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Haruhiko Ehara
- Laboratory for Transcription Structural Biology, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Hongha Vu
- Department of Biology Education, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Jeong Ho Chang
- Department of Biology Education, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Matthew J Schellenberg
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Shun-Ichi Sekine
- Laboratory for Transcription Structural Biology, RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| |
Collapse
|
2
|
Sarkar M, Martufi M, Roman-Trufero M, Wang YF, Whilding C, Dormann D, Sabbattini P, Dillon N. CNOT3 interacts with the Aurora B and MAPK/ERK kinases to promote survival of differentiating mesendodermal progenitor cells. Mol Biol Cell 2021; 32:ar40. [PMID: 34613789 PMCID: PMC8694085 DOI: 10.1091/mbc.e21-02-0089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 09/23/2021] [Accepted: 10/01/2021] [Indexed: 11/18/2022] Open
Abstract
Mesendoderm cells are key intermediate progenitors that form at the early primitive streak (PrS) and give rise to mesoderm and endoderm in the gastrulating embryo. We have identified an interaction between CNOT3 and the cell cycle kinase Aurora B that requires sequences in the NOT box domain of CNOT3 and regulates MAPK/ERK signaling during mesendoderm differentiation. Aurora B phosphorylates CNOT3 at two sites located close to a nuclear localization signal and promotes localization of CNOT3 to the nuclei of mouse embryonic stem cells (ESCs) and metastatic lung cancer cells. ESCs that have both sites mutated give rise to embryoid bodies that are largely devoid of mesoderm and endoderm and are composed mainly of cells with ectodermal characteristics. The mutant ESCs are also compromised in their ability to differentiate into mesendoderm in response to FGF2, BMP4, and Wnt3 due to reduced survival and proliferation of differentiating mesendoderm cells. We also show that the double mutation alters the balance of interaction of CNOT3 with Aurora B and with ERK and reduces phosphorylation of ERK in response to FGF2. Our results identify a potential adaptor function for CNOT3 that regulates the Ras/MEK/ERK pathway during embryogenesis.
Collapse
Affiliation(s)
- Moumita Sarkar
- Gene Regulation and Chromatin Group, Imperial College London, London W12 0NN, UK
| | - Matteo Martufi
- Gene Regulation and Chromatin Group, Imperial College London, London W12 0NN, UK
| | - Monica Roman-Trufero
- Gene Regulation and Chromatin Group, Imperial College London, London W12 0NN, UK
| | - Yi-Fang Wang
- Bioinformatics and Computing, Imperial College London, London W12 0NN, UK
| | - Chad Whilding
- Microscopy Facility, MRC London Institute of Medical Sciences, Imperial College London, London W12 0NN, UK
| | - Dirk Dormann
- Microscopy Facility, MRC London Institute of Medical Sciences, Imperial College London, London W12 0NN, UK
| | | | - Niall Dillon
- Gene Regulation and Chromatin Group, Imperial College London, London W12 0NN, UK
- Institute of Clinical Sciences, Imperial College London, London W12 0NN, UK
| |
Collapse
|
3
|
Yoshizawa R, Umeki N, Yamamoto A, Okada M, Murata M, Sako Y. p52Shc regulates the sustainability of ERK activation in a RAF-independent manner. Mol Biol Cell 2021; 32:1838-1848. [PMID: 34260260 PMCID: PMC8684710 DOI: 10.1091/mbc.e21-01-0007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
p52SHC (SHC) and GRB2 are adaptor proteins involved in the RAS/MAPK (ERK) pathway mediating signals from cell-surface receptors to various cytoplasmic proteins. To further examine their roles in signal transduction, we studied the translocation of fluorescently labeled SHC and GRB2 to the cell surface, caused by the activation of ERBB receptors by heregulin (HRG). We simultaneously evaluated activated ERK translocation to the nucleus. Unexpectedly, the translocation dynamics of SHC were sustained when those of GRB2 were transient. The sustained localization of SHC positively correlated with the sustained nuclear localization of ERK, which became more transient after SHC knockdown. SHC-mediated PI3K activation was required to maintain the sustainability of the ERK translocation regulating MEK but not RAF. In cells overexpressing ERBB1, SHC translocation became transient, and the HRG-induced cell fate shifted from a differentiation to a proliferation bias. Our results indicate that SHC and GRB2 functions are not redundant but that SHC plays the critical role in the temporal regulation of ERK activation.
Collapse
Affiliation(s)
- Ryo Yoshizawa
- Cellular Informatics Lab, RIKEN, Wako, Saitama 351-0198, Japan.,Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Komaba, Meguro, Tokyo 153-8902, Japan
| | - Nobuhisa Umeki
- Cellular Informatics Lab, RIKEN, Wako, Saitama 351-0198, Japan
| | | | - Mariko Okada
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan.,Center for Drug Design and Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki 567-0085, Japan
| | - Masayuki Murata
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Komaba, Meguro, Tokyo 153-8902, Japan
| | - Yasushi Sako
- Cellular Informatics Lab, RIKEN, Wako, Saitama 351-0198, Japan
| |
Collapse
|
4
|
Jurado M, Castaño Ó, Zorzano A. Stochastic modulation evidences a transitory EGF-Ras-ERK MAPK activity induced by PRMT5. Comput Biol Med 2021; 133:104339. [PMID: 33910125 DOI: 10.1016/j.compbiomed.2021.104339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 03/06/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023]
Abstract
The extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK) pathway involves a three-step cascade of kinases that transduce signals and promote processes such as cell growth, development, and apoptosis. An aberrant response of this pathway is related to the proliferation of cell diseases and tumors. By using simulation modeling, we document that the protein arginine methyltransferase 5 (PRMT5) modulates the MAPK pathway and thus avoids an aberrant behavior. PRMT5 methylates the Raf kinase, reducing its catalytic activity and thereby, reducing the activation of ERK in time and amplitude. Two minimal computational models of the epidermal growth factor (EGF)-Ras-ERK MAPK pathway influenced by PRMT5 were proposed: a first model in which PRMT5 is activated by EGF and a second one in which PRMT5 is stimulated by the cascade response. The reported results show that PRMT5 reduces the time duration and the expression of the activated ERK in both cases, but only in the first model PRMT5 limits the EGF range that generates an ERK activation. Based on our data, we propose the protein PRMT5 as a regulatory factor to develop strategies to fight against an excessive activity of the MAPK pathway, which could be of use in chronic diseases and cancer.
Collapse
Affiliation(s)
- Manuel Jurado
- Biotechnology Ph.D. Programme, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Óscar Castaño
- Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain; Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain; CIBER in Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid, Spain; Bioelectronics Unit and Nanobioengineering Lab., Institute for Nanoscience and Nanotechnology of the University of Barcelona (IN2UB), Barcelona, Spain.
| | - Antonio Zorzano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; CIBER of Diabetes and Associated Metabolic Diseases, Barcelona, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
| |
Collapse
|
5
|
Miao T, Little AC, Aronshtam A, Marquis T, Fenn SL, Hristova M, Krementsov DN, van der Vliet A, Spees JL, Oldinski RA. Internalized FGF-2-Loaded Nanoparticles Increase Nuclear ERK1/2 Content and Result in Lung Cancer Cell Death. NANOMATERIALS 2020; 10:nano10040612. [PMID: 32230722 PMCID: PMC7221911 DOI: 10.3390/nano10040612] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/13/2020] [Accepted: 03/16/2020] [Indexed: 12/15/2022]
Abstract
Innovative cancer treatments, which improve adjuvant therapy and reduce adverse events, are desperately needed. Nanoparticles provide controlled intracellular biomolecule delivery in the absence of activating external cell surface receptors. Prior reports suggest that intracrine signaling, following overexpression of basic fibroblast growth factor (FGF-2) after viral transduction, has a toxic effect on diseased cells. Herein, the research goals were to (1) encapsulate recombinant FGF-2 within stable, alginate-based nanoparticles (ABNs) for non-specific cellular uptake, and (2) determine the effects of ABN-mediated intracellular delivery of FGF-2 on cancer cell proliferation/survival. In culture, human alveolar adenocarcinoma basal epithelial cell line (A549s) and immortalized human bronchial epithelial cell line (HBE1s) internalized ABNs through non-selective endocytosis. Compared to A549s exposed to empty (i.e., blank) ABNs, the intracellular delivery of FGF-2 via ABNs significantly increased the levels of lactate dehydrogenase, indicating that FGF-2-ABN treatment decreased the transformed cell integrity. Noticeably, the nontransformed cells were not significantly affected by FGF-2-loaded ABN treatment. Furthermore, FGF-2-loaded ABNs significantly increased nuclear levels of activated-extracellular signal-regulated kinase ½ (ERK1/2) in A549s but had no significant effect on HBE1 nuclear ERK1/2 expression. Our novel intracellular delivery method of FGF-2 via nanoparticles resulted in increased cancer cell death via increased nuclear ERK1/2 activation.
Collapse
Affiliation(s)
- Tianxin Miao
- Bioengineering Program, College of Engineering and Mathematical Sciences, Larner College of Medicine, College of Engineering and Mathematical Sciences, University of Vermont, Burlington, VT 05405, USA (S.L.F.)
| | - Andrew C. Little
- Cellular, Molecular and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, USA; (A.C.L.); (A.v.d.V.)
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA;
| | - Alexander Aronshtam
- Department of Medicine, Stem Cell Core, Larner College of Medicine, University of Vermont, Colchester, VT 05446, USA; (A.A.); (T.M.)
| | - Taylor Marquis
- Department of Medicine, Stem Cell Core, Larner College of Medicine, University of Vermont, Colchester, VT 05446, USA; (A.A.); (T.M.)
| | - Spencer L. Fenn
- Bioengineering Program, College of Engineering and Mathematical Sciences, Larner College of Medicine, College of Engineering and Mathematical Sciences, University of Vermont, Burlington, VT 05405, USA (S.L.F.)
| | - Milena Hristova
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA;
| | - Dimitry N. Krementsov
- Department of Biomedical and Health Sciences, College of Nursing and Health Sciences, University of Vermont, Burlington, VT 05405, USA;
| | - Albert van der Vliet
- Cellular, Molecular and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, USA; (A.C.L.); (A.v.d.V.)
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA;
| | - Jeffrey L. Spees
- Cellular, Molecular and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, USA; (A.C.L.); (A.v.d.V.)
- Department of Medicine, Stem Cell Core, Larner College of Medicine, University of Vermont, Colchester, VT 05446, USA; (A.A.); (T.M.)
- Correspondence: (J.L.S.); (R.A.O.); Tel.: +1-802-656-2388 (J.L.S.); +1-802-656-3338 (R.A.O.); Fax: +1-802-656-8932 (J.L.S.); +1-802-656-3358 (R.A.O.)
| | - Rachael A. Oldinski
- Bioengineering Program, College of Engineering and Mathematical Sciences, Larner College of Medicine, College of Engineering and Mathematical Sciences, University of Vermont, Burlington, VT 05405, USA (S.L.F.)
- Department of Mechanical Engineering, College of Engineering and Mathematical Sciences, University of Vermont, Burlington, VT 05405, USA
- Department of Electrical and Biomedical Engineering, College of Engineering and Mathematical Sciences, University of Vermont, Burlington, VT 05405, USA
- Materials Science Program, College of Engineering and Mathematical Sciences, University of Vermont, Burlington, VT 05405, USA
- Correspondence: (J.L.S.); (R.A.O.); Tel.: +1-802-656-2388 (J.L.S.); +1-802-656-3338 (R.A.O.); Fax: +1-802-656-8932 (J.L.S.); +1-802-656-3358 (R.A.O.)
| |
Collapse
|
6
|
Morales ML, Arenas A, Ortiz-Ruiz A, Leivas A, Rapado I, Rodríguez-García A, Castro N, Zagorac I, Quintela-Fandino M, Gómez-López G, Gallardo M, Ayala R, Linares M, Martínez-López J. MEK inhibition enhances the response to tyrosine kinase inhibitors in acute myeloid leukemia. Sci Rep 2019; 9:18630. [PMID: 31819100 PMCID: PMC6901485 DOI: 10.1038/s41598-019-54901-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 11/19/2019] [Indexed: 12/28/2022] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3) is a key driver of acute myeloid leukemia (AML). Several tyrosine kinase inhibitors (TKIs) targeting FLT3 have been evaluated clinically, but their effects are limited when used in monotherapy due to the emergence of drug-resistance. Thus, a better understanding of drug-resistance pathways could be a good strategy to explore and evaluate new combinational therapies for AML. Here, we used phosphoproteomics to identify differentially-phosphorylated proteins in patients with AML and TKI resistance. We then studied resistance mechanisms in vitro and evaluated the efficacy and safety of rational combinational therapy in vitro, ex vivo and in vivo in mice. Proteomic and immunohistochemical studies showed the sustained activation of ERK1/2 in bone marrow samples of patients with AML after developing resistance to FLT3 inhibitors, which was identified as a common resistance pathway. We examined the concomitant inhibition of MEK-ERK1/2 and FLT3 as a strategy to overcome drug-resistance, finding that the MEK inhibitor trametinib remained potent in TKI-resistant cells and exerted strong synergy when combined with the TKI midostaurin in cells with mutated and wild-type FLT3. Importantly, this combination was not toxic to CD34+ cells from healthy donors, but produced survival improvements in vivo when compared with single therapy groups. Thus, our data point to trametinib plus midostaurin as a potentially beneficial therapy in patients with AML.
Collapse
Affiliation(s)
- María Luz Morales
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Alicia Arenas
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Alejandra Ortiz-Ruiz
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Alejandra Leivas
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Inmaculada Rapado
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
- Servicio de Hematología, Hospital 12 de Octubre, Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), ISCIII, Madrid, Spain
| | - Alba Rodríguez-García
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Nerea Castro
- Servicio de Hematología, Hospital 12 de Octubre, Madrid, Spain
| | - Ivana Zagorac
- Breast Cancer Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Miguel Quintela-Fandino
- Breast Cancer Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Gonzalo Gómez-López
- Bioinformatics Unit, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Miguel Gallardo
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Rosa Ayala
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
- Servicio de Hematología, Hospital 12 de Octubre, Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), ISCIII, Madrid, Spain
- Universidad Complutense de Madrid, Madrid, Spain
| | - María Linares
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain.
- Universidad Complutense de Madrid, Madrid, Spain.
| | - Joaquín Martínez-López
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
- Servicio de Hematología, Hospital 12 de Octubre, Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), ISCIII, Madrid, Spain
- Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
7
|
Chen Z, Zhang W, Zhang N, Zhou Y, Hu G, Xue M, Liu J, Li Y. Down-regulation of insulin-like growth factor binding protein 5 is involved in intervertebral disc degeneration via the ERK signalling pathway. J Cell Mol Med 2019; 23:6368-6377. [PMID: 31290273 PMCID: PMC6714225 DOI: 10.1111/jcmm.14525] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 06/07/2019] [Accepted: 06/11/2019] [Indexed: 12/27/2022] Open
Abstract
It is obvious that epigenetic processes influence the evolution of intervertebral disc degeneration (IDD). However, its molecular mechanisms are poorly understood. Therefore, we tested the hypothesis that IGFBP5, a potential regulator of IDD, modulates IDD via the ERK signalling pathway. We showed that IGFBP5 mRNA was significantly down-regulated in degenerative nucleus pulposus (NP) tissues. IGFBP5 was shown to significantly promote NP cell proliferation and inhibit apoptosis in vitro, which was confirmed by MTT, flow cytometry and colony formation assays. Furthermore, IGFBP5 was shown to exert its effects by inhibiting the ERK signalling pathway. The effects induced by IGFBP5 overexpression on NP cells were similar to those induced by treatment with an ERK pathway inhibitor (PD98059). Moreover, qRT-PCR and Western blot analyses were performed to examine the levels of apoptosis-related factors, including Bax, caspase-3 and Bcl2. The silencing of IGFBP5 up-regulated the levels of Bax and caspase-3 and down-regulated the level of Bcl2, thereby contributing to the development of human IDD. Furthermore, these results were confirmed in vivo using an IDD rat model, which showed that the induction of Igfbp5 mRNA expression abrogated the effects of IGFBP5 silencing on intervertebral discs. Overall, our findings elucidate the role of IGFBP5 in the pathogenesis of IDD and provide a potential novel therapeutic target for IDD.
Collapse
Affiliation(s)
- Zhonghui Chen
- Orthopedic SurgeryRenmin Hospital of Wuhan University, Hubei General HospitalWuchang District, WuhanChina
| | - Weibing Zhang
- Orthopedic SurgeryRenmin Hospital of Wuhan University, Hubei General HospitalWuchang District, WuhanChina
| | - Nu Zhang
- Orthopedic SurgeryRenmin Hospital of Wuhan University, Hubei General HospitalWuchang District, WuhanChina
| | - Yan Zhou
- Orthopedic SurgeryRenmin Hospital of Wuhan University, Hubei General HospitalWuchang District, WuhanChina
| | - Geliang Hu
- Orthopedic SurgeryRenmin Hospital of Wuhan University, Hubei General HospitalWuchang District, WuhanChina
| | - Mingdi Xue
- Orthopedic SurgeryRenmin Hospital of Wuhan University, Hubei General HospitalWuchang District, WuhanChina
| | - Junhua Liu
- Orthopedic SurgeryChibi Third Renmin HospitalChibiChina
| | - Yaming Li
- Orthopedic SurgeryRenmin Hospital of Wuhan University, Hubei General HospitalWuchang District, WuhanChina
| |
Collapse
|
8
|
Hey F, Andreadi C, Noble C, Patel B, Jin H, Kamata T, Straatman K, Luo J, Balmanno K, Jones DT, Collins VP, Cook SJ, Caunt CJ, Pritchard C. Over-expressed, N-terminally truncated BRAF is detected in the nucleus of cells with nuclear phosphorylated MEK and ERK. Heliyon 2018; 4:e01065. [PMID: 30603699 PMCID: PMC6304467 DOI: 10.1016/j.heliyon.2018.e01065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 09/12/2018] [Accepted: 12/14/2018] [Indexed: 12/31/2022] Open
Abstract
BRAF is a cytoplasmic protein kinase, which activates the MEK-ERK signalling pathway. Deregulation of the pathway is associated with the presence of BRAF mutations in human cancer, the most common being V600E BRAF, although structural rearrangements, which remove N-terminal regulatory sequences, have also been reported. RAF-MEK-ERK signalling is normally thought to occur in the cytoplasm of the cell. However, in an investigation of BRAF localisation using fluorescence microscopy combined with subcellular fractionation of Green Fluorescent Protein (GFP)-tagged proteins expressed in NIH3T3 cells, surprisingly, we detected N-terminally truncated BRAF (ΔBRAF) in both nuclear and cytoplasmic compartments. In contrast, ΔCRAF and full-length, wild-type BRAF (WTBRAF) were detected at lower levels in the nucleus while full-length V600EBRAF was virtually excluded from this compartment. Similar results were obtained using ΔBRAF tagged with the hormone-binding domain of the oestrogen receptor (hbER) and with the KIAA1549-ΔBRAF translocation mutant found in human pilocytic astrocytomas. Here we show that GFP-ΔBRAF nuclear translocation does not involve a canonical Nuclear Localisation Signal (NLS), but is suppressed by N-terminal sequences. Nuclear GFP-ΔBRAF retains MEK/ERK activating potential and is associated with the accumulation of phosphorylated MEK and ERK in the nucleus. In contrast, full-length GFP-WTBRAF and GFP-V600EBRAF are associated with the accumulation of phosphorylated ERK but not phosphorylated MEK in the nucleus. These data have implications for cancers bearing single nucleotide variants or N-terminal deleted structural variants of BRAF.
Collapse
Affiliation(s)
- Fiona Hey
- Department of Molecular Cell Biology, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Catherine Andreadi
- Leicester Cancer Research Centre, Clinical Sciences Building, University of Leicester, Leicester Royal Infirmary, Leicester LE2 7LX, UK
| | - Catherine Noble
- Department of Molecular Cell Biology, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Bipin Patel
- Department of Molecular Cell Biology, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Hong Jin
- Department of Molecular Cell Biology, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Tamihiro Kamata
- Leicester Cancer Research Centre, Clinical Sciences Building, University of Leicester, Leicester Royal Infirmary, Leicester LE2 7LX, UK
| | - Kees Straatman
- Core Biotechnology Services, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Jinli Luo
- Leicester Cancer Research Centre, Clinical Sciences Building, University of Leicester, Leicester Royal Infirmary, Leicester LE2 7LX, UK
| | - Kathryn Balmanno
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - David T.W. Jones
- Department of Pathology, Division of Molecular Histopathology, University of Cambridge, Cambridge CB2 0QQ, UK
| | - V. Peter Collins
- Department of Pathology, Division of Molecular Histopathology, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Simon J. Cook
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Christopher J. Caunt
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Catrin Pritchard
- Leicester Cancer Research Centre, Clinical Sciences Building, University of Leicester, Leicester Royal Infirmary, Leicester LE2 7LX, UK
| |
Collapse
|
9
|
Systems Modeling Identifies Divergent Receptor Tyrosine Kinase Reprogramming to MAPK Pathway Inhibition. Cell Mol Bioeng 2018; 11:451-469. [PMID: 30524510 PMCID: PMC6244947 DOI: 10.1007/s12195-018-0542-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 07/17/2018] [Indexed: 12/25/2022] Open
Abstract
Introduction Targeted cancer therapeutics have demonstrated more limited clinical efficacy than anticipated, due to both intrinsic and acquired drug resistance. Underlying mechanisms have been largely attributed to genetic changes, but a substantial proportion of resistance observations remain unexplained by genomic properties. Emerging evidence shows that receptor tyrosine kinase (RTK) reprogramming is a major alternative process causing targeted drug resistance, separate from genetic alterations. Hence, the contributions of mechanisms leading to this process need to be more rigorously assessed. Methods To parse contributions of multiple mechanisms to RTK reprogramming, we have developed a quantitative multi-receptor and multi-mechanistic experimental framework and kinetic model. Results We find that RTK reprogramming mechanisms are disparate among RTKs and nodes of intervention in the MAPK pathway. Mek inhibition induces increased Axl and Her2 levels in triple negative breast cancer (TNBC) cells while Met and EGFR levels remain unchanged, with Axl and Her2 sharing re-wiring through increased synthesis and differing secondary contributing mechanisms. While three Mek inhibitors exhibited mechanistic similarity, three Erk inhibitors elicited effects different from the Mek inhibitors and from each other, with MAPK pathway target-specific effects correlating with Erk subcellular localization. Furthermore, we find that Mek inhibitor-induced RTK reprogramming occurs through both BET bromodomain dependent and independent mechanisms, motivating combination treatment with BET and Axl inhibition to overcome RTK reprogramming. Conclusions Our findings suggest that RTK reprogramming occurs through multiple mechanisms in a MAPK pathway target-specific manner, highlighting the need for comprehensive resistance mechanism profiling strategies during pharmacological development. Electronic supplementary material The online version of this article (10.1007/s12195-018-0542-y) contains supplementary material, which is available to authorized users.
Collapse
|
10
|
Davis TB, Yang M, Schell MJ, Wang H, Ma L, Pledger WJ, Yeatman TJ. PTPRS Regulates Colorectal Cancer RAS Pathway Activity by Inactivating Erk and Preventing Its Nuclear Translocation. Sci Rep 2018; 8:9296. [PMID: 29915291 PMCID: PMC6006154 DOI: 10.1038/s41598-018-27584-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 02/26/2018] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) growth and progression is frequently driven by RAS pathway activation through upstream growth factor receptor activation or through mutational activation of KRAS or BRAF. Here we describe an additional mechanism by which the RAS pathway may be modulated in CRC. PTPRS, a receptor-type protein tyrosine phosphatase, appears to regulate RAS pathway activation through ERK. PTPRS modulates ERK phosphorylation and subsequent translocation to the nucleus. Native mutations in PTPRS, present in ~10% of CRC, may reduce its phosphatase activity while increasing ERK activation and downstream transcriptional signaling.
Collapse
Affiliation(s)
- Thomas B Davis
- Gibbs Cancer Center & Research Institute, 380 Serpentine Drive, Spartanburg, SC, 29303, USA
| | - Mingli Yang
- Gibbs Cancer Center & Research Institute, 380 Serpentine Drive, Spartanburg, SC, 29303, USA
| | - Michael J Schell
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Heiman Wang
- Gibbs Cancer Center & Research Institute, 380 Serpentine Drive, Spartanburg, SC, 29303, USA
| | - Le Ma
- Gibbs Cancer Center & Research Institute, 380 Serpentine Drive, Spartanburg, SC, 29303, USA
| | - W Jack Pledger
- Gibbs Cancer Center & Research Institute, 380 Serpentine Drive, Spartanburg, SC, 29303, USA
- Department of Molecular Medicine, VCOM, 350 Howard Street, Spartanburg, SC, 29303, USA
| | - Timothy J Yeatman
- Gibbs Cancer Center & Research Institute, 380 Serpentine Drive, Spartanburg, SC, 29303, USA.
| |
Collapse
|
11
|
Brietz A, Schuch KV, Wangorsch G, Lorenz K, Dandekar T. Analyzing ERK 1/2 signalling and targets. MOLECULAR BIOSYSTEMS 2017; 12:2436-46. [PMID: 27301697 DOI: 10.1039/c6mb00255b] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The ERK cascade (e.g. Raf-1) protects the heart from cell death and ischemic injury but can also turn maladaptive. Furthermore, an additional autophosphorylation of ERK2 at Thr188 (Erk1 at Thr208) allows ERK to phosphorylate nuclear targets involved in hypertrophy, stressing this additional phosphorylation as a promising pharmacological target. An in silico model was assembled and setup to reproduce different phosphorylation states of ERK 1/2 and various types of stimuli (hypertrophic versus non-hypertrophic). Synergistic and antagonistic receptor stimuli can be predicted in a semi-quantitative model, simulated time courses were experimentally validated. Furthermore, we detected new targets of ERK 1/2, which possibly contribute to the development of pathological hypertrophy. In addition we modeled further interaction partners involved in the protective and maladaptive cascade. Experimental validation included different gene expression data sets supporting key components and novel interaction partners as well as time courses in chronic heart failure.
Collapse
Affiliation(s)
- Alexandra Brietz
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany.
| | | | - Gaby Wangorsch
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany.
| | - Kristina Lorenz
- Biomedizinsche Forschung, Leibniz Institut für Analytische Wissenschaften - ISAS - e.V, Bunsen-Kirchhoff Straße 11, 44139 Dortmund, Germany and West German Heart and Vascular Center Essen, University Hospital Essen-Duisburg, Duisburg, Germany
| | - Thomas Dandekar
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
12
|
Sipthorp J, Lebraud H, Gilley R, Kidger AM, Okkenhaug H, Saba-El-Leil M, Meloche S, Caunt CJ, Cook SJ, Heightman TD. Visualization of Endogenous ERK1/2 in Cells with a Bioorthogonal Covalent Probe. Bioconjug Chem 2017; 28:1677-1683. [PMID: 28449575 DOI: 10.1021/acs.bioconjchem.7b00152] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The RAS-RAF-MEK-ERK pathway has been intensively studied in oncology, with RAS known to be mutated in ∼30% of all human cancers. The recent emergence of ERK1/2 inhibitors and their ongoing clinical investigation demands a better understanding of ERK1/2 behavior following small-molecule inhibition. Although fluorescent fusion proteins and fluorescent antibodies are well-established methods of visualizing proteins, we show that ERK1/2 can be visualized via a less-invasive approach based on a two-step process using inverse electron demand Diels-Alder cycloaddition. Our previously reported trans-cyclooctene-tagged covalent ERK1/2 inhibitor was used in a series of imaging experiments following a click reaction with a tetrazine-tagged fluorescent dye. Although limitations were encountered with this approach, endogenous ERK1/2 was successfully imaged in cells, and "on-target" staining was confirmed by over-expressing DUSP5, a nuclear ERK1/2 phosphatase that anchors ERK1/2 in the nucleus.
Collapse
Affiliation(s)
- James Sipthorp
- Signalling Laboratory, The Babraham Institute , Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Honorine Lebraud
- Astex Pharmaceuticals , 436 Cambridge Science Park, Cambridge CB4 0QA, U.K
| | - Rebecca Gilley
- Signalling Laboratory, The Babraham Institute , Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Andrew M Kidger
- Signalling Laboratory, The Babraham Institute , Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Hanneke Okkenhaug
- Signalling Laboratory, The Babraham Institute , Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Marc Saba-El-Leil
- Institute of Research in Immunology and Cancer and Department of Pharmacology, Université de Montréal , Montréal, Québec, Canada H3C 3J7
| | - Sylvain Meloche
- Institute of Research in Immunology and Cancer and Department of Pharmacology, Université de Montréal , Montréal, Québec, Canada H3C 3J7
| | - Christopher J Caunt
- Department of Biology and Biochemistry, University of Bath , Claverton Down, Bath BA2 7AY, U.K
| | - Simon J Cook
- Signalling Laboratory, The Babraham Institute , Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Tom D Heightman
- Astex Pharmaceuticals , 436 Cambridge Science Park, Cambridge CB4 0QA, U.K
| |
Collapse
|
13
|
Vera M, Biswas J, Senecal A, Singer RH, Park HY. Single-Cell and Single-Molecule Analysis of Gene Expression Regulation. Annu Rev Genet 2017; 50:267-291. [PMID: 27893965 DOI: 10.1146/annurev-genet-120215-034854] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent advancements in single-cell and single-molecule imaging technologies have resolved biological processes in time and space that are fundamental to understanding the regulation of gene expression. Observations of single-molecule events in their cellular context have revealed highly dynamic aspects of transcriptional and post-transcriptional control in eukaryotic cells. This approach can relate transcription with mRNA abundance and lifetimes. Another key aspect of single-cell analysis is the cell-to-cell variability among populations of cells. Definition of heterogeneity has revealed stochastic processes, determined characteristics of under-represented cell types or transitional states, and integrated cellular behaviors in the context of multicellular organisms. In this review, we discuss novel aspects of gene expression of eukaryotic cells and multicellular organisms revealed by the latest advances in single-cell and single-molecule imaging technology.
Collapse
Affiliation(s)
- Maria Vera
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York, NY 10461; , , ,
| | - Jeetayu Biswas
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York, NY 10461; , , ,
| | - Adrien Senecal
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York, NY 10461; , , ,
| | - Robert H Singer
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York, NY 10461; , , , .,Janelia Research Campus of the HHMI, Ashburn, Virginia 20147
| | - Hye Yoon Park
- Department of Physics and Astronomy, Seoul National University, Seoul, 08826, Korea; .,Institute of Molecular Biology and Genetics, Seoul National University, Seoul, 08826, Korea
| |
Collapse
|
14
|
Shindo Y, Iwamoto K, Mouri K, Hibino K, Tomita M, Kosako H, Sako Y, Takahashi K. Conversion of graded phosphorylation into switch-like nuclear translocation via autoregulatory mechanisms in ERK signalling. Nat Commun 2016; 7:10485. [PMID: 26786866 PMCID: PMC4736105 DOI: 10.1038/ncomms10485] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 12/17/2015] [Indexed: 11/29/2022] Open
Abstract
The phosphorylation cascade in the extracellular signal-regulated kinase (ERK) pathway is a versatile reaction network motif that can potentially act as a switch, oscillator or memory. Nevertheless, there is accumulating evidence that the phosphorylation response is mostly linear to extracellular signals in mammalian cells. Here we find that subsequent nuclear translocation gives rise to a switch-like increase in nuclear ERK concentration in response to signal input. The switch-like response disappears in the presence of ERK inhibitor, suggesting the existence of autoregulatory mechanisms for ERK nuclear translocation involved in conversion from a graded to a switch-like response. In vitro reconstruction of ERK nuclear translocation indicates that ERK-mediated phosphorylation of nucleoporins regulates ERK translocation. A mathematical model and knockdown experiments suggest a contribution of nucleoporins to regulation of the ERK nuclear translocation response. Taken together, this study provides evidence that nuclear translocation with autoregulatory mechanisms acts as a switch in ERK signalling. While ERK signalling can produce switch-like cell behaviour, phosphorylation of ERK increases linearly with extracellular signals. Here, the authors solve this seeming contradiction by showing that nuclear translocation of ERK behaves in a switch-like manner and is controlled by ERK activity.
Collapse
Affiliation(s)
- Yuki Shindo
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan.,Laboratory for Biochemical Simulation, RIKEN Quantitative Biology Center, Suita, Osaka 565-0874, Japan
| | - Kazunari Iwamoto
- Laboratory for Biochemical Simulation, RIKEN Quantitative Biology Center, Suita, Osaka 565-0874, Japan
| | - Kazunari Mouri
- Cellular Informatics Laboratory, RIKEN, Wako, Saitama 351-0198, Japan
| | - Kayo Hibino
- Cellular Informatics Laboratory, RIKEN, Wako, Saitama 351-0198, Japan.,Laboratory for Cell Signaling Dynamics, RIKEN Quantitative Biology Center, Suita, Osaka 565-0874, Japan
| | - Masaru Tomita
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0017, Japan
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Yasushi Sako
- Cellular Informatics Laboratory, RIKEN, Wako, Saitama 351-0198, Japan
| | - Koichi Takahashi
- Laboratory for Biochemical Simulation, RIKEN Quantitative Biology Center, Suita, Osaka 565-0874, Japan.,Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0017, Japan
| |
Collapse
|
15
|
Zhang M, Sun F, Chen F, Zhou B, Duan Y, Su H, Lin X. Subcellular proteomic approach for identifying the signaling effectors of protein kinase C-β₂ under high glucose conditions in human umbilical vein endothelial cells. Mol Med Rep 2015; 12:7247-62. [PMID: 26459836 PMCID: PMC4626174 DOI: 10.3892/mmr.2015.4403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 08/05/2015] [Indexed: 11/06/2022] Open
Abstract
The high glucose‑induced activation of protein kinase C‑β2 (PKC‑β2) has an essential role in the pathophysiology of diabetes‑associated vascular disease. In the present study, human umbilical vein endothelial cells (HUVECs) were cultured in high and normal glucose conditions prior to being infected with a recombinant adenovirus to induce the overexpression of PKC‑β2. The activity of PKC‑β2 was also decreased using a selective PKC‑β2 inhibitor. A series of two‑dimensional electrophoresis images detected ~800 spots in the nuclei, and ~600 spots in the cytosol. Following intra‑ and inter‑group cross‑matching, 38 significantly altered spots were identified as high glucose‑induced and PKC‑β2‑associated nuclear proteins. In addition to the observation that the regulation of key proteins involved in the nuclear factor (NF)‑κB signaling cascade occurred in the cytosol, various transcription factors, including peroxisome proliferator‑activated receptor δ (PPAR‑δ), were also altered in the nuclei. A human protein‑protein interaction network of potential connections of PKC‑β2‑associated proteins was constructed in the proteomics investigation using Biological General Repository for Interaction Datasets. The results indicated that PKC‑β2 may be involved in high glucose‑induced glucose and lipid crosstalk by regulating PPAR‑δ. In addition, NF‑κB inhibitor‑interacting Ras‑like protein 1 may be important in the PKC‑β2‑NF‑κB inhibitor‑NF‑κB signaling pathway in HUVECs under high‑glucose conditions.
Collapse
Affiliation(s)
- Min Zhang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Fang Sun
- Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University, Chongqing 400042, P.R. China
| | - Fangfang Chen
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Bo Zhou
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yaqian Duan
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hong Su
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xuebo Lin
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
16
|
Lin CC, Lin WN, Hou WC, Hsiao LD, Yang CM. Endothelin-1 induces VCAM-1 expression-mediated inflammation via receptor tyrosine kinases and Elk/p300 in human tracheal smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2015; 309:L211-25. [PMID: 26071554 DOI: 10.1152/ajplung.00232.2014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 05/14/2015] [Indexed: 11/22/2022] Open
Abstract
The elevated level of endothelin-1 (ET-1) has been detected in the bronchoalveolar lavage of patients with severe asthma, acute lung injury, acute respiratory distress syndrome, and sepsis. ET-1 may affect vessel tone together with lung physiology and pathology. Vascular cell adhesion molecule-1 (VCAM-1) is one kind of adhesion molecules participating in the process of polymorphonuclear leukocyte transmigration and regulating the occurrence and amplification of tissue inflammation. However, the molecular mechanisms underlying ET-1-mediated expression of VCAM-1 on human tracheal smooth muscle cells (HTSMCs) were largely unknown. Here we reported that ET-1 stimulated expression of VCAM-1 gene on HTSMCs, which was blocked by pretreatment with the inhibitors of ET receptors, Src, matrix metalloproteinases (MMPs), epidermal growth factor receptor (EGFR), platelet-derived growth factor receptor (PDGFR), phosphatidylinositol 3-kinase (PI3K), AKT, MEK1/2, and p300, suggesting the participation of these signaling components in ET-1-regulated HTSMC responses. Furthermore, transfection with small-interfering RNA (siRNA) of Src, AKT, p42 mitogen-activated protein kinase (MAPK), or p300 downregulated the respective proteins and significantly attenuated ET-1-induced VCAM-1 expression. ET-1 also stimulated phosphorylation of Src, EGFR, PDGFR, AKT, p42/p44 MAPK, and Elk-1 and acetylation of histone H4 on HTSMCs. Immunoprecipitation assay showed the association between Elk-1 and p300 in the nucleus. Adhesion assay revealed that the adhesion of THP-1 to HTSMCs challenged with ET-1 was increased, which was attenuated by the inhibitors of ET receptors, Src, MMPs, EGFR, PDGFR, PI3K, AKT, p42/p44 MAPK, and p300. Taken together, these data suggested that ET-1 promotes occurrence and amplification of pathology-related airway inflammation via enhancing VCAM-1 expression in an ET receptor/Src/MMP/EGFR, PDGFR/PI3K/AKT/p42/p44 MAPK/Elk-1/p300 pathway in HTSMCs.
Collapse
Affiliation(s)
- Chih-Chung Lin
- Department of Anesthetics, Chang Gung Memorial Hospital and Chang Gung University, Tao-Yuan, Taiwan
| | - Wei-Ning Lin
- Graduate Institute of Basic Medicine, Fu Jen Catholic University, New Taipei City, Taiwan; and
| | - Wei-Chen Hou
- Department of Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Li-Der Hsiao
- Department of Anesthetics, Chang Gung Memorial Hospital and Chang Gung University, Tao-Yuan, Taiwan
| | - Chuen-Mao Yang
- Department of Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| |
Collapse
|
17
|
Vidal M, Wieland T, Lohse MJ, Lorenz K. β-Adrenergic receptor stimulation causes cardiac hypertrophy via a Gβγ/Erk-dependent pathway. Cardiovasc Res 2012; 96:255-64. [PMID: 22843704 DOI: 10.1093/cvr/cvs249] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
AIMS Activation of the β(1)-adrenergic receptor and its G protein, G(s), induces cardiac hypertrophy. However, activation of classic Gα(s) effectors, adenylyl cyclases (AC) and protein kinase A, is not sufficient for induction of hypertrophy, which suggests the involvement of additional pathway(s) activated by G(s). Recently, we discovered that βγ subunits of G(q) induce phosphorylation of the extracellular regulated kinases 1 and 2 (Erk1/2) at threonine188 and thereby induce hypertrophy. Here we investigated whether β-adrenergic receptors might also induce cardiac hypertrophy via Erk(Thr188) phosphorylation. METHODS AND RESULTS β-Adrenergic receptor activation induced Erk(Thr188) phosphorylation in mouse hearts and in neonatal cardiomyocytes. Inhibition of Erk1/2 or overexpression of Erk(Thr188) phosphorylation-deficient mutants (Erk2(T188A) and Erk2(T188S)) significantly attenuated β-adrenergic cardiomyocyte hypertrophy in vitro. Erk activity was stimulated by both isoproterenol and the direct AC activator forskolin, but only isoproterenol induced Erk(Thr188) phosphorylation. Erk(Thr188) phosphorylation required Gβγ released from G(s) and was prevented by Gβγ inhibition. Similarly, isoproterenol, but not forskolin, induced nuclear accumulation of Erk and cardiomyocyte hypertrophy. Long-term application of isoproterenol in mice caused left ventricular hypertrophy and cardiac remodelling, and this was reduced in Erk2(T188S) transgenic mice, supporting the physiological relevance of Erk(Thr188) phosphorylation. CONCLUSIONS Activation of G(s) by β-adrenergic receptors leads to (i) canonical Erk1/2 activation via AC, and (ii) release of Gβγ, which then associates with activated Erk1/2 and induces Erk(Thr188) phosphorylation, causing nuclear accumulation of Erk and ultimately cardiomyocyte hypertrophy. These findings reveal a new pathway critically involved in β-adrenergically mediated cardiac hypertrophy and may yield new therapeutic strategies against hypertrophic remodelling.
Collapse
Affiliation(s)
- Marie Vidal
- Institute of Pharmacology and Toxicology, University of Würzburg, Versbacher Strasse 9, 97078 Würzburg, Germany
| | | | | | | |
Collapse
|
18
|
Harrington HA, Komorowski M, Beguerisse-Díaz M, Ratto GM, Stumpf MPH. Mathematical modeling reveals the functional implications of the different nuclear shuttling rates of Erk1 and Erk2. Phys Biol 2012; 9:036001. [PMID: 22551942 DOI: 10.1088/1478-3975/9/3/036001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The mitogen-activated protein kinase (MAPK) family of proteins is involved in regulating cellular fates such as proliferation, differentiation and apoptosis. In particular, the dynamics of the Erk/Mek system, which has become the canonical example for MAPK signaling systems, have attracted considerable attention. Erk is encoded by two genes, Erk1 and Erk2, that until recently had been considered equivalent as they differ only subtly at the sequence level. However, these proteins exhibit radically different trafficking between cytoplasm and nucleus and this fact may have functional implications. Here we use spatially resolved data on Erk1/2 to develop and analyze spatio-temporal models of these cascades, and we discuss how sensitivity analysis can be used to discriminate between mechanisms. Our models elucidate some of the factors governing the interplay between signaling processes and the Erk1/2 localization in different cellular compartments, including competition between Erk1 and Erk2. Our approach is applicable to a wide range of signaling systems, such as activation cascades, where translocation of molecules occurs. Our study provides a first model of Erk1 and Erk2 activation and their nuclear shuttling dynamics, revealing a role in the regulation of the efficiency of nuclear signaling.
Collapse
|
19
|
Minashima T, Small W, Moss SE, Kirsch T. Intracellular modulation of signaling pathways by annexin A6 regulates terminal differentiation of chondrocytes. J Biol Chem 2012; 287:14803-15. [PMID: 22399299 DOI: 10.1074/jbc.m111.297861] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Annexin A6 (AnxA6) is highly expressed in hypertrophic and terminally differentiated growth plate chondrocytes. Rib chondrocytes isolated from newborn AnxA6-/- mice showed delayed terminal differentiation as indicated by reduced terminal differentiation markers, including alkaline phosphatase, matrix metalloproteases-13, osteocalcin, and runx2, and reduced mineralization. Lack of AnxA6 in chondrocytes led to a decreased intracellular Ca(2+) concentration and protein kinase C α (PKCα) activity, ultimately resulting in reduced extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK) activities. The 45 C-terminal amino acids of AnxA6 (AnxA6(1-627)) were responsible for the direct binding of AnxA6 to PKCα. Consequently, transfection of AnxA6-/- chondrocytes with full-length AnxA6 rescued the reduced expression of terminal differentiation markers, whereas transfection of AnxA6-/- chondrocytes with AnxA6(1-627) did not or only partially rescued the decreased mRNA levels of terminal differentiation markers. In addition, lack of AnxA6 in matrix vesicles, which initiate the mineralization process in growth plate cartilage, resulted in reduced alkaline phosphatase activity and Ca(2+) and inorganic phosphate (P(i)) content and the inability to form hydroxyapatite-like crystals in vitro. Histological analysis of femoral, tibial, and rib growth plates from newborn mice revealed that the hypertrophic zone of growth plates from newborn AnxA6-/- mice was reduced in size. In addition, reduced mineralization was evident in the hypertrophic zone of AnxA6-/- growth plate cartilage, although apoptosis was not altered compared with wild type growth plates. In conclusion, AnxA6 via its stimulatory actions on PKCα and its role in mediating Ca(2+) flux across membranes regulates terminal differentiation and mineralization events of chondrocytes.
Collapse
Affiliation(s)
- Takeshi Minashima
- Musculoskeletal Research Center, Department of Orthopaedic Surgery, New York University Hospital for Joint Diseases, New York, New York 10003, USA
| | | | | | | |
Collapse
|