1
|
Timofeev O, Giron P, Lawo S, Pichler M, Noeparast M. ERK pathway agonism for cancer therapy: evidence, insights, and a target discovery framework. NPJ Precis Oncol 2024; 8:70. [PMID: 38485987 PMCID: PMC10940698 DOI: 10.1038/s41698-024-00554-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/16/2024] [Indexed: 03/18/2024] Open
Abstract
At least 40% of human cancers are associated with aberrant ERK pathway activity (ERKp). Inhibitors targeting various effectors within the ERKp have been developed and explored for over two decades. Conversely, a substantial body of evidence suggests that both normal human cells and, notably to a greater extent, cancer cells exhibit susceptibility to hyperactivation of ERKp. However, this vulnerability of cancer cells remains relatively unexplored. In this review, we reexamine the evidence on the selective lethality of highly elevated ERKp activity in human cancer cells of varying backgrounds. We synthesize the insights proposed for harnessing this vulnerability of ERK-associated cancers for therapeutical approaches and contextualize these insights within established pharmacological cancer-targeting models. Moreover, we compile the intriguing preclinical findings of ERK pathway agonism in diverse cancer models. Lastly, we present a conceptual framework for target discovery regarding ERKp agonism, emphasizing the utilization of mutual exclusivity among oncogenes to develop novel targeted therapies for precision oncology.
Collapse
Affiliation(s)
- Oleg Timofeev
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps University, 35043, Marburg, Germany
| | - Philippe Giron
- Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Clinical Sciences, Research group Genetics, Reproduction and Development, Centre for Medical Genetics, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Steffen Lawo
- CRISPR Screening Core Facility, Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany
| | - Martin Pichler
- Translational Oncology, II. Med Clinics Hematology and Oncology, 86156, Augsburg, Germany
| | - Maxim Noeparast
- Translational Oncology, II. Med Clinics Hematology and Oncology, 86156, Augsburg, Germany.
| |
Collapse
|
2
|
Pandey A, Shen C, Feng S, Enosi Tuipulotu D, Ngo C, Liu C, Kurera M, Mathur A, Venkataraman S, Zhang J, Talaulikar D, Song R, Wong JJL, Teoh N, Kaakoush NO, Man SM. Ku70 senses cytosolic DNA and assembles a tumor-suppressive signalosome. SCIENCE ADVANCES 2024; 10:eadh3409. [PMID: 38277448 PMCID: PMC10816715 DOI: 10.1126/sciadv.adh3409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 12/26/2023] [Indexed: 01/28/2024]
Abstract
The innate immune response contributes to the development or attenuation of acute and chronic diseases, including cancer. Microbial DNA and mislocalized DNA from damaged host cells can activate different host responses that shape disease outcomes. Here, we show that mice and humans lacking a single allele of the DNA repair protein Ku70 had increased susceptibility to the development of intestinal cancer. Mechanistically, Ku70 translocates from the nucleus into the cytoplasm where it binds to cytosolic DNA and interacts with the GTPase Ras and the kinase Raf, forming a tripartite protein complex and docking at Rab5+Rab7+ early-late endosomes. This Ku70-Ras-Raf signalosome activates the MEK-ERK pathways, leading to impaired activation of cell cycle proteins Cdc25A and CDK1, reducing cell proliferation and tumorigenesis. We also identified the domains of Ku70, Ras, and Raf involved in activating the Ku70 signaling pathway. Therapeutics targeting components of the Ku70 signalosome could improve the treatment outcomes in cancer.
Collapse
Affiliation(s)
- Abhimanu Pandey
- Division of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Cheng Shen
- Division of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Shouya Feng
- Division of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Daniel Enosi Tuipulotu
- Division of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Chinh Ngo
- Division of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Cheng Liu
- Conjoint Gastroenterology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia
- School of Medicine, University of Queensland, Herston, Australia
- Mater Pathology, Mater Hospital, South Brisbane, Australia
| | - Melan Kurera
- Division of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Anukriti Mathur
- Division of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Shweta Venkataraman
- Division of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Jing Zhang
- Division of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Dipti Talaulikar
- Division of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
- Haematology Translational Research Unit, ACT Pathology, Canberra Health Services, Canberra, Australian Capital Territory, Australia
- Department of Human Genomics, ACT Pathology, Canberra, Australian Capital Territory, Australia
- School of Medicine and Psychology, College of Health and Medicine, The Australian National University, Canberra, Australia
| | - Renhua Song
- Epigenetics and RNA Biology Program Centenary Institute, The University of Sydney, Camperdown 2050, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown 2050, Australia
| | - Justin J.-L. Wong
- Epigenetics and RNA Biology Program Centenary Institute, The University of Sydney, Camperdown 2050, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown 2050, Australia
| | - Narci Teoh
- Gastroenterology and Hepatology Unit, The Australian National University Medical School at The Canberra Hospital, The Australian National University, Canberra, Australia
| | - Nadeem O. Kaakoush
- School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Si Ming Man
- Division of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| |
Collapse
|
3
|
Ahmad F, Ma L, Wei W, Liu Y, Hakim I, Daugherty A, Mujahid S, Radin AA, Chua MS, So S. Identification and validation of microtubule depolymerizing agent, CYT997, as a potential drug candidate for hepatocellular carcinoma. Liver Int 2023; 43:2794-2807. [PMID: 37833852 DOI: 10.1111/liv.15756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND AND AIMS Hepatocellular carcinoma (HCC) is a typically fatal malignancy with limited treatment options and poor survival rates, despite recent FDA approvals of newer treatment options. We aim to address this unmet need by using a proprietary computational drug discovery platform that identifies drug candidates with the potential to advance rapidly and successfully through preclinical studies. METHODS We generated an in silico model of HCC biology to identify the top 10 small molecules with predicted efficacy. The most promising candidate, CYT997, was tested for its in vitro effects on cell viability and cell death, colony formation, cell cycle changes, and cell migration/invasion in HCC cells. We used an HCC patient-derived xenograft (PDX) mouse model to assess its in vivo efficacy. RESULTS CYT997 was significantly more cytotoxic against HCC cells than against primary human hepatocytes, and sensitized HCC cells to sorafenib. It arrested cell cycle at the G2/M phase with associated up-regulations of p21, p-MEK1/2, p-ERK, and down-regulation of cyclin B1. Cell apoptosis and senescence-like morphology were also observed. CYT997 inhibited HCC cell migration and invasion, and down-regulated the expressions of acetylated tubulins, β-tubulin, glypican-3 (GPC3), β-catenin, and c-Myc. In vivo, CYT997 (20 mg/kg, three times weekly by oral gavage) significantly inhibited PDX growth, while being non-toxic to mice. Immunohistochemistry confirmed the down-regulation of GPC3, c-Myc, and Ki-67, supporting its anti-proliferative effect. CONCLUSION CYT997 is a potentially efficacious and non-toxic drug candidate for HCC therapy. Its ability to down-regulate GPC3, β-catenin, and c-Myc highlights a novel mechanism of action.
Collapse
Affiliation(s)
- Faiz Ahmad
- Asian Liver Center, Department of Surgery, School of Medicine, Stanford, California, USA
| | - Li Ma
- Asian Liver Center, Department of Surgery, School of Medicine, Stanford, California, USA
| | - Wei Wei
- Asian Liver Center, Department of Surgery, School of Medicine, Stanford, California, USA
| | - Yi Liu
- Asian Liver Center, Department of Surgery, School of Medicine, Stanford, California, USA
| | - Isaac Hakim
- Aria Pharmaceuticals, Palo Alto, California, USA
| | | | - Sana Mujahid
- Aria Pharmaceuticals, Palo Alto, California, USA
| | | | - Mei-Sze Chua
- Asian Liver Center, Department of Surgery, School of Medicine, Stanford, California, USA
| | - Samuel So
- Asian Liver Center, Department of Surgery, School of Medicine, Stanford, California, USA
| |
Collapse
|
4
|
Lin XW, Fan ZZ, Liu YH, Li J, Ma Q, Yan RH. High MEK/ERK signalling is a key regulator of diapause maintenance in the cotton bollworm, Helicoverpa armigera. INSECT MOLECULAR BIOLOGY 2021; 30:508-518. [PMID: 34086372 DOI: 10.1111/imb.12721] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 06/12/2023]
Abstract
MEK/ERK signalling has been identified as a key factor that terminates diapause in Sarcophaga crassipalpis and Bombyx mori. Paradoxically, high p-MEK/p-ERK signalling induces diapause in pupae of the moth Helicoverpa armigera; however, the regulatory mechanism is unknown. In the present study, we show that p-MEK and p-ERK are elevated in the brain of diapause-destined pupae and suppression of MEK/ERK activity terminates diapause progress. Reactive oxygen species (ROS) activate MEK/ERK signalling, causing large-scale phosphorylation of downstream proteins. The levels of ubiquitin-conjugated proteins are also significantly reduced when ROS or p-ERK level decreased. Moreover, terminated diapause progress by 20-hydroxyecdysone injection significantly decreases p-MEK, p-ERK and phospho-ribosomal S6 kinase levels, while phospho-MAPK substrates and ubiquitin-conjugated protein levels increase. Our data demonstrate that high MEK/ERK signalling mediated by ROS promotes diapause maintenance via increasing phosphorylation and degradation of downstream substrates. The results of this study may provide important information for understanding the regulatory mechanisms during insect diapause.
Collapse
Affiliation(s)
- X W Lin
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests (Hainan University), Ministry of Education, Haikou, China
- College of Plant Protection, Hainan University, Haikou, China
| | - Z Z Fan
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests (Hainan University), Ministry of Education, Haikou, China
- College of Plant Protection, Hainan University, Haikou, China
| | - Y H Liu
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests (Hainan University), Ministry of Education, Haikou, China
- College of Plant Protection, Hainan University, Haikou, China
| | - J Li
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests (Hainan University), Ministry of Education, Haikou, China
- College of Plant Protection, Hainan University, Haikou, China
| | - Q Ma
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests (Hainan University), Ministry of Education, Haikou, China
- College of Plant Protection, Hainan University, Haikou, China
| | - R H Yan
- Key Laboratory of Green Prevention and Control of Tropical Plant Diseases and Pests (Hainan University), Ministry of Education, Haikou, China
- College of Plant Protection, Hainan University, Haikou, China
| |
Collapse
|
5
|
Santarelli R, Pompili C, Gilardini Montani MS, Romeo MA, Gonnella R, D'Orazi G, Cirone M. Lovastatin reduces PEL cell survival by phosphorylating ERK1/2 that blocks the autophagic flux and engages a cross-talk with p53 to activate p21. IUBMB Life 2021; 73:968-977. [PMID: 33987937 PMCID: PMC8361952 DOI: 10.1002/iub.2503] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 03/10/2021] [Accepted: 05/11/2021] [Indexed: 12/19/2022]
Abstract
Statins are inhibitors of the mevalonate pathway that besides being cholesterol lowering agents, display anti‐cancer properties. This is because cholesterol is an essential component of cell membranes but also because the mevalonate pathway controls protein farnesylation and geranylation, processes essential for the activity of GTPase family proteins. In this study, we found that Lovastatin exerted a dose‐ and time‐dependent cytotoxic effect against PEL cells, an aggressive B cell lymphoma strictly associated with the gammaherpesvirus KSHV and characterized by a poor response to conventional chemotherapies. At molecular level, Lovastatin by dephosphorylating STAT3, induced ERK1/2 activation that inhibited autophagy and phosphorylated p53ser15 that in turn maintained ERK1/2 activated and up‐regulated p21. However, p21 played a pro‐survival role in this setting, as its inhibition by UC2288 further reduced cell survival in PEL cells undergoing Lovastatin treatment. In conclusion, this study suggests that Lovastatin may represent a valid therapeutic alternative against PEL cells, especially if used in combination with p21 inhibitors.
Collapse
Affiliation(s)
- Roberta Santarelli
- Department of Experimental Medicine, "Sapienza" University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Chiara Pompili
- Department of Experimental Medicine, "Sapienza" University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Maria Saveria Gilardini Montani
- Department of Experimental Medicine, "Sapienza" University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Maria Anele Romeo
- Department of Experimental Medicine, "Sapienza" University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Roberta Gonnella
- Department of Experimental Medicine, "Sapienza" University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Gabriella D'Orazi
- Translational Research Area, Regina Elena National Cancer Institute, Rome, Italy.,Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Mara Cirone
- Department of Experimental Medicine, "Sapienza" University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
6
|
Alterations in Tau Protein Level and Phosphorylation State in the Brain of the Autistic-Like Rats Induced by Prenatal Exposure to Valproic Acid. Int J Mol Sci 2021; 22:ijms22063209. [PMID: 33809910 PMCID: PMC8004207 DOI: 10.3390/ijms22063209] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/23/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by deficient social interaction and communication besides repetitive, stereotyped behaviours. A characteristic feature of ASD is altered dendritic spine density and morphology associated with synaptic plasticity disturbances. Since microtubules (MTs) regulate dendritic spine morphology and play an important role in spine development and plasticity the aim of the present study was to investigate the alterations in the content of neuronal α/β-tubulin and Tau protein level as well as phosphorylation state in the valproic acid (VPA)-induced rat model of autism. Our results indicated that maternal exposure to VPA induces: (1) decrease the level of α/β-tubulin along with Tau accumulation in the hippocampus and cerebral cortex; (2) excessive Tau phosphorylation and activation of Tau-kinases: CDK5, ERK1/2, and p70S6K in the cerebral cortex; (3) up-regulation of mTOR kinase-dependent signalling in the hippocampus and cerebral cortex of adolescent rat offspring. Moreover, immunohistochemical staining showed histopathological changes in neurons (chromatolysis) in both analysed brain structures of rats prenatally exposed to VPA. The observed changes in Tau protein together with an excessive decrease in α/β-tubulin level may suggest destabilization and thus dysfunction of the MT cytoskeleton network, which in consequence may lead to the disturbance in synaptic plasticity and the development of autistic-like behaviours.
Collapse
|
7
|
Rose S, Ezan F, Cuvellier M, Bruyère A, Legagneux V, Langouët S, Baffet G. Generation of proliferating human adult hepatocytes using optimized 3D culture conditions. Sci Rep 2021; 11:515. [PMID: 33436872 PMCID: PMC7804446 DOI: 10.1038/s41598-020-80019-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 11/10/2020] [Indexed: 02/08/2023] Open
Abstract
Generating the proliferation of differentiated normal adult human hepatocytes is a major challenge and an expected central step in understanding the microenvironmental conditions that regulate the phenotype of human hepatocytes in vitro. In this work, we described optimized 3D culture conditions of primary human hepatocytes (PHH) to trigger two waves of proliferation and we identified matrix stiffness and cell-cell interactions as the main actors necessary for this proliferation. We demonstrated that DNA replication and overexpression of cell cycle markers are modulate by the matrix stiffness while PHH cultured in 3D without prior cellular interactions did not proliferate. Besides, we showed that PHH carry out an additional cell cycle after transient inhibition of MAPK MER1/2-ERK1/2 signaling pathway. Collagen cultured hepatocytes are organized as characteristic hollow spheroids able to maintain survival, cell polarity and hepatic differentiation for long-term culture periods of at least 28 days. Remarkably, we demonstrated by transcriptomic analysis and functional experiments that proliferating cells are mature hepatocytes with high detoxication capacities. In conclusion, the advanced 3D model described here, named Hepoid, is particularly relevant for obtaining normal human proliferating hepatocytes. By allowing concomitant proliferation and differentiation, it constitutes a promising tool for many pharmacological and biotechnological applications.
Collapse
Affiliation(s)
- Sophie Rose
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France
| | - Frédéric Ezan
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France
| | - Marie Cuvellier
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France
| | - Arnaud Bruyère
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France
| | - Vincent Legagneux
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France
| | - Sophie Langouët
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France.
| | - Georges Baffet
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France.
| |
Collapse
|
8
|
Wu PK, Becker A, Park JI. Growth Inhibitory Signaling of the Raf/MEK/ERK Pathway. Int J Mol Sci 2020; 21:ijms21155436. [PMID: 32751750 PMCID: PMC7432891 DOI: 10.3390/ijms21155436] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 07/28/2020] [Indexed: 12/14/2022] Open
Abstract
In response to extracellular stimuli, the Raf/MEK/extracellular signal-regulated kinase (ERK) pathway regulates diverse cellular processes. While mainly known as a mitogenic signaling pathway, the Raf/MEK/ERK pathway can mediate not only cell proliferation and survival but also cell cycle arrest and death in different cell types. Growing evidence suggests that the cell fate toward these paradoxical physiological outputs may be determined not only at downstream effector levels but also at the pathway level, which involves the magnitude of pathway activity, spatial-temporal regulation, and non-canonical functions of the molecular switches in this pathway. This review discusses recent updates on the molecular mechanisms underlying the pathway-mediated growth inhibitory signaling, with a major focus on the regulation mediated at the pathway level.
Collapse
Affiliation(s)
- Pui-Kei Wu
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- Correspondence: (P.-K.W.); (J.-I.P.)
| | - Andrew Becker
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - Jong-In Park
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Correspondence: (P.-K.W.); (J.-I.P.)
| |
Collapse
|
9
|
PI3K pathway protein analyses in metastatic breast cancer patients receiving standard everolimus and exemestane. J Cancer Res Clin Oncol 2020; 146:3013-3023. [PMID: 32566979 PMCID: PMC7519923 DOI: 10.1007/s00432-020-03291-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 06/12/2020] [Indexed: 11/26/2022]
Abstract
Purpose Everolimus plus exemestane (EVE/EXE) is a registered treatment option for ER-positive, HER2-negative (ER +/HER2-) metastatic breast cancer (MBC), but resistance mechanisms limit efficacy. We aimed to find markers that might help select patients with a higher chance on benefit from EVE/EXE. Methods Immunohistochemistry (IHC) of PTEN, p-AKT(Thr308), p-AKT(Ser473), p-4EBP1, p-p70S6K, p-S6RP(Ser240/244), p-ERK1/2 and p-S6RP (Ser235/236) was performed on primary tumour tissue and on biopsies immediately taken from ER +/HER2- MBC patients before the start of standard EVE/EXE (Eudract 2013-004120-11). Unsupervised hierarchical clustering was executed to create heatmaps to distinguish subgroups of preferentially activated and less-activated PI3K/MAPK proteins. Uni- and multivariate Cox models were used for associations with PFS. Results Primary tumour tissue from 145 patients was retrieved. Median PFS was 5.4 months. Patients without (neo)adjuvant therapy (p = 0.03) or bone only disease (p = 0.04) had longer PFS on EVE/EXE. In primary tumours, neither single proteins nor PI3K/MAPK-associated heatmap subgroups were significantly associated with PFS. In 21 patients a non-osseous biopsy obtained before dosing was useful for continuous scoring, which demonstrated upregulation of several proteins as compared to readings in corresponding primary tumour tissues. These comparisons revealed that increased expression of p-4EBP1 was significantly associated with worse PFS (multivariate HR 3.69, p = 0.05). Conclusions IHC of single proteins or heatmap subgroups of the differentially activated PI3K/MAPK pathways was not able to discriminate patients on EVE/EXE with poor or better PFS. Upregulation of p-4EBP1 in pre-treatment biopsies as compared to levels in primary tumours pointed towards shorter PFS. Electronic supplementary material The online version of this article (10.1007/s00432-020-03291-x) contains supplementary material, which is available to authorized users.
Collapse
|
10
|
Hastings JF, Gonzalez Rajal A, Latham SL, Han JZ, McCloy RA, O'Donnell YE, Phimmachanh M, Murphy AD, Nagrial A, Daneshvar D, Chin V, Watkins DN, Burgess A, Croucher DR. Analysis of pulsed cisplatin signalling dynamics identifies effectors of resistance in lung adenocarcinoma. eLife 2020; 9:53367. [PMID: 32513387 PMCID: PMC7282820 DOI: 10.7554/elife.53367] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 05/05/2020] [Indexed: 12/12/2022] Open
Abstract
The identification of clinically viable strategies for overcoming resistance to platinum chemotherapy in lung adenocarcinoma has previously been hampered by inappropriately tailored in vitro assays of drug response. Therefore, using a pulse model that closely mimics the in vivo pharmacokinetics of platinum therapy, we profiled cisplatin-induced signalling, DNA-damage and apoptotic responses across a panel of human lung adenocarcinoma cell lines. By coupling this data to real-time, single-cell imaging of cell cycle and apoptosis we provide a fine-grained stratification of response, where a P70S6K-mediated signalling axis promotes resistance on a TP53 wildtype or null background, but not a mutant TP53 background. This finding highlights the value of in vitro models that match the physiological pharmacokinetics of drug exposure. Furthermore, it also demonstrates the importance of a mechanistic understanding of the interplay between somatic mutations and the signalling networks that govern drug response for the implementation of any consistently effective, patient-specific therapy. Lung adenocarcinoma is the most common type of lung cancer, and it emerges because of a variety of harmful genetic changes, or mutations. Two lung cancer patients – or indeed, two different sets of cancerous cells within a patient – may therefore carry different damaging mutations. A group of drugs called platinum-based chemotherapies are currently the most effective way to treat lung adenocarcinoma. Yet, only 30% of patients actually respond to the therapy. Many studies conducted in laboratory settings have tried to understand why most cases are resistant to treatment, with limited success. Here, Hastings, Gonzalez-Rajal et al. propose that previous research has been inconclusive because studies done in the laboratory do not reflect how the treatment is actually administered. In patients, platinum-based drugs are cleared from the body within a few hours, but during experiments, the treatment is continually administered to cells growing in a dish. Hastings, Gonzalez-Rajal et al. therefore developed a laboratory method that mimics the way cells are exposed to platinum-based chemotherapy in the body. These experiments showed that the lung adenocarcinoma cells which resisted treatment also carried high levels of a protein known as P70S6K. Pairing platinum-based chemotherapy with a drug that blocks the activity of P70S6K killed these resistant cells. This combination also treated human lung adenocarcinoma tumours growing under the skin of mice. However, it was ineffective on cancerous cells that carry a mutation in a protein called p53, which is often defective in cancers. Overall, this work demonstrates the need to refine how drugs are tested in the laboratory to better reflect real-life conditions. It also underlines the importance of personalizing drug combinations to the genetic background of each tumour, a concept that will be vital to consider in future clinical trials.
Collapse
Affiliation(s)
- Jordan F Hastings
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia
| | | | - Sharissa L Latham
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia.,St Vincent's Hospital Clinical School, University of New South Wales, Sydney, Australia
| | - Jeremy Zr Han
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia
| | - Rachael A McCloy
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia
| | - Yolande Ei O'Donnell
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia
| | - Monica Phimmachanh
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia
| | - Alexander D Murphy
- Crown Princess Mary Cancer Centre, Westmead and Blacktown Hospitals, Sydney, Australia
| | - Adnan Nagrial
- Crown Princess Mary Cancer Centre, Westmead and Blacktown Hospitals, Sydney, Australia
| | - Dariush Daneshvar
- Crown Princess Mary Cancer Centre, Westmead and Blacktown Hospitals, Sydney, Australia
| | - Venessa Chin
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia.,St Vincent's Hospital Clinical School, University of New South Wales, Sydney, Australia.,St Vincent's Hospital Sydney, Darlinghurst, Australia
| | - D Neil Watkins
- Hudson Institute of Medical Research, Victoria, Australia.,Department of Molecular and Translational Medicine, School of Medicine, Nursing and Health Sciences, Monash University, Victoria, Australia.,Research Institute in Oncology and Hematology, Cancer Care Manitoba, Winnipeg, Canada.,Department of Internal Medicine, Rady Faculty of Health Science, University of Manitoba, Winnipeg, Canada
| | - Andrew Burgess
- ANZAC Research Institute, Concord, Australia.,The University of Sydney Concord Clinical School, Faculty of Medicine and Health, Sydney, Australia
| | - David R Croucher
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, Australia.,St Vincent's Hospital Clinical School, University of New South Wales, Sydney, Australia.,School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
11
|
Naito K, Kurihara K, Moteki H, Kimura M, Natsume H, Ogihara M. Effect of Selective Serotonin (5-HT) 2B Receptor Agonist BW723C86 on Epidermal Growth Factor/Transforming Growth Factor-α Receptor Tyrosine Kinase and Ribosomal p70 S6 Kinase Activities in Primary Cultures of Adult Rat Hepatocytes. Biol Pharm Bull 2019; 42:631-637. [PMID: 30713268 DOI: 10.1248/bpb.b18-00831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Serotonin (5-hydroxytryptamine; 5-HT) can induce hepatocyte DNA synthesis and proliferation by autocrine secretion of transforming growth factor (TGF)-α through 5-HT2B receptor/phospholipase C (PLC)/Ca2+ and a signaling pathway involving epidermal growth factor (EGF)/TGF-α receptor tyrosine kinase (RTK)/extracellular signal-regulated kinase 2 (ERK2)/mammalian target of rapamycin (mTOR). In the present study, we investigated whether 5-HT or a selective 5-HT2B receptor agonist BW723C86, would stimulate phosphorylation of TGF-α RTK and ribosomal p70 S6 kinase (p70S6K) in primary cultures of adult rat hepatocytes. Western blotting analysis was used to detect 5-HT- or BW723C86 (10-6 M)-induced phosphorylation of EGF/TGF-α RTK and p70S6K. Our results showed that 5-HT- or BW723C86 (10-6 M)-induced phosphorylation of EGF/TGF-α RTK peaked at between 5 and 10 min. On the other hand, 5-HT- or BW723C86 (10-6 M)-induced phosphorylation of p70S6K peaked at about 30 min. Furthermore, a selective 5-HT2B receptor antagonist LY272015, a specific PLC inhibitor U-73122, a membrane-permeable Ca2+ chelator BAPTA/AM, an L-type Ca2+ channel blocker verapamil, somatostatin, and a specific p70S6K inhibitor LY2584702 completely abolished the phosphorylation of p70S6K induced by both 5-HT and BW723C86. These results indicate that phosphorylation of p70S6K is dependent on the 5-HT2B-receptor-mediated autocrine secretion of TGF-α. In addition, these results demonstrate that the hepatocyte proliferating action of 5-HT and BW723C86 are mediated by phosphorylation of p70S6K, a downstream element of the EGF/TGF-α RTK signaling pathway.
Collapse
Affiliation(s)
- Kota Naito
- Departments of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Josai University
| | - Kazuki Kurihara
- Departments of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Josai University
| | - Hajime Moteki
- Departments of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Josai University
| | - Mitsutoshi Kimura
- Departments of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Josai University
| | - Hideshi Natsume
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Josai University
| | - Masahiko Ogihara
- Departments of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Josai University
| |
Collapse
|
12
|
Metformin Promotes Neuronal Differentiation via Crosstalk between Cdk5 and Sox6 in Neuroblastoma Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:1765182. [PMID: 30911317 PMCID: PMC6399528 DOI: 10.1155/2019/1765182] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/21/2019] [Accepted: 02/07/2019] [Indexed: 12/19/2022]
Abstract
Metformin has recently emerged as a key player in promotion of neuroblastoma differentiation and neurite outgrowth. However, molecular mechanisms of how metformin promotes cellular differentiation have not yet been fully elucidated. In this study, we investigated how metformin promotes cell differentiation, via an inhibition of cell proliferation, by culturing SH-SY5Y neuroblastoma cells with or without metformin. Pretreatment with reactive oxygen species (ROS) scavenger, NAC, revealed that ROS plays a crucial role in induction of cell differentiation. Cell differentiation was observed under various morphological criteria: extension of neuritic processes and neuronal differentiation markers. Treatment with metformin significantly increased neurite length, number of cells with neurite, and expression of neuronal differentiation markers, β-tubulin III and tyrosine hydroxylase (TH) compared with untreated control. Further investigation found that metformin significantly decreased Cdk5 but increased Sox6 during cell differentiation. Analysis of the mechanism underlying these changes using Cdk5 inhibitor, roscovitine, indicated that expressions of Cdk5 and Sox6 corresponded to metformin treatment. These results suggested that metformin produces neuronal differentiation via Cdk5 and Sox6. In addition, phosphorylated Erk1/2 was decreased while phosphorylated Akt was increased in metformin treatment. Taken together, these findings suggest that metformin promotes neuronal differentiation via ROS activation through Cdk5/Sox6 crosstalk, relating to Erk1/2 and Akt signaling.
Collapse
|
13
|
Yuan X, Wang Y, Li L, Zhou W, Tian D, Lu C, Yu S, Zhao J, Peng S. PM 2.5 induces embryonic growth retardation: Potential involvement of ROS-MAPKs-apoptosis and G0/G1 arrest pathways. ENVIRONMENTAL TOXICOLOGY 2016; 31:2028-2044. [PMID: 26472167 DOI: 10.1002/tox.22203] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 09/25/2015] [Accepted: 09/26/2015] [Indexed: 06/05/2023]
Abstract
Airborne fine particulate matter (PM2.5 ) is an "invisible killer" to human health. There is increasing evidence revealing the adverse effects of PM2.5 on the early embryonic development and pregnancy outcome, but the molecular mechanism underlying PM2.5 -induced embryotoxicity is largely unknown. Previous studies have documented that exposure to PM triggers ROS generation, leads to subsequent activation of MAPKs signaling, and results in corresponding cell biological changes including enhanced apoptosis and altered cell cycle in the cardiopulmonary system. Here, we investigated whether ROS-MAPKs-apoptosis/cell cycle arrest pathways play an important role in PM2.5 -induced embryotoxicity using the rat whole embryo culture system. The results showed that PM2.5 treatment led to embryonic growth retardation at concentrations of 50 μg/ml and above, as evidenced by the reduced yolk sac diameter, crown-rump length, head length and somite number. PM2.5 -induced embryonic growth retardation was accompanied by cell apoptosis and G0/G1 phase arrest. Furthermore, ROS generation and subsequent activation of JNK and ERK might be involved in PM2.5 -induced apoptosis and G0/G1 phase arrest by downregulating Bcl-2/Bax protein ratio and upregulating p15INK4B , p16INK4A , and p21WAF1/CIP1 transcription level. In conclusion, our results indicate that ROS-JNK/ERK-apoptosis and G0/G1 arrest pathways are involved in PM2.5 -induced embryotoxicity, which not only provides insights into the molecular mechanism of PM2.5 -induced embryotoxicity, but also may help to identify specific interventions to improve adverse pregnancy outcomes of PM2.5 . © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 2028-2044, 2016.
Collapse
Affiliation(s)
- Xiaoyan Yuan
- Evaluation and Research Center for Toxicology, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, 20 Dongdajie Street, Fengtai District, Beijing, 100071, People's Republic of China
| | - Yimei Wang
- Evaluation and Research Center for Toxicology, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, 20 Dongdajie Street, Fengtai District, Beijing, 100071, People's Republic of China
| | - Lizhong Li
- Evaluation and Research Center for Toxicology, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, 20 Dongdajie Street, Fengtai District, Beijing, 100071, People's Republic of China
| | - Wei Zhou
- Evaluation and Research Center for Toxicology, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, 20 Dongdajie Street, Fengtai District, Beijing, 100071, People's Republic of China
| | - Dongdong Tian
- Evaluation and Research Center for Toxicology, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, 20 Dongdajie Street, Fengtai District, Beijing, 100071, People's Republic of China
| | - Chunfeng Lu
- Evaluation and Research Center for Toxicology, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, 20 Dongdajie Street, Fengtai District, Beijing, 100071, People's Republic of China
| | - Shouzhong Yu
- Evaluation and Research Center for Toxicology, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, 20 Dongdajie Street, Fengtai District, Beijing, 100071, People's Republic of China
| | - Jun Zhao
- Evaluation and Research Center for Toxicology, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, 20 Dongdajie Street, Fengtai District, Beijing, 100071, People's Republic of China
| | - Shuangqing Peng
- Evaluation and Research Center for Toxicology, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, 20 Dongdajie Street, Fengtai District, Beijing, 100071, People's Republic of China
| |
Collapse
|
14
|
Translational control of myelin basic protein expression by ERK2 MAP kinase regulates timely remyelination in the adult brain. J Neurosci 2015; 35:7850-65. [PMID: 25995471 DOI: 10.1523/jneurosci.4380-14.2015] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Successful myelin repair in the adult CNS requires the robust and timely production of myelin proteins to generate new myelin sheaths. The underlying regulatory mechanisms and complex molecular basis of myelin regeneration, however, remain poorly understood. Here, we investigate the role of ERK MAP kinase signaling in this process. Conditional deletion of Erk2 from cells of the oligodendrocyte lineage resulted in delayed remyelination following demyelinating injury to the adult mouse corpus callosum. The delayed repair occurred as a result of a specific deficit in the translation of the major myelin protein, MBP. In the absence of ERK2, activation of the ribosomal protein S6 kinase (p70S6K) and its downstream target, ribosomal protein S6 (S6RP), was impaired at a critical time when premyelinating oligodendrocytes were transitioning to mature cells capable of generating new myelin sheaths. Thus, we have described an important link between the ERK MAP kinase signaling cascade and the translational machinery specifically in remyelinating oligodendrocytes in vivo. These results suggest an important role for ERK2 in the translational control of MBP, a myelin protein that appears critical for ensuring the timely generation of new myelin sheaths following demyelinating injury in the adult CNS.
Collapse
|
15
|
ERK1/2 can feedback-regulate cellular MEK1/2 levels. Cell Signal 2015; 27:1939-48. [PMID: 26163823 DOI: 10.1016/j.cellsig.2015.07.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 06/30/2015] [Accepted: 07/07/2015] [Indexed: 12/11/2022]
Abstract
Signal transduction of the Raf/MEK/ERK pathway is regulated by various feedback mechanisms. Given the greater molar ratio between Raf-MEK than between MEK-ERK in cells, it may be possible that MEK1/2 levels are regulated to modulate Raf/MEK/ERK activity upon pathway stimulation. Nevertheless, it has not been reported whether MEK1/2 expression can be subject to a feedback regulation. Here, we report that the Raf/MEK/ERK pathway can feedback-regulate cellular MEK1 and MEK2 levels. In different cell types, ΔRaf-1:ER- or B-Raf(V600E)-mediated MEK/ERK activation increased MEK1 but decreased MEK2 levels. These regulations were abrogated by ERK1/2 knockdown mediated by RNA interference, suggesting the presence of a feedback mechanism that regulates MEK1/2 levels. Subsequently, analyses using qPCR and luciferase reporters of the DNA promoter and 3' untranslated region revealed that the feedback MEK1 upregulation was in part attributed to increased transcription. However, the feedback MEK2 downregulation was only observed at protein levels, which was blocked by the proteasome inhibitors, MG132 and bortezomib, suggesting that the MEK2 regulation is mediated at a post-translational level. These results suggest that the Raf/MEK/ERK pathway can feedback-regulate cellular levels of MEK1 and MEK2, wherein MEK1 levels are upregulated at transcriptional level whereas MEK2 levels are downregulated at posttranslational level.
Collapse
|
16
|
Wu PK, Hong SK, Yoon SH, Park JI. Active ERK2 is sufficient to mediate growth arrest and differentiation signaling. FEBS J 2015; 282:1017-30. [PMID: 25639353 DOI: 10.1111/febs.13197] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 01/07/2015] [Accepted: 01/13/2015] [Indexed: 12/01/2022]
Abstract
Although extracellular signal-regulated kinases (ERK1/2) have been shown to be required in Raf/MEK/ERK pathway signaling, its sufficiency for mediating the pathway signaling has not been firmly established. In an effort to address this, we evaluated previously described ERK2 mutants that exhibit enhanced autophosphorylation of TEY sites in the activation loop in terms of their ability to induce growth arrest and differentiation in LNCaP and PC12 cells. We demonstrate that expression of ERK2-L73P/S151D, containing Lys73Pro and Ser151Asp substitutions that synergistically promote ERK autophosphorylation, is sufficient to induce growth arrest and differentiation, whereas expression of ERK2-I84A and ERK2-R65S/D319N is not as effective. When compared to the constitutively active MEK1-ΔN3/S218E/S222D, expression of ERK2-L73P/S151D only mildly increased ERK kinase activity in cells, as assessed using the ERK substrates p90(RSK) and ETS domain-containing protein (ELK1). However, ERK2-L73P/S151D expression effectively induced down-regulation of androgen receptors, Retinoblastoma (Rb) protein and E2F1 transcription factor, and up-regulation of p16(INK4A) and p21(CIP1), accompanied by cell-cycle arrest and morphological differentiation in LNCaP cells and neurite-like processes in PC12 cells. These effects and the TEY site phosphorylation of ERK2-L73P/S151D were abrogated upon introduction of the active site-disabling Lys52Arg mutation, suggesting that its autoactivation drives this signaling. Moreover, introduction of mutations Asp316/319Ala or Asp319Asn, which impair the common docking site/D-domain-based physical interaction of ERK, did not significantly affect ERK2-L73P/S151D signaling, suggesting that ERK2 mediates growth arrest and differentiation independently of the conventional ERK-target interaction mechanism. Thus, our study presents convincing evidence of ERK sufficiency for Raf/MEK/ERK signaling.
Collapse
Affiliation(s)
- Pui-Kei Wu
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | | | | |
Collapse
|
17
|
Sheppard CL, Lee LCY, Hill EV, Henderson DJP, Anthony DF, Houslay DM, Yalla KC, Cairns LS, Dunlop AJ, Baillie GS, Huston E, Houslay MD. Mitotic activation of the DISC1-inducible cyclic AMP phosphodiesterase-4D9 (PDE4D9), through multi-site phosphorylation, influences cell cycle progression. Cell Signal 2014; 26:1958-74. [PMID: 24815749 DOI: 10.1016/j.cellsig.2014.04.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 04/28/2014] [Accepted: 04/29/2014] [Indexed: 10/25/2022]
Abstract
In Rat-1 cells, the dramatic decrease in the levels of both intracellular cyclic 3'5' adenosine monophosphate (cyclic AMP; cAMP) and in the activity of cAMP-activated protein kinase A (PKA) observed in mitosis was paralleled by a profound increase in cAMP hydrolyzing phosphodiesterase-4 (PDE4) activity. The decrease in PKA activity, which occurs during mitosis, was attributable to PDE4 activation as the PDE4 selective inhibitor, rolipram, but not the phosphodiesterase-3 (PDE3) inhibitor, cilostamide, specifically ablated this cell cycle-dependent effect. PDE4 inhibition caused Rat-1 cells to move from S phase into G2/M more rapidly, to transit through G2/M more quickly and to remain in G1 for a longer period. Inhibition of PDE3 elicited no observable effects on cell cycle dynamics. Selective immunopurification of each of the four PDE4 sub-families identified PDE4D as being selectively activated in mitosis. Subsequent analysis uncovered PDE4D9, an isoform whose expression can be regulated by Disrupted-In-Schizophrenia 1 (DISC1)/activating transcription factor 4 (ATF4) complex, as the sole PDE4 species activated during mitosis in Rat-1 cells. PDE4D9 becomes activated in mitosis through dual phosphorylation at Ser585 and Ser245, involving the combined action of ERK and an unidentified 'switch' kinase that has previously been shown to be activated by H2O2. Additionally, in mitosis, PDE4D9 also becomes phosphorylated at Ser67 and Ser81, through the action of MK2 (MAPKAPK2) and AMP kinase (AMPK), respectively. The multisite phosphorylation of PDE4D9 by all four of these protein kinases leads to decreased mobility (band-shift) of PDE4D9 on SDS-PAGE. PDE4D9 is predominantly concentrated in the perinuclear region of Rat-1 cells but with a fraction distributed asymmetrically at the cell margins. Our investigations demonstrate that the diminished levels of cAMP and PKA activity that characterise mitosis are due to enhanced cAMP degradation by PDE4D9. PDE4D9, was found to locate primarily not only in the perinuclear region of Rat-1 cells but also at the cell margins. We propose that the sequestration of PDE4D9 in a specific complex together with AMPK, ERK, MK2 and the H2O2-activatable 'switch' kinase allows for its selective multi-site phosphorylation, activation and regulation in mitosis.
Collapse
Affiliation(s)
- Catherine L Sheppard
- Institute of Neuroscience and Psychology, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - Louisa C Y Lee
- Institute of Neuroscience and Psychology, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - Elaine V Hill
- Institute of Neuroscience and Psychology, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - David J P Henderson
- Institute of Neuroscience and Psychology, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - Diana F Anthony
- Institute of Neuroscience and Psychology, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - Daniel M Houslay
- Institute of Neuroscience and Psychology, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - Krishna C Yalla
- Institute of Neuroscience and Psychology, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - Lynne S Cairns
- Institute of Neuroscience and Psychology, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - Allan J Dunlop
- Institute of Neuroscience and Psychology, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - George S Baillie
- Institute of Cardiovascular and Medical Sciences, Wolfson Link and Davidson Buildings, University of Glasgow, University Avenue, Glasgow G12 8QQ, Scotland, UK
| | - Elaine Huston
- Institute of Pharmaceutical Science, King's College London, 5th Floor, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Miles D Houslay
- Institute of Pharmaceutical Science, King's College London, 5th Floor, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK.
| |
Collapse
|
18
|
Abstract
The Raf/MEK/extracellular signal-regulated kinase (ERK) pathway has a pivotal role in facilitating cell proliferation, and its deregulated activation is a central signature of many epithelial cancers. However paradoxically, sustained activity of Raf/MEK/ERK can also result in growth arrest in many different cell types. This anti-proliferative Raf/MEK/ERK signaling also has physiological significance, as exemplified by its potential as a tumor suppressive mechanism. Therefore, significant questions include in which cell types and by what mechanisms this pathway can mediate such an opposing context of signaling. Particularly, our understating of the role of ERK1 and ERK2, the focal points of pathway signaling, in growth arrest signaling is still limited. This review discusses these aspects of Raf/MEK/ERK-mediated growth arrest signaling.
Collapse
|