1
|
Lombardi Z, Gardini L, Kashchuk AV, Menconi A, Lulli M, Tusa I, Tubita A, Maresca L, Stecca B, Capitanio M, Rovida E. Importin subunit beta-1 mediates ERK5 nuclear translocation, and its inhibition synergizes with ERK5 kinase inhibitors in reducing cancer cell proliferation. Mol Oncol 2025; 19:99-113. [PMID: 38965815 PMCID: PMC11705758 DOI: 10.1002/1878-0261.13674] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 04/30/2024] [Accepted: 05/27/2024] [Indexed: 07/06/2024] Open
Abstract
The mitogen-activated protein kinase (MAPK) extracellular signal-regulated kinase 5 (ERK5) is emerging as a promising target in cancer. Indeed, alterations of the MEK5/ERK5 pathway are present in many types of cancer, including melanoma. One of the key events in MAPK signalling is MAPK nuclear translocation and its subsequent regulation of gene expression. Likewise, the effects of ERK5 in supporting cancer cell proliferation have been linked to its nuclear localization. Despite many processes regulating ERK5 nuclear translocation having been determined, the nuclear transporters involved have not yet been identified. Here, we investigated the role of importin subunit alpha (α importin) and importin subunit beta-1 (importin β1) in ERK5 nuclear shuttling to identify additional targets for cancer treatment. Either importin β1 knockdown or the α/β1 importin inhibitor ivermectin reduced the nuclear amount of overexpressed and endogenous ERK5 in HEK293T and A375 melanoma cells, respectively. These results were confirmed in single-molecule microscopy in HeLa cells. Moreover, immunofluorescence analysis showed that ivermectin impairs epidermal growth factor (EGF)-induced ERK5 nuclear shuttling in HeLa cells. Both co-immunoprecipitation experiments and proximity ligation assay provided evidence that ERK5 and importin β1 interact and that this interaction is further induced by EGF administration and prevented by ivermectin treatment. The combination of ivermectin and the ERK5 inhibitor AX15836 synergistically reduced cell viability and colony formation ability in A375 and HeLa cells and was more effective than single treatments in preventing the growth of A375 and HeLa spheroids. The increased reduction of cell viability upon the same combination was also observed in patient-derived metastatic melanoma cells. The combination of ivermectin and ERK5 inhibitors other than AX15836 provided similar effects on cell viability. The identification of importin β1 as the nuclear transporter of ERK5 may be exploited for additional ERK5-inhibiting strategies for cancer therapy.
Collapse
Affiliation(s)
- Zoe Lombardi
- Department of Clinical and Experimental Biomedical SciencesUniversity of FlorenceItaly
| | - Lucia Gardini
- National Institute of Optics, National Research CouncilFlorenceItaly
- European Laboratory of Non‐Linear Spectroscopy (LENS)FlorenceItaly
| | - Anatolii V. Kashchuk
- European Laboratory of Non‐Linear Spectroscopy (LENS)FlorenceItaly
- Department of Physics and AstronomyUniversity of FlorenceItaly
| | - Alessio Menconi
- Department of Clinical and Experimental Biomedical SciencesUniversity of FlorenceItaly
| | - Matteo Lulli
- Department of Clinical and Experimental Biomedical SciencesUniversity of FlorenceItaly
| | - Ignazia Tusa
- Department of Clinical and Experimental Biomedical SciencesUniversity of FlorenceItaly
| | - Alessandro Tubita
- Department of Clinical and Experimental Biomedical SciencesUniversity of FlorenceItaly
| | - Luisa Maresca
- Core Research Laboratory – Institute for Cancer Research and Prevention (ISPRO)FlorenceItaly
| | - Barbara Stecca
- Core Research Laboratory – Institute for Cancer Research and Prevention (ISPRO)FlorenceItaly
| | - Marco Capitanio
- European Laboratory of Non‐Linear Spectroscopy (LENS)FlorenceItaly
- Department of Physics and AstronomyUniversity of FlorenceItaly
| | - Elisabetta Rovida
- Department of Clinical and Experimental Biomedical SciencesUniversity of FlorenceItaly
| |
Collapse
|
2
|
Aoki-Utsubo C, Kameoka M, Deng L, Hanafi M, Dewi BE, Sudarmono P, Wakita T, Hotta H. Statins enhance extracellular release of hepatitis C virus particles through ERK5 activation. Microbiol Immunol 2024; 68:359-370. [PMID: 39073705 DOI: 10.1111/1348-0421.13166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/13/2024] [Accepted: 07/09/2024] [Indexed: 07/30/2024]
Abstract
Statins, such as lovastatin, have been known to inhibit 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase. Statins were reported to moderately suppress hepatitis C virus (HCV) replication in cultured cells harboring HCV RNA replicons. We report here using an HCV cell culture (HCVcc) system that high concentrations of lovastatin (5-20 μg/mL) markedly enhanced the release of HCV infectious particles (virion) in the culture supernatants by up to 40 times, without enhancing HCV RNA replication, HCV protein synthesis, or HCV virion assembly in the cells. We also found that lovastatin increased the phosphorylation (activation) level of extracellular-signal-regulated kinase 5 (ERK5) in both the infected and uninfected cells in a dose-dependent manner. The lovastatin-mediated increase of HCV virion release was partially reversed by selective ERK5 inhibitors, BIX02189 and XMD8-92, or by ERK5 knockdown using small interfering RNA (siRNA). Moreover, we demonstrated that other cholesterol-lowering statins, but not dehydrolovastatin that is incapable of inhibiting HMG-CoA reductase and activating ERK5, enhanced HCV virion release to the same extent as observed with lovastatin. These results collectively suggest that statins markedly enhance HCV virion release from infected cells through HMG-CoA reductase inhibition and ERK5 activation.
Collapse
Affiliation(s)
- Chie Aoki-Utsubo
- Department of Public Health, Graduate School of Health Sciences, Kobe University, Kobe, Japan
| | - Masanori Kameoka
- Department of Public Health, Graduate School of Health Sciences, Kobe University, Kobe, Japan
| | - Lin Deng
- Division of Infectious Disease Control, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Muhammad Hanafi
- Research Center for Chemistry, National Research and Innovation Agency (BRIN), Serpong, Indonesia
| | - Beti Ernawati Dewi
- Department of Microbiology, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Pratiwi Sudarmono
- Department of Microbiology, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Takaji Wakita
- National Institute of Infectious Diseases, Tokyo, Japan
| | - Hak Hotta
- Department of Public Health, Graduate School of Health Sciences, Kobe University, Kobe, Japan
- Faculty of Clinical Nutrition and Dietetics, Konan Women's University, Kobe, Japan
| |
Collapse
|
3
|
Espinosa-Gil S, Ivanova S, Alari-Pahissa E, Denizli M, Villafranca-Magdalena B, Viñas-Casas M, Bolinaga-Ayala I, Gámez-García A, Faundez-Vidiella C, Colas E, Lopez-Botet M, Zorzano A, Lizcano JM. MAP kinase ERK5 modulates cancer cell sensitivity to extrinsic apoptosis induced by death-receptor agonists. Cell Death Dis 2023; 14:715. [PMID: 37919293 PMCID: PMC10622508 DOI: 10.1038/s41419-023-06229-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/13/2023] [Accepted: 10/18/2023] [Indexed: 11/04/2023]
Abstract
Death receptor ligand TRAIL is a promising cancer therapy due to its ability to selectively trigger extrinsic apoptosis in cancer cells. However, TRAIL-based therapies in humans have shown limitations, mainly due inherent or acquired resistance of tumor cells. To address this issue, current efforts are focussed on dissecting the intracellular signaling pathways involved in resistance to TRAIL, to identify strategies that sensitize cancer cells to TRAIL-induced cytotoxicity. In this work, we describe the oncogenic MEK5-ERK5 pathway as a critical regulator of cancer cell resistance to the apoptosis induced by death receptor ligands. Using 2D and 3D cell cultures and transcriptomic analyses, we show that ERK5 controls the proteostasis of TP53INP2, a protein necessary for full activation of caspase-8 in response to TNFα, FasL or TRAIL. Mechanistically, ERK5 phosphorylates and induces ubiquitylation and proteasomal degradation of TP53INP2, resulting in cancer cell resistance to TRAIL. Concordantly, ERK5 inhibition or genetic deletion, by stabilizing TP53INP2, sensitizes cancer cells to the apoptosis induced by recombinant TRAIL and TRAIL/FasL expressed by Natural Killer cells. The MEK5-ERK5 pathway regulates cancer cell proliferation and survival, and ERK5 inhibitors have shown anticancer activity in preclinical models of solid tumors. Using endometrial cancer patient-derived xenograft organoids, we propose ERK5 inhibition as an effective strategy to sensitize cancer cells to TRAIL-based therapies.
Collapse
Affiliation(s)
- Sergio Espinosa-Gil
- Departament de Bioquímica i Biologia Molecular and Institut de Neurociències. Facultat de Medicina, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Protein Kinases in Cancer Research. Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Saska Ivanova
- IRB Institute for Research in Biomedicine, Barcelona, Spain
- CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Melek Denizli
- Biomedical Research Group in Gynecology, Vall Hebron Institute of Research, Universitat Autònoma de Barcelona. CIBERONC, Barcelona, Spain
| | - Beatriz Villafranca-Magdalena
- Biomedical Research Group in Gynecology, Vall Hebron Institute of Research, Universitat Autònoma de Barcelona. CIBERONC, Barcelona, Spain
| | - Maria Viñas-Casas
- Departament de Bioquímica i Biologia Molecular and Institut de Neurociències. Facultat de Medicina, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Protein Kinases in Cancer Research. Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Idoia Bolinaga-Ayala
- Departament de Bioquímica i Biologia Molecular and Institut de Neurociències. Facultat de Medicina, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Protein Kinases in Cancer Research. Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Andrés Gámez-García
- Departament de Bioquímica i Biologia Molecular and Institut de Neurociències. Facultat de Medicina, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Claudia Faundez-Vidiella
- Departament de Bioquímica i Biologia Molecular and Institut de Neurociències. Facultat de Medicina, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Protein Kinases in Cancer Research. Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Eva Colas
- Biomedical Research Group in Gynecology, Vall Hebron Institute of Research, Universitat Autònoma de Barcelona. CIBERONC, Barcelona, Spain
| | - Miguel Lopez-Botet
- University Pompeu Fabra, Barcelona, Spain
- Immunology laboratory, Dpt. of Pathology, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Antonio Zorzano
- IRB Institute for Research in Biomedicine, Barcelona, Spain
- CIBERDEM, Instituto de Salud Carlos III, Madrid, Spain
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biología, Universitat de Barcelona, Barcelona, Spain
| | - José Miguel Lizcano
- Departament de Bioquímica i Biologia Molecular and Institut de Neurociències. Facultat de Medicina, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.
- Protein Kinases in Cancer Research. Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain.
| |
Collapse
|
4
|
de Mattos K, Dumas FO, Campolina-Silva GH, Belleannée C, Viger RS, Tremblay JJ. ERK5 Cooperates With MEF2C to Regulate Nr4a1 Transcription in MA-10 and MLTC-1 Leydig Cells. Endocrinology 2023; 164:bqad120. [PMID: 37539861 PMCID: PMC10435423 DOI: 10.1210/endocr/bqad120] [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: 12/06/2022] [Revised: 03/30/2023] [Accepted: 08/02/2023] [Indexed: 08/05/2023]
Abstract
Leydig cells produce hormones required for the development and maintenance of sex characteristics and fertility in males. MEF2 transcription factors are important regulators of Leydig cell gene expression and steroidogenesis. ERK5 is an atypical member of the MAP kinase family that modulates transcription factor activity, either by direct phosphorylation or by acting as a transcriptional coactivator. While MEF2 and ERK5 are known to cooperate transcriptionally, the presence and role of ERK5 in Leydig cells remained unknown. Our goal was to determine whether ERK5 is present in Leydig cells and whether it cooperates with MEF2 to regulate gene expression. We found that ERK5 is present in Leydig cells in testicular tissue and immortalized cell lines. ERK5 knockdown in human chorionic gonadotrophin-treated MA-10 Leydig cells reduced steroidogenesis and decreased Star and Nr4a1 expression. Luciferase assays using a synthetic reporter plasmid containing 3 MEF2 elements revealed that ERK5 enhances MEF2-dependent promoter activation. Although ERK5 did not cooperate with MEF2 on the Star promoter in Leydig cell lines, we found that ERK5 and MEF2C do cooperate on the Nr4a1 promoter, which contains 2 adjacent MEF2 elements. Mutation of each MEF2 element in a short version of the Nr4a1 promoter significantly decreased the ERK5/MEF2C cooperation, indicating that both MEF2 elements need to be intact. The ERK5/MEF2C cooperation did not require phosphorylation of MEF2C on Ser387. Taken together, our data identify ERK5 as a new regulator of MEF2 activity in Leydig cells and provide potential new insights into mechanisms that regulate Leydig cell gene expression and function.
Collapse
Affiliation(s)
- Karine de Mattos
- Reproduction, Mother and Child Health, Centre de recherche du centre hospitalier universitaire de Québec, Université Laval, Québec City, QC, G1V 4G2, Canada
| | - Félix-Olivier Dumas
- Reproduction, Mother and Child Health, Centre de recherche du centre hospitalier universitaire de Québec, Université Laval, Québec City, QC, G1V 4G2, Canada
| | - Gabriel Henrique Campolina-Silva
- Reproduction, Mother and Child Health, Centre de recherche du centre hospitalier universitaire de Québec, Université Laval, Québec City, QC, G1V 4G2, Canada
| | - Clémence Belleannée
- Reproduction, Mother and Child Health, Centre de recherche du centre hospitalier universitaire de Québec, Université Laval, Québec City, QC, G1V 4G2, Canada
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Université Laval, Québec City, QC, G1V 0A6, Canada
| | - Robert S Viger
- Reproduction, Mother and Child Health, Centre de recherche du centre hospitalier universitaire de Québec, Université Laval, Québec City, QC, G1V 4G2, Canada
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Université Laval, Québec City, QC, G1V 0A6, Canada
| | - Jacques J Tremblay
- Reproduction, Mother and Child Health, Centre de recherche du centre hospitalier universitaire de Québec, Université Laval, Québec City, QC, G1V 4G2, Canada
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Université Laval, Québec City, QC, G1V 0A6, Canada
| |
Collapse
|
5
|
Mondru AK, Aljasir MA, Alrumayh A, Nithianandarajah GN, Ahmed K, Muller J, Goldring CEP, Wilm B, Cross MJ. VEGF Stimulates Activation of ERK5 in the Absence of C-Terminal Phosphorylation Preventing Nuclear Localization and Facilitating AKT Activation in Endothelial Cells. Cells 2023; 12:967. [PMID: 36980305 PMCID: PMC10047687 DOI: 10.3390/cells12060967] [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: 02/01/2023] [Revised: 03/02/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
Extracellular-signal-regulated kinase 5 (ERK5) is critical for normal cardiovascular development. Previous studies have defined a canonical pathway for ERK5 activation, showing that ligand stimulation leads to MEK5 activation resulting in dual phosphorylation of ERK5 on Thr218/Tyr220 residues within the activation loop. ERK5 then undergoes a conformational change, facilitating phosphorylation on residues in the C-terminal domain and translocation to the nucleus where it regulates MEF2 transcriptional activity. Our previous research into the importance of ERK5 in endothelial cells highlighted its role in VEGF-mediated tubular morphogenesis and cell survival, suggesting that ERK5 played a unique role in endothelial cells. Our current data show that in contrast to EGF-stimulated HeLa cells, VEGF-mediated ERK5 activation in human dermal microvascular endothelial cells (HDMECs) does not result in C-terminal phosphorylation of ERK5 and translocation to the nucleus, but instead to a more plasma membrane/cytoplasmic localisation. Furthermore, the use of small-molecule inhibitors to MEK5 and ERK5 shows that instead of regulating MEF2 activity, VEGF-mediated ERK5 is important for regulating AKT activity. Our data define a novel pathway for ERK5 activation in endothelial cells leading to cell survival.
Collapse
Affiliation(s)
- Anil Kumar Mondru
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
| | - Mohammad A. Aljasir
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
| | - Ahmed Alrumayh
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
| | - Gopika N. Nithianandarajah
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
| | - Katie Ahmed
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
| | - Jurgen Muller
- Cardiovascular Research Group, School of Pharmacy and Medical Sciences, University of Bradford, Bradford BD7 1DP, UK
| | - Christopher E. P. Goldring
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
| | - Bettina Wilm
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK
| | - Michael J. Cross
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GE, UK
| |
Collapse
|
6
|
You I, Donovan KA, Krupnick NM, Boghossian AS, Rees MG, Ronan MM, Roth JA, Fischer ES, Wang ES, Gray NS. Acute pharmacological degradation of ERK5 does not inhibit cellular immune response or proliferation. Cell Chem Biol 2022; 29:1630-1638.e7. [PMID: 36220104 PMCID: PMC9675722 DOI: 10.1016/j.chembiol.2022.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/26/2022] [Accepted: 09/17/2022] [Indexed: 01/31/2023]
Abstract
Recent interest in the role that extracellular signal-regulated kinase 5 (ERK5) plays in various diseases, particularly cancer and inflammation, has grown. Phenotypes observed from genetic knockdown or deletion of ERK5 suggested that targeting ERK5 could have therapeutic potential in various disease settings, motivating the development ATP-competitive ERK5 inhibitors. However, these inhibitors were unable to recapitulate the effects of genetic loss of ERK5, suggesting that ERK5 may have key kinase-independent roles. To investigate potential non-catalytic functions of ERK5, we report the development of INY-06-061, a potent and selective heterobifunctional degrader of ERK5. In contrast to results reported through genetic knockdown of ERK5, INY-06-061-induced ERK5 degradation did not induce anti-proliferative effects in multiple cancer cell lines or suppress inflammatory responses in primary endothelial cells. Thus, we developed and characterized a chemical tool useful for validating phenotypes reported to be associated with genetic ERK5 ablation and for guiding future ERK5-directed drug discovery efforts.
Collapse
Affiliation(s)
- Inchul You
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Katherine A Donovan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Noah M Krupnick
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | - Matthew G Rees
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Melissa M Ronan
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jennifer A Roth
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Eric S Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Eric S Wang
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| | - Nathanael S Gray
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
7
|
OXTR High stroma fibroblasts control the invasion pattern of oral squamous cell carcinoma via ERK5 signaling. Nat Commun 2022; 13:5124. [PMID: 36045118 PMCID: PMC9433374 DOI: 10.1038/s41467-022-32787-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 08/17/2022] [Indexed: 11/20/2022] Open
Abstract
The Pattern Of Invasion (POI) of tumor cells into adjacent normal tissues clinically predicts postoperative tumor metastasis/recurrence of early oral squamous cell carcinoma (OSCC), but the mechanisms underlying the development of these subtypes remain unclear. Focusing on the highest score of POIs (Worst POI, WPOI) present within each tumor, we observe a disease progression-driven shift of WPOI towards the high-risk type 4/5, associated with a mesenchymal phenotype in advanced OSCC. WPOI 4-5-derived cancer-associated fibroblasts (CAFsWPOI4-5), characterized by high oxytocin receptor expression (OXTRHigh), contribute to local-regional metastasis. OXTRHigh CAFs induce a desmoplastic stroma and CCL26 is required for the invasive phenotype of CCR3+ tumors. Mechanistically, OXTR activates nuclear ERK5 transcription signaling via Gαq and CDC37 to maintain high levels of OXTR and CCL26. ERK5 ablation reprograms the pro-invasive phenotype of OXTRHigh CAFs. Therefore, targeting ERK5 signaling in OXTRHigh CAFs is a potential therapeutic strategy for OSCC patients with WPOI 4-5. Worst pattern of invasion (WPOI) is a parameter used to quantify tumor invasiveness of oral squamous cell carcinoma (OSCC). Here the authors show that a fibroblast subset characterized by the expression of the oxytocin receptor is enriched in highly invasive WPOI 4-5 OSCC tumors and can be targeted to reduce the desmoplastic stroma and tumor metastasis.
Collapse
|
8
|
Muñoz-Díaz E, Sáez-Vásquez J. Nuclear dynamics: Formation of bodies and trafficking in plant nuclei. FRONTIERS IN PLANT SCIENCE 2022; 13:984163. [PMID: 36082296 PMCID: PMC9445803 DOI: 10.3389/fpls.2022.984163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/04/2022] [Indexed: 06/01/2023]
Abstract
The existence of the nucleus distinguishes prokaryotes and eukaryotes. Apart from containing most of the genetic material, the nucleus possesses several nuclear bodies composed of protein and RNA molecules. The nucleus is separated from the cytoplasm by a double membrane, regulating the trafficking of molecules in- and outwards. Here, we investigate the composition and function of the different plant nuclear bodies and molecular clues involved in nuclear trafficking. The behavior of the nucleolus, Cajal bodies, dicing bodies, nuclear speckles, cyclophilin-containing bodies, photobodies and DNA damage foci is analyzed in response to different abiotic stresses. Furthermore, we research the literature to collect the different protein localization signals that rule nucleocytoplasmic trafficking. These signals include the different types of nuclear localization signals (NLSs) for nuclear import, and the nuclear export signals (NESs) for nuclear export. In contrast to these unidirectional-movement signals, the existence of nucleocytoplasmic shuttling signals (NSSs) allows bidirectional movement through the nuclear envelope. Likewise, nucleolar signals are also described, which mainly include the nucleolar localization signals (NoLSs) controlling nucleolar import. In contrast, few examples of nucleolar export signals, called nucleoplasmic localization signals (NpLSs) or nucleolar export signals (NoESs), have been reported. The existence of consensus sequences for these localization signals led to the generation of prediction tools, allowing the detection of these signals from an amino acid sequence. Additionally, the effect of high temperatures as well as different post-translational modifications in nuclear and nucleolar import and export is discussed.
Collapse
Affiliation(s)
- Eduardo Muñoz-Díaz
- Centre National de la Recherche Scientifique (CNRS), Laboratoire Génome et Développement des Plantes, UMR 5096, Perpignan, France
- Univ. Perpignan Via Domitia, Laboratoire Génome et Développement des Plantes, UMR 5096, Perpignan, France
| | - Julio Sáez-Vásquez
- Centre National de la Recherche Scientifique (CNRS), Laboratoire Génome et Développement des Plantes, UMR 5096, Perpignan, France
- Univ. Perpignan Via Domitia, Laboratoire Génome et Développement des Plantes, UMR 5096, Perpignan, France
| |
Collapse
|
9
|
Howell SJ, Lee CA, Batoki JC, Zapadka TE, Lindstrom SI, Taylor BE, Taylor PR. Retinal Inflammation, Oxidative Stress, and Vascular Impairment Is Ablated in Diabetic Mice Receiving XMD8-92 Treatment. Front Pharmacol 2021; 12:732630. [PMID: 34456740 PMCID: PMC8385489 DOI: 10.3389/fphar.2021.732630] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022] Open
Abstract
The global number of diabetics continues to rise annually. As diabetes progresses, almost all of Type I and more than half of Type II diabetics develop diabetic retinopathy. Diabetic retinopathy is a microvascular disease of the retina, and is the leading cause of blindness in the working-age population worldwide. With such a significant health impact, new drugs are required to halt the blinding threat posed by this visual disorder. The cause of diabetic retinopathy is multifactorial, and an optimal therapeutic would halt inflammation, cease photoreceptor cell dysfunction, and ablate vascular impairment. XMD8-92 is a small molecule inhibitor that blocks inflammatory activity downstream of ERK5 (extracellular signal-related kinase 5) and BRD4 (bromodomain 4). ERK5 elicits inflammation, is increased in Type II diabetics, and plays a pathologic role in diabetic nephropathy, while BRD4 induces retinal inflammation and plays a role in retinal degeneration. Further, we provide evidence that suggests both pERK5 and BRD4 expression are increased in the retinas of our STZ (streptozotocin)-induced diabetic mice. Taken together, we hypothesized that XMD8-92 would be a good therapeutic candidate for diabetic retinopathy, and tested XMD8-92 in a murine model of diabetic retinopathy. In the current study, we developed an in vivo treatment regimen by administering one 100 μL subcutaneous injection of saline containing 20 μM of XMD8-92 weekly, to STZ-induced diabetic mice. XMD8-92 treatments significantly decreased diabetes-mediated retinal inflammation, VEGF production, and oxidative stress. Further, XMD8-92 halted the degradation of ZO-1 (zonula occludens-1), which is a tight junction protein associated with vascular permeability in the retina. Finally, XMD8-92 treatment ablated diabetes-mediated vascular leakage and capillary degeneration, which are the clinical hallmarks of non-proliferative diabetic retinopathy. Taken together, this study provides strong evidence that XMD8-92 could be a potentially novel therapeutic for diabetic retinopathy.
Collapse
Affiliation(s)
- Scott J. Howell
- Department of Ophthalmology and Visual Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Louis Stokes Cleveland VA Medical Center, VA Northeast Ohio Healthcare System, Cleveland, OH, United States
| | - Chieh A. Lee
- Department of Ophthalmology and Visual Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Julia C. Batoki
- Department of Ophthalmology and Visual Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Thomas E. Zapadka
- Department of Ophthalmology and Visual Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Louis Stokes Cleveland VA Medical Center, VA Northeast Ohio Healthcare System, Cleveland, OH, United States
| | - Sarah I. Lindstrom
- Department of Ophthalmology and Visual Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Brooklyn E. Taylor
- Department of Ophthalmology and Visual Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Patricia R. Taylor
- Department of Ophthalmology and Visual Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Louis Stokes Cleveland VA Medical Center, VA Northeast Ohio Healthcare System, Cleveland, OH, United States
| |
Collapse
|
10
|
Paudel R, Fusi L, Schmidt M. The MEK5/ERK5 Pathway in Health and Disease. Int J Mol Sci 2021; 22:ijms22147594. [PMID: 34299213 PMCID: PMC8303459 DOI: 10.3390/ijms22147594] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
The MEK5/ERK5 mitogen-activated protein kinases (MAPK) cascade is a unique signaling module activated by both mitogens and stress stimuli, including cytokines, fluid shear stress, high osmolarity, and oxidative stress. Physiologically, it is mainly known as a mechanoreceptive pathway in the endothelium, where it transduces the various vasoprotective effects of laminar blood flow. However, it also maintains integrity in other tissues exposed to mechanical stress, including bone, cartilage, and muscle, where it exerts a key function as a survival and differentiation pathway. Beyond its diverse physiological roles, the MEK5/ERK5 pathway has also been implicated in various diseases, including cancer, where it has recently emerged as a major escape route, sustaining tumor cell survival and proliferation under drug stress. In addition, MEK5/ERK5 dysfunction may foster cardiovascular diseases such as atherosclerosis. Here, we highlight the importance of the MEK5/ERK5 pathway in health and disease, focusing on its role as a protective cascade in mechanical stress-exposed healthy tissues and its function as a therapy resistance pathway in cancers. We discuss the perspective of targeting this cascade for cancer treatment and weigh its chances and potential risks when considering its emerging role as a protective stress response pathway.
Collapse
|
11
|
González-Rubio G, Sellers-Moya Á, Martín H, Molina M. A walk-through MAPK structure and functionality with the 30-year-old yeast MAPK Slt2. Int Microbiol 2021; 24:531-543. [PMID: 33993419 DOI: 10.1007/s10123-021-00183-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 01/10/2023]
Abstract
Mitogen-activated protein kinases (MAPKs) are evolutionarily conserved signaling proteins involved in the regulation of most eukaryotic cellular processes. They are downstream components of essential signal transduction pathways activated by the external stimuli, in which the signal is conveyed through phosphorylation cascades. The excellent genetic and biochemical tractability of simple eukaryotes such as Saccharomyces cerevisiae has significantly contributed to gain fundamental information into the physiology of these key proteins. The budding yeast MAPK Slt2 was identified 30 years ago and was later revealed as a fundamental element of the cell wall integrity (CWI) pathway, one of the five MAPK routes of S. cerevisiae. As occurs with other MAPKs, whereas Slt2 displays the core typical structural traits of eukaryotic protein kinases, it also features conserved domains among MAPKs that allow an exquisite spatio-temporal regulation of their activity and binding to activating kinases, downregulatory phosphatases, or nuclear transcription factors. Additionally, Slt2 bears a regulatory extra C-terminal tail unique among S. cerevisiae MAPKs. Here, we review the structural and functional basis for the signaling role of Slt2 in the context of the molecular architecture of this important family of protein kinases.
Collapse
Affiliation(s)
- Gema González-Rubio
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Universidad Complutense de Madrid, Pza. Ramón y Cajal s/n, 28040, Madrid, Spain
| | - Ángela Sellers-Moya
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Universidad Complutense de Madrid, Pza. Ramón y Cajal s/n, 28040, Madrid, Spain
| | - Humberto Martín
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Universidad Complutense de Madrid, Pza. Ramón y Cajal s/n, 28040, Madrid, Spain.
| | - María Molina
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Universidad Complutense de Madrid, Pza. Ramón y Cajal s/n, 28040, Madrid, Spain.
| |
Collapse
|
12
|
Tanida T, Matsuda KI, Tanaka M. Novel metabolic system for lactic acid via LRPGC1/ERRγ signaling pathway. FASEB J 2020; 34:13239-13256. [PMID: 32851675 DOI: 10.1096/fj.202000492r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/25/2020] [Accepted: 07/10/2020] [Indexed: 12/17/2022]
Abstract
Lactic acid (LA) is a byproduct of glycolysis resulting from intense exercise or a metabolic defect in aerobic processes. LA metabolism is essential to prevent lactic acidosis, but the mechanism through which LA regulates its own metabolism is largely unknown. Here, we identified a LA-responsive protein, named LRPGC1, which has a distinct role from PGC1α, a key metabolic regulator, and report that LRPGC1 particularly mediates LA response to activate liver LA metabolism. Following LA stimulation, LRPGC1, but not PGC1α, translocates from the cytoplasm to the nucleus through deactivation of nuclear export signals, interacts with the nuclear receptor ERRγ, and upregulates TFAM, which ensures mitochondrial biogenesis. Knockout of PGC1 gene in HepG2 hepatocarcinoma cells decreased the LA consumption and TFAM expression, which were rescued by LRPGC1 expression, but not by PGC1α. These LRPGC1-induced effects were mediated by ERRγ, concomitantly with mitochondrial activation. The response element for LRPGC1/ERRγ signaling pathway was identified in TFAM promoter. Notably, the survival rate of a mouse model of lactic acidosis was reduced by the liver-targeted silencing of Lrpgc1, while it was significantly ameliorated by the pharmacological activation of ERRγ. These findings demonstrate LA-responsive transactivation via LRPGC1 that highlight an intrinsic molecular mechanism for LA homeostasis.
Collapse
Affiliation(s)
- Takashi Tanida
- Department of Anatomy and Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ken Ichi Matsuda
- Department of Anatomy and Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masaki Tanaka
- Department of Anatomy and Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
13
|
He Z, Tao D, Xiong J, Lou F, Zhang J, Chen J, Dai W, Sun J, Wang Y. Phosphorylation of 5-LOX: The Potential Set-point of Inflammation. Neurochem Res 2020; 45:2245-2257. [PMID: 32671628 DOI: 10.1007/s11064-020-03090-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 06/11/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022]
Abstract
Inflammation secondary to tissue injuries serves as a double-edged sword that determines the prognosis of tissue repair. As one of the most important enzymes controlling the inflammation process by producing leukotrienes, 5-lipoxygenase (5-LOX, also called 5-LO) has been one of the therapeutic targets in regulating inflammation for a long time. Although a large number of 5-LOX inhibitors have been explored, only a few of them can be applied clinically. Surprisingly, phosphorylation of 5-LOX reveals great significance in regulating the subcellular localization of 5-LOX, which has proven to be an important mechanism underlying the enzymatic activities of 5-LOX. There are at least three phosphorylation sites in 5-LOX jointly to determine the final inflammatory outcomes, and adjustment of phosphorylation of 5-LOX at different phosphorylation sites brings hope to provide an unrecognized means to regulate inflammation. The present review intends to shed more lights into the set-point-like mechanisms of phosphorylation of 5-LOX and its possible clinical application by summarizing the biological properties of 5-LOX, the relationship of 5-LOX with neurodegenerative diseases and brain injuries, the phosphorylation of 5-LOX at different sites, the regulatory effects and mechanisms of phosphorylated 5-LOX upon inflammation, as well as the potential anti-inflammatory application through balancing the phosphorylation-depended set-point.
Collapse
Affiliation(s)
- Zonglin He
- Department of Physiology, Basic Medical School, Jinan University, Huangpu Avenue 601, Tianhe District, Guangzhou, Guangdong Province, China.,Faculty of Medicine, International school, Jinan University, Huangpu Avenue 601, Tianhe District, Guangzhou, Guangdong Province, China
| | - Di Tao
- Department of Physiology, Basic Medical School, Jinan University, Huangpu Avenue 601, Tianhe District, Guangzhou, Guangdong Province, China.,Faculty of Medicine, International school, Jinan University, Huangpu Avenue 601, Tianhe District, Guangzhou, Guangdong Province, China
| | - Jiaming Xiong
- Department of Physiology, Basic Medical School, Jinan University, Huangpu Avenue 601, Tianhe District, Guangzhou, Guangdong Province, China
| | - Fangfang Lou
- Department of Physiology, Basic Medical School, Jinan University, Huangpu Avenue 601, Tianhe District, Guangzhou, Guangdong Province, China
| | - Jiayuan Zhang
- Department of Physiology, Basic Medical School, Jinan University, Huangpu Avenue 601, Tianhe District, Guangzhou, Guangdong Province, China
| | - Jinxia Chen
- Department of Physiology, Basic Medical School, Jinan University, Huangpu Avenue 601, Tianhe District, Guangzhou, Guangdong Province, China
| | - Weixi Dai
- Department of Physiology, Basic Medical School, Jinan University, Huangpu Avenue 601, Tianhe District, Guangzhou, Guangdong Province, China.,Faculty of Medicine, International school, Jinan University, Huangpu Avenue 601, Tianhe District, Guangzhou, Guangdong Province, China
| | - Jing Sun
- Department of Physiology, Basic Medical School, Jinan University, Huangpu Avenue 601, Tianhe District, Guangzhou, Guangdong Province, China
| | - Yuechun Wang
- Department of Physiology, Basic Medical School, Jinan University, Huangpu Avenue 601, Tianhe District, Guangzhou, Guangdong Province, China.
| |
Collapse
|
14
|
Stubbs EB. Targeting the blood-nerve barrier for the management of immune-mediated peripheral neuropathies. Exp Neurol 2020; 331:113385. [PMID: 32562668 DOI: 10.1016/j.expneurol.2020.113385] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/03/2020] [Accepted: 06/15/2020] [Indexed: 02/07/2023]
Abstract
Healthy peripheral nerves encounter, with increased frequency, numerous chemical, biological, and biomechanical forces. Over time and with increasing age, these forces collectively contribute to the pathophysiology of a spectrum of traumatic, metabolic, and/or immune-mediated peripheral nerve disorders. The blood-nerve barrier (BNB) serves as a critical first-line defense against chemical and biologic insults while biomechanical forces are continuously buffered by a dense array of longitudinally orientated epineural collagen fibers exhibiting high-tensile strength. As emphasized throughout this Experimental Neurology Special Issue, the BNB is best characterized as a functionally dynamic multicellular vascular unit comprised of not only highly specialized endoneurial endothelial cells, but also associated perineurial cells, pericytes, Schwann cells, basement membrane, and invested axons. The composition of the BNB, while anatomically distinct, is not functionally dissimilar to that of the well characterized neurovascular unit of the central nervous system. While the BNB lacks a glial limitans and an astrocytic endfoot layer, the primary function of both vascular units is to establish, maintain, and protect an optimal endoneurial (PNS) or interstitial (CNS) fluid microenvironment that is vital for proper neuronal function. Altered endoneurial homeostasis as a secondary consequence of BNB dysregulation is considered an early pathological event in the course of a variety of traumatic, immune-mediated, or metabolically acquired peripheral neuropathies. In this review, emerging experimental advancements targeting the endoneurial microvasculature for the therapeutic management of immune-mediated inflammatory peripheral neuropathies, including the AIDP variant of Guillain-Barré syndrome, are discussed.
Collapse
Affiliation(s)
- Evan B Stubbs
- Research Service (151), Department of Veterans Affairs, Edward Hines Jr. VA Hospital, Hines, IL 60141, USA; Department of Ophthalmology, Loyola University Health Science Division, Maywood, IL 60153, USA.
| |
Collapse
|
15
|
Erazo T, Espinosa-Gil S, Diéguez-Martínez N, Gómez N, Lizcano JM. SUMOylation Is Required for ERK5 Nuclear Translocation and ERK5-Mediated Cancer Cell Proliferation. Int J Mol Sci 2020; 21:ijms21062203. [PMID: 32209980 PMCID: PMC7139592 DOI: 10.3390/ijms21062203] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/16/2020] [Accepted: 03/21/2020] [Indexed: 01/09/2023] Open
Abstract
The MAP kinase ERK5 contains an N-terminal kinase domain and a unique C-terminal tail including a nuclear localization signal and a transcriptional activation domain. ERK5 is activated in response to growth factors and stresses and regulates transcription at the nucleus by either phosphorylation or interaction with transcription factors. MEK5-ERK5 pathway plays an important role regulating cancer cell proliferation and survival. Therefore, it is important to define the precise molecular mechanisms implicated in ERK5 nucleo-cytoplasmic shuttling. We previously described that the molecular chaperone Hsp90 stabilizes and anchors ERK5 at the cytosol and that ERK5 nuclear shuttling requires Hsp90 dissociation. Here, we show that MEK5 or overexpression of Cdc37—mechanisms that increase nuclear ERK5—induced ERK5 Small Ubiquitin-related Modifier (SUMO)-2 modification at residues Lys6/Lys22 in cancer cells. Furthermore, mutation of these SUMO sites abolished the ability of ERK5 to translocate to the nucleus and to promote prostatic cancer PC-3 cell proliferation. We also show that overexpression of the SUMO protease SENP2 completely abolished endogenous ERK5 nuclear localization in response to epidermal growth factor (EGF) stimulation. These results allow us to propose a more precise mechanism: in response to MEK5 activation, ERK5 SUMOylation favors the dissociation of Hsp90 from the complex, allowing ERK5 nuclear shuttling and activation of the transcription.
Collapse
|
16
|
Targeted Avenues for Cancer Treatment: The MEK5-ERK5 Signaling Pathway. Trends Mol Med 2020; 26:394-407. [PMID: 32277933 DOI: 10.1016/j.molmed.2020.01.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/20/2019] [Accepted: 01/21/2020] [Indexed: 12/13/2022]
Abstract
Twenty years have passed since extracellular signal-regulated kinase 5 (ERK5) and its upstream activator, mitogen-activated protein kinase 5 (MEK5), first emerged onto the cancer research scene. Although we have come a long way in defining the liaison between dysregulated MEK5-ERK5 signaling and the pathogenesis of epithelial and nonepithelial malignancies, selective targeting of this unique pathway remains elusive. Here, we provide an updated review of the existing evidence for a correlation between aberrant MEK5-ERK5 (phospho)proteomic/transcriptomic profiles, aggressive cancer states, and poor patient outcomes. We then focus on emerging insights from preclinical models regarding the relevance of upregulated ERK5 activity in promoting tumor growth, metastasis, therapy resistance, undifferentiated traits, and immunosuppression, highlighting the opportunities, prospects, and challenges of selectively blocking this cascade for antineoplastic treatment and chemosensitization.
Collapse
|
17
|
Pearson AJ, Fullwood P, Toro Tapia G, Prise I, Smith MP, Xu Q, Jordan A, Giurisato E, Whitmarsh AJ, Francavilla C, Tournier C. Discovery of a Gatekeeper Residue in the C-Terminal Tail of the Extracellular Signal-Regulated Protein Kinase 5 (ERK5). Int J Mol Sci 2020; 21:E929. [PMID: 32023819 PMCID: PMC7037328 DOI: 10.3390/ijms21030929] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/23/2020] [Accepted: 01/29/2020] [Indexed: 01/17/2023] Open
Abstract
The extracellular signal-regulated protein kinase 5 (ERK5) is a non-redundant mitogen-activated protein kinase (MAPK) that exhibits a unique C-terminal extension which comprises distinct structural and functional properties. Here, we sought to elucidate the significance of phosphoacceptor sites in the C-terminal transactivation domain of ERK5. We have found that Thr732 acted as a functional gatekeeper residue controlling C-terminal-mediated nuclear translocation and transcriptional enhancement. Consistently, using a non-bias quantitative mass spectrometry approach, we demonstrated that phosphorylation at Thr732 conferred selectivity for binding interactions of ERK5 with proteins related to chromatin and RNA biology, whereas a number of metabolic regulators were associated with full-length wild type ERK5. Additionally, our proteomic analysis revealed that phosphorylation of the Ser730-Glu-Thr732-Pro motif could occur independently of dual phosphorylation at Thr218-Glu-Tyr220 in the activation loop. Collectively, our results firmly establish the significance of C-terminal phosphorylation in regulating ERK5 function. The post-translational modification of ERK5 on its C-terminal tail might be of particular relevance in cancer cells where ERK5 has be found to be hyperphosphoryated.
Collapse
Affiliation(s)
- Adam J. Pearson
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (A.J.P.); (Q.X.); (E.G.)
| | - Paul Fullwood
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (P.F.); (G.T.T.); (M.P.S.); (A.J.W.); (C.F.)
| | - Gabriela Toro Tapia
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (P.F.); (G.T.T.); (M.P.S.); (A.J.W.); (C.F.)
| | - Ian Prise
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK;
| | - Michael P. Smith
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (P.F.); (G.T.T.); (M.P.S.); (A.J.W.); (C.F.)
| | - Qiuping Xu
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (A.J.P.); (Q.X.); (E.G.)
| | - Allan Jordan
- Drug Discovery Unit, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M13 9PT, UK;
| | - Emanuele Giurisato
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (A.J.P.); (Q.X.); (E.G.)
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Alan J. Whitmarsh
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (P.F.); (G.T.T.); (M.P.S.); (A.J.W.); (C.F.)
| | - Chiara Francavilla
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (P.F.); (G.T.T.); (M.P.S.); (A.J.W.); (C.F.)
| | - Cathy Tournier
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (A.J.P.); (Q.X.); (E.G.)
| |
Collapse
|
18
|
Beyond Kinase Activity: ERK5 Nucleo-Cytoplasmic Shuttling as a Novel Target for Anticancer Therapy. Int J Mol Sci 2020; 21:ijms21030938. [PMID: 32023850 PMCID: PMC7038028 DOI: 10.3390/ijms21030938] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 01/18/2023] Open
Abstract
The importance of mitogen-activated protein kinases (MAPK) in human pathology is underlined by the relevance of abnormalities of MAPK-related signaling pathways to a number of different diseases, including inflammatory disorders and cancer. One of the key events in MAPK signaling, especially with respect to pro-proliferative effects that are crucial for the onset and progression of cancer, is MAPK nuclear translocation and its role in the regulation of gene expression. The extracellular signal-regulated kinase 5 (ERK5) is the most recently discovered classical MAPK and it is emerging as a possible target for cancer treatment. The bigger size of ERK5 when compared to other MAPK enables multiple levels of regulation of its expression and activity. In particular, the phosphorylation of kinase domain and C-terminus, as well as post-translational modifications and chaperone binding, are involved in ERK5 regulation. Likewise, different mechanisms control ERK5 nucleo-cytoplasmic shuttling, underscoring the key role of ERK5 in the nuclear compartment. In this review, we will focus on the mechanisms involved in ERK5 trafficking between cytoplasm and nucleus, and discuss how these processes might be exploited to design new strategies for cancer treatment.
Collapse
|
19
|
Guo YJ, Pan WW, Liu SB, Shen ZF, Xu Y, Hu LL. ERK/MAPK signalling pathway and tumorigenesis. Exp Ther Med 2020; 19:1997-2007. [PMID: 32104259 PMCID: PMC7027163 DOI: 10.3892/etm.2020.8454] [Citation(s) in RCA: 738] [Impact Index Per Article: 147.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 12/04/2019] [Indexed: 12/12/2022] Open
Abstract
Mitogen-activated protein kinase (MAPK) cascades are key signalling pathways that regulate a wide variety of cellular processes, including proliferation, differentiation, apoptosis and stress responses. The MAPK pathway includes three main kinases, MAPK kinase kinase, MAPK kinase and MAPK, which activate and phosphorylate downstream proteins. The extracellular signal-regulated kinases ERK1 and ERK2 are evolutionarily conserved, ubiquitous serine-threonine kinases that regulate cellular signalling under both normal and pathological conditions. ERK expression is critical for development and their hyperactivation plays a major role in cancer development and progression. The Ras/Raf/MAPK (MEK)/ERK pathway is the most important signalling cascade among all MAPK signal transduction pathways, and plays a crucial role in the survival and development of tumour cells. The present review discusses recent studies on Ras and ERK pathway members. With respect to processes downstream of ERK activation, the role of ERK in tumour proliferation, invasion and metastasis is highlighted, and the role of the ERK/MAPK signalling pathway in tumour extracellular matrix degradation and tumour angiogenesis is emphasised.
Collapse
Affiliation(s)
- Yan-Jun Guo
- Department of Human Anatomy and Embryology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Wei-Wei Pan
- Department of Human Anatomy and Embryology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Sheng-Bing Liu
- Department of Human Anatomy and Embryology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Zhong-Fei Shen
- Department of Human Anatomy and Embryology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Ying Xu
- Department of Human Anatomy and Embryology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Ling-Ling Hu
- Department of Human Anatomy and Embryology, College of Medicine, Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| |
Collapse
|
20
|
Xue Z, Wang J, Yu W, Li D, Zhang Y, Wan F, Kou X. Biochanin A protects against PM 2.5-induced acute pulmonary cell injury by interacting with the target protein MEK5. Food Funct 2019; 10:7188-7203. [PMID: 31608342 DOI: 10.1039/c9fo01382b] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Epidemiological studies have shown that exposure to ambient fine particulate matter (PM2.5) is associated with an increased risk for cardiopulmonary diseases. The MEK5/ERK5 and NF-κB signaling pathways are closely related to the regulation of acute pulmonary cell injury (APCI) and may play an important role in the underlying pathophysiological mechanisms. Related studies have shown that Biochanin A (BCA) effectively interferes with APCI, but the underlying mechanism through which this occurs is not fully understood. Previously, based on proteomic and bioinformatic research, we found the indispensable role of MEK5 in mediating remission effects of BCA against PM2.5-induced lung toxicity. Therefore, using A549 adenocarcinoma human alveolar basal epithelial cells (A549 cells), we combined western blot and qRT-PCR to study the protective signaling pathways induced by BCA, indicating that MEK5/ERK5 and NF-κB are both involved in mediating APCI in response to PM2.5, and MEK5/ERK5 positively activated NF-κB and its downstream cellular regulatory factors. BCA significantly suppressed PM2.5-induced upregulation of MEK5/ERK5 expression and phosphorylation and activation of NF-κB. Furthermore, due to the specificity of the MEK5/ERK5 protein structure, the binding sites and binding patterns of BCA and MEK5 were analyzed using molecular docking correlation techniques, which showed that there are stable hydrogen bonds between BCA and the PB1 domain of MEK5 as well as its kinase domain. BCA forms a stable complex with MEK5, which has potential effects on MEKK2/3-MEK5-ERK5 ternary interactions, p62/αPKC-mediated NF-κB regulation, and inhibition of MEK5 target protein phosphorylation. Therefore, our study suggests that MEK5 is an important regulator of intracellular signaling of APCI in response to PM2.5 exposure. BCA may exert anti-APCI activity by targeting MEK5 to inhibit activation of the MEK5/ERK5/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Zhaohui Xue
- Department of Food Science, School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China.
| | - Junyu Wang
- Department of Food Science, School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China.
| | - Wancong Yu
- Tianjin Academy of Agricultural Science, 300381, Tianjin, China
| | - Dan Li
- Department of Food Science, School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China.
| | - Yixia Zhang
- Department of Food Science, School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China.
| | - Fang Wan
- Department of Food Science, School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China.
| | - Xiaohong Kou
- Department of Food Science, School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China.
| |
Collapse
|
21
|
EGFR-c-Src-Mediated HDAC3 Phosphorylation Exacerbates Invasion of Breast Cancer Cells. Cells 2019; 8:cells8080930. [PMID: 31430896 PMCID: PMC6721651 DOI: 10.3390/cells8080930] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/08/2019] [Accepted: 08/14/2019] [Indexed: 01/09/2023] Open
Abstract
Breast cancer is one of the leading causes of morbidity and mortality among women. Epidermal growth factor receptor (EGFR) and proto-oncogene tyrosine-protein kinase Src (c-Src) are critical components of the signaling pathways that are associated with breast cancer. However, the regulatory mechanism of histone deacetylase 3 (HDAC3) in these pathways remains unclear. Using the Net Phos 3.1 program for the analysis of kinase consensus motifs, we found two c-Src-mediated putative phosphorylation sites, tyrosine (Tyr, Y)-328 and Y331 on HDAC3, and generated a phospho-specific HDAC3 antibody against these sites. c-Src-mediated phosphorylation was observed in the cells expressing wild-type HDAC3 (HDAC3WT), but not in cells overexpressing phosphorylation-defective HDAC3 (HDAC3Y328/331A). Phosphorylated HDAC3 showed relatively higher deacetylase activity, and PP2, which is a c-Src inhibitor, blocked HDAC3 phosphorylation and reduced its enzymatic activity. EGF treatment resulted in HDAC3 phosphorylation in both MDA-MB-231 and EGFR-overexpressing MCF7 (MCF7-EGFR) cells, but not in MCF7 cells. Total internal reflection fluorescence analysis showed that HDAC3 was recruited to the plasma membrane following EGF stimulation. HDAC3 inhibition with either c-Src knockdown or PP2 treatment significantly ameliorated the invasiveness of breast cancer cells. Altogether, our findings reveal an EGF signaling cascade involving EGFR, c-Src, and HDAC3 in breast cancer cells.
Collapse
|
22
|
Maik-Rachline G, Hacohen-Lev-Ran A, Seger R. Nuclear ERK: Mechanism of Translocation, Substrates, and Role in Cancer. Int J Mol Sci 2019; 20:ijms20051194. [PMID: 30857244 PMCID: PMC6429060 DOI: 10.3390/ijms20051194] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/03/2019] [Accepted: 03/04/2019] [Indexed: 12/15/2022] Open
Abstract
The extracellular signal-regulated kinases 1/2 (ERK) are central signaling components that regulate stimulated cellular processes such as proliferation and differentiation. When dysregulated, these kinases participate in the induction and maintenance of various pathologies, primarily cancer. While ERK is localized in the cytoplasm of resting cells, many of its substrates are nuclear, and indeed, extracellular stimulation induces a rapid and robust nuclear translocation of ERK. Similarly to other signaling components that shuttle to the nucleus upon stimulation, ERK does not use the canonical importinα/β mechanism of nuclear translocation. Rather, it has its own unique nuclear translocation signal (NTS) that interacts with importin7 to allow stimulated shuttling via the nuclear pores. Prevention of the nuclear translocation inhibits proliferation of B-Raf- and N/K-Ras-transformed cancers. This effect is distinct from the one achieved by catalytic Raf and MEK inhibitors used clinically, as cells treated with the translocation inhibitors develop resistance much more slowly. In this review, we describe the mechanism of ERK translocation, present all its nuclear substrates, discuss its role in cancer and compare its translocation to the translocation of other signaling components. We also present proof of principle data for the use of nuclear ERK translocation as an anti-cancer target. It is likely that the prevention of nuclear ERK translocation will eventually serve as a way to combat Ras and Raf transformed cancers with less side-effects than the currently used drugs.
Collapse
Affiliation(s)
- Galia Maik-Rachline
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | - Avital Hacohen-Lev-Ran
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | - Rony Seger
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
23
|
Flores K, Yadav SS, Katz AA, Seger R. The Nuclear Translocation of Mitogen-Activated Protein Kinases: Molecular Mechanisms and Use as Novel Therapeutic Target. Neuroendocrinology 2019; 108:121-131. [PMID: 30261516 DOI: 10.1159/000494085] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 09/26/2018] [Indexed: 11/19/2022]
Abstract
The mitogen-activated protein kinase (MAPK) cascades are central signaling pathways that play a central role in the regulation of most stimulated cellular processes including proliferation, differentiation, stress response and apoptosis. Currently 4 such cascades are known, each termed by its downstream MAPK components: the extracellular signal-regulated kinase 1/2 (ERK1/2), cJun-N-terminal kinase (JNK), p38 and ERK5. One of the hallmarks of these cascades is the stimulated nuclear translocation of their MAPK components using distinct mechanisms. ERK1/2 are shuttled into the nucleus by importin7, JNK and p38 by a dimer of importin3 with either importin9 or importin7, and ERK5 by importin-α/β. Dysregulation of these cascades often results in diseases, including cancer and inflammation, as well as developmental and neurological disorders. Much effort has been invested over the years in developing inhibitors to the MAPK cascades to combat these diseases. Although some inhibitors are already in clinical use or clinical trials, their effects are hampered by development of resistance or adverse side-effects. Recently, our group developed 2 myristoylated peptides: EPE peptide, which inhibits the interaction of ERK1/2 with importin7, and PERY peptide, which prevents JNK/p38 interaction with either importin7 or importin9. These peptides block the nuclear translocation of their corresponding kinases, resulting in prevention of several cancers, while the PERY peptide also inhibits inflammation-induced diseases. These peptides provide a proof of concept for the use of the nuclear translocation of MAPKs as therapeutic targets for cancer and/or inflammation.
Collapse
Affiliation(s)
- Karen Flores
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Suresh Singh Yadav
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Arieh A Katz
- Department of Integrative Biomedical Sciences and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Rony Seger
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot,
| |
Collapse
|
24
|
Adams NR, Vasquez YM, Mo Q, Gibbons W, Kovanci E, DeMayo FJ. WNK lysine deficient protein kinase 1 regulates human endometrial stromal cell decidualization, proliferation, and migration in part through mitogen-activated protein kinase 7. Biol Reprod 2018; 97:400-412. [PMID: 29025069 DOI: 10.1093/biolre/iox108] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 08/28/2017] [Indexed: 12/21/2022] Open
Abstract
The differentiation of endometrial stromal cells into decidual cells, termed decidualization, is an integral step in the establishment of pregnancy. The mitogen-activated protein kinase homolog, WNK lysine deficient protein kinase 1 (WNK1), is activated downstream of epidermal growth factor receptor during decidualization. Primary human endometrial stromal cells (HESCs) were subjected to small interfering RNA knockdown of WNK1 followed by in vitro decidualization. This abrogated expression of the decidual marker genes, insulin like growth factor binding protein 1 (IGFBP1) and prolactin (PRL), and prevented adoption of decidual cell morphology. Analysis of the WNK1-dependent transcriptome by RNA-Seq demonstrated that WNK1 regulates the expression of 1858 genes during decidualization. Gene ontology and upstream regulator pathway analysis showed that WNK1 regulates cell migration, differentiation, and proliferation. WNK1 was required for many of the gene expression changes that drive decidualization, including the induction of the inflammatory cytokines, C-C motif chemokine ligand 8 (CCL8), interleukin 1 beta (IL1B), and interleukin 15 (IL15), and the repression of transforming growth factor-beta (TGF-beta) pathway genes, including early growth response 2 (EGR2), SMAD family member 3 (SMAD3), integrin subunit alpha 2 (ITGA2), integrin subunit alpha 4 (ITGA4), and integrin subunit beta 3 (ITGB3). In addition to abrogating decidualization, WNK1 knockdown decreased the migration and proliferation of HESCs. Furthermore, mitogen-activated protein kinase 7 (MAPK7), a known downstream target of WNK1, was activated during decidualization in a WNK1-dependent manner. Small interfering RNA knockdown of MAPK7 demonstrated that MAPK7 regulates a subset of WNK1-regulated genes and controls the migration and proliferation of HESCs. These results indicate that WNK1 and MAPK7 promote migration and proliferation during decidualization and regulate the expression of inflammatory cytokines and TGF-beta pathway genes in HESCs.
Collapse
Affiliation(s)
- Nyssa R Adams
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA.,Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Yasmin M Vasquez
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Qianxing Mo
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - William Gibbons
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas, USA
| | - Ertug Kovanci
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas, USA
| | - Francesco J DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| |
Collapse
|
25
|
Maik-Rachline G, Zehorai E, Hanoch T, Blenis J, Seger R. The nuclear translocation of the kinases p38 and JNK promotes inflammation-induced cancer. Sci Signal 2018; 11:11/525/eaao3428. [PMID: 29636389 DOI: 10.1126/scisignal.aao3428] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The stimulated nuclear translocation of signaling proteins, such as MAPKs, is a necessity for the initiation and regulation of their physiological functions. Previously, we determined that nuclear translocation of the MAPKs p38 and JNK involves binding to heterodimers comprising importin 3 and either importin 7 or importin 9. Here, we identified the importin-binding region in p38 and JNK and developed a myristoylated peptide targeting this site that we called PERY. The PERY peptide specifically blocked the interaction of p38 and JNK with the importins, restricted their nuclear translocation, and inhibited phosphorylation of their nuclear (but not cytoplasmic) substrates. Through these effects, the PERY peptide reduced the proliferation of several (but not all) cancer cell lines in culture and inhibited the growth of a human breast cancer xenograft in mice. In addition, the PERY peptide substantially inhibited inflammation in mice, as manifested in models of colitis and colitis-associated colon cancer. The PERY peptide more effectively prevented colon cancer development than did a commercial p38 inhibitor. In vivo analysis further suggested that this effect was mediated by PERY peptide-induced prevention of the nuclear translocation of p38 in macrophages. Together, these results support the use of the nuclear translocation of p38 and JNK as a novel drug target to treat various cancers and inflammation-induced diseases.
Collapse
Affiliation(s)
- Galia Maik-Rachline
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Elder Zehorai
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Tamar Hanoch
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - John Blenis
- Weill Cornell Medicine, New York, NY 10021, USA
| | - Rony Seger
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
26
|
Live-cell measurements of kinase activity in single cells using translocation reporters. Nat Protoc 2017; 13:155-169. [PMID: 29266096 DOI: 10.1038/nprot.2017.128] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Although kinases are important regulators of many cellular processes, measuring their activity in live cells remains challenging. We have developed kinase translocation reporters (KTRs), which enable multiplexed measurements of the dynamics of kinase activity at a single-cell level. These KTRs are composed of an engineered construct in which a kinase substrate is fused to a bipartite nuclear localization signal (bNLS) and nuclear export signal (NES), as well as to a fluorescent protein for microscopy-based detection of its localization. The negative charge introduced by phosphorylation of the substrate is used to directly modulate nuclear import and export, thereby regulating the reporter's distribution between the cytoplasm and nucleus. The relative cytoplasmic versus nuclear fluorescence of the KTR construct (the C/N ratio) is used as a proxy for the kinase activity in living, single cells. Multiple KTRs can be studied in the same cell by fusing them to different fluorescent proteins. Here, we present a protocol to execute and analyze live-cell microscopy experiments using KTRs. We describe strategies for development of new KTRs and procedures for lentiviral expression of KTRs in a cell line of choice. Cells are then plated in a 96-well plate, from which multichannel fluorescent images are acquired with automated time-lapse microscopy. We provide detailed guidance for a computational analysis and parameterization pipeline. The entire procedure, from virus production to data analysis, can be completed in ∼10 d.
Collapse
|
27
|
Wilkinson EL, Sidaway JE, Cross MJ. Statin regulated ERK5 stimulates tight junction formation and reduces permeability in human cardiac endothelial cells. J Cell Physiol 2017. [PMID: 28639275 PMCID: PMC5655747 DOI: 10.1002/jcp.26064] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The MEKK3/MEK5/ERK5 signaling axis is required for cardiovascular development in vivo. We analyzed the physiological role of ERK5 in cardiac endothelial cells and the consequence of activation of this kinase by the statin class of HMG Co‐A reductase inhibitor drugs. We utilized human cardiac microvascular endothelial cells (HCMECs) and altered ERK5 expression using siRNA mediated gene silencing or overexpression of constitutively active MEK5 and ERK5 to reveal a role for ERK5 in regulating endothelial tight junction formation and cell permeability. Statin treatment of HCMECs stimulated activation of ERK5 and translocation to the plasma membrane resulting in co‐localization with the tight junction protein ZO‐1 and a concomitant reduction in endothelial cell permeability. Statin mediated activation of ERK5 was a consequence of reduced isoprenoid synthesis following HMG Co‐A reductase inhibition. Statin pretreatment could overcome the effect of doxorubicin in reducing endothelial tight junction formation and prevent increased permeability. Our data provide the first evidence for the role of ERK5 in regulating endothelial tight junction formation and endothelial cell permeability. Statin mediated ERK5 activation and the resulting decrease in cardiac endothelial cell permeability may contribute to the cardioprotective effects of statins in reducing doxorubicin‐induced cardiotoxicity.
Collapse
Affiliation(s)
- Emma L Wilkinson
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| | - James E Sidaway
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| | - Michael J Cross
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool, Liverpool, UK
| |
Collapse
|
28
|
Hoang VT, Yan TJ, Cavanaugh JE, Flaherty PT, Beckman BS, Burow ME. Oncogenic signaling of MEK5-ERK5. Cancer Lett 2017; 392:51-59. [PMID: 28153789 PMCID: PMC5901897 DOI: 10.1016/j.canlet.2017.01.034] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 01/17/2017] [Accepted: 01/23/2017] [Indexed: 12/17/2022]
Abstract
Mitogen-activated protein kinases (MAPKs) regulate diverse cellular processes including proliferation, cell survival, differentiation, and apoptosis. While conventional MAPK constituents have well-defined roles in oncogenesis, the MEK5 pathway has only recently emerged in cancer research. In this review, we consider the MEK5 signaling cascade, focusing specifically on its involvement in drug resistance and regulation of aggressive cancer phenotypes. Moreover, we explore the role of MEK5/ERK5 in tumorigenesis and metastatic progression, discussing the discrepancies in preclinical studies and assessing its viability as a therapeutic target for anti-cancer agents.
Collapse
Affiliation(s)
- Van T Hoang
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA, USA
| | - Thomas J Yan
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA, USA
| | - Jane E Cavanaugh
- Department of Pharmacological Sciences, Division of Medicinal Chemistry, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | - Patrick T Flaherty
- Department of Pharmacological Sciences, Division of Medicinal Chemistry, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | | | - Matthew E Burow
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA, USA; Department of Pharmacology, Tulane University, New Orleans, LA, USA; Tulane Cancer Center, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
29
|
Zheng R, Studzinski GP. Nuclear ERK5 inhibits progression of leukemic monocytes to macrophages by regulating the transcription factor PU.1 and heat shock protein HSP70. Leuk Lymphoma 2016; 58:1468-1480. [PMID: 27748139 DOI: 10.1080/10428194.2016.1243675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Differentiation therapy can supplement the therapy of APL, but other subtypes of AML are treated principally with cytotoxic agents, with few lasting remissions. While the induction of monocyte followed by macrophage differentiation by vitamin D derivatives (VDDs) is dramatic in cultured AML cells of all subtypes, attempts to translate this to the clinic have not been effective. Thus, better understanding of the mechanisms underlying VDD-induced differentiation may improve this approach. The key events in this form of differentiation include increased expression of CD11b, and the transcription factor PU.1 is known to be a part of this process. We show here that in the transition of monocytes to macrophages induced by a VDD, ERK5, a member of the MAPK family of signaling molecules, prevents PU.1 expression. However, upon ERK5 inhibition PU.1 protein is stabilized by HSP70.Thus, ERK5 may be a target for manipulation of the immunoregulatory actions of macrophages in cancer.
Collapse
Affiliation(s)
- Ruifang Zheng
- a Department of Pathology and Laboratory Medicine , New Jersey Medical School, Rutgers University , Newark , NJ , USA
| | - George P Studzinski
- a Department of Pathology and Laboratory Medicine , New Jersey Medical School, Rutgers University , Newark , NJ , USA
| |
Collapse
|
30
|
Seo HG, Kim HB, Kang MJ, Ryum JH, Yi EC, Cho JW. Identification of the nuclear localisation signal of O-GlcNAc transferase and its nuclear import regulation. Sci Rep 2016; 6:34614. [PMID: 27713473 PMCID: PMC5054401 DOI: 10.1038/srep34614] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 08/22/2016] [Indexed: 01/22/2023] Open
Abstract
Nucleocytoplasmic O-GlcNAc transferase (OGT) attaches a single GlcNAc to hydroxyl groups of serine and threonine residues. Although the cellular localisation of OGT is important to regulate a variety of cellular processes, the molecular mechanisms regulating the nuclear localisation of OGT is unclear. Here, we characterised three amino acids (DFP; residues 451-453) as the nuclear localisation signal of OGT and demonstrated that this motif mediated the nuclear import of non-diffusible β-galactosidase. OGT bound the importin α5 protein, and this association was abolished when the DFP motif of OGT was mutated or deleted. We also revealed that O-GlcNAcylation of Ser389, which resides in the tetratricopeptide repeats, plays an important role in the nuclear localisation of OGT. Our findings may explain how OGT, which possesses a NLS, exists in the nucleus and cytosol simultaneously.
Collapse
Affiliation(s)
- Hyeon Gyu Seo
- Department of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Han Byeol Kim
- Department of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Min Jueng Kang
- Department of Molecular Medicine and Biopharmaceutical Sciences, School of Convergence Science and Technology and College of Medicine or College of Pharmacy, Seoul National University, 28 Yeongeon-dong, Jongno-gu, Seoul 03080, Republic of Korea
| | - Joo Hwan Ryum
- Department of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Eugene C Yi
- Department of Molecular Medicine and Biopharmaceutical Sciences, School of Convergence Science and Technology and College of Medicine or College of Pharmacy, Seoul National University, 28 Yeongeon-dong, Jongno-gu, Seoul 03080, Republic of Korea
| | - Jin Won Cho
- Department of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
31
|
Wrobel K, Zhao YC, Kulkoyluoglu E, Chen KLA, Hieronymi K, Holloway J, Li S, Ray T, Ray PS, Landesman Y, Lipka AE, Smith RL, Madak-Erdogan Z. ERα-XPO1 Cross Talk Controls Tamoxifen Sensitivity in Tumors by Altering ERK5 Cellular Localization. Mol Endocrinol 2016; 30:1029-1045. [PMID: 27533791 PMCID: PMC5045498 DOI: 10.1210/me.2016-1101] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 08/12/2016] [Indexed: 12/22/2022] Open
Abstract
Most breast cancer deaths occur in women with recurrent, estrogen receptor (ER)-α(+), metastatic tumors. There is a critical need for therapeutic approaches that include novel, targetable mechanism-based strategies by which ERα (+) tumors can be resensitized to endocrine therapies. The objective of this study was to validate a group of nuclear transport genes as potential biomarkers to predict the risk of endocrine therapy failure and to evaluate the inhibition of XPO1, one of these genes as a novel means to enhance the effectiveness of endocrine therapies. Using advanced statistical methods, we found that expression levels of several of nuclear transport genes including XPO1 were associated with poor survival and predicted recurrence of tamoxifen-treated breast tumors in human breast cancer gene expression data sets. In mechanistic studies we showed that the expression of XPO1 determined the cellular localization of the key signaling proteins and the response to tamoxifen. We demonstrated that combined targeting of XPO1 and ERα in several tamoxifen-resistant cell lines and tumor xenografts with the XPO1 inhibitor, Selinexor, and tamoxifen restored tamoxifen sensitivity and prevented recurrence in vivo. The nuclear transport pathways have not previously been implicated in the development of endocrine resistance, and given the need for better strategies for selecting patients to receive endocrine modulatory reagents and improving therapy response of relapsed ERα(+) tumors, our findings show great promise for uncovering the role these pathways play in reducing cancer recurrences.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Hormonal/pharmacology
- Biological Transport/drug effects
- Biological Transport/genetics
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Cell Line, Tumor
- Cell Nucleus/drug effects
- Cell Nucleus/genetics
- Cell Nucleus/metabolism
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Humans
- Karyopherins/genetics
- Karyopherins/metabolism
- MCF-7 Cells
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Mitogen-Activated Protein Kinase 7/genetics
- Mitogen-Activated Protein Kinase 7/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Tamoxifen/pharmacology
- Exportin 1 Protein
Collapse
Affiliation(s)
- Kinga Wrobel
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Yiru Chen Zhao
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Eylem Kulkoyluoglu
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Karen Lee Ann Chen
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Kadriye Hieronymi
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Jamie Holloway
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Sarah Li
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Tania Ray
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Partha Sarathi Ray
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Yosef Landesman
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Alexander Edward Lipka
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Rebecca Lee Smith
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Zeynep Madak-Erdogan
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| |
Collapse
|
32
|
ERK5 kinase activity is dispensable for cellular immune response and proliferation. Proc Natl Acad Sci U S A 2016; 113:11865-11870. [PMID: 27679845 DOI: 10.1073/pnas.1609019113] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Unlike other members of the MAPK family, ERK5 contains a large C-terminal domain with transcriptional activation capability in addition to an N-terminal canonical kinase domain. Genetic deletion of ERK5 is embryonic lethal, and tissue-restricted deletions have profound effects on erythroid development, cardiac function, and neurogenesis. In addition, depletion of ERK5 is antiinflammatory and antitumorigenic. Small molecule inhibition of ERK5 has been shown to have promising activity in cell and animal models of inflammation and oncology. Here we report the synthesis and biological characterization of potent, selective ERK5 inhibitors. In contrast to both genetic depletion/deletion of ERK5 and inhibition with previously reported compounds, inhibition of the kinase with the most selective of the new inhibitors had no antiinflammatory or antiproliferative activity. The source of efficacy in previously reported ERK5 inhibitors is shown to be off-target activity on bromodomains, conserved protein modules involved in recognition of acetyl-lysine residues during transcriptional processes. It is likely that phenotypes reported from genetic deletion or depletion of ERK5 arise from removal of a noncatalytic function of ERK5. The newly reported inhibitors should be useful in determining which of the many reported phenotypes are due to kinase activity and delineate which can be pharmacologically targeted.
Collapse
|
33
|
Gomez N, Erazo T, Lizcano JM. ERK5 and Cell Proliferation: Nuclear Localization Is What Matters. Front Cell Dev Biol 2016; 4:105. [PMID: 27713878 PMCID: PMC5031611 DOI: 10.3389/fcell.2016.00105] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 09/06/2016] [Indexed: 12/31/2022] Open
Abstract
ERK5, the last MAP kinase family member discovered, is activated by the upstream kinase MEK5 in response to growth factors and stress stimulation. MEK5-ERK5 pathway has been associated to different cellular processes, playing a crucial role in cell proliferation in normal and cancer cells by mechanisms that are both dependent and independent of its kinase activity. Thus, nuclear ERK5 activates transcription factors by either direct phosphorylation or acting as co-activator thanks to a unique transcriptional activation TAD domain located at its C-terminal tail. Consequently, ERK5 has been proposed as an interesting target to tackle different cancers, and either inhibitors of ERK5 activity or silencing the protein have shown antiproliferative activity in cancer cells and to block tumor growth in animal models. Here, we review the different mechanisms involved in ERK5 nuclear translocation and their consequences. Inactive ERK5 resides in the cytosol, forming a complex with Hsp90-Cdc37 superchaperone. In a canonical mechanism, MEK5-dependent activation results in ERK5 C-terminal autophosphorylation, Hsp90 dissociation, and nuclear translocation. This mechanism integrates signals such as growth factors and stresses that activate the MEK5-ERK5 pathway. Importantly, two other mechanisms, MEK5-independent, have been recently described. These mechanisms allow nuclear shuttling of kinase-inactive forms of ERK5. Although lacking kinase activity, these forms activate transcription by interacting with transcription factors through the TAD domain. Both mechanisms also require Hsp90 dissociation previous to nuclear translocation. One mechanism involves phosphorylation of the C-terminal tail of ERK5 by kinases that are activated during mitosis, such as Cyclin-dependent kinase-1. The second mechanism involves overexpression of chaperone Cdc37, an oncogene that is overexpressed in cancers such as prostate adenocarcinoma, where it collaborates with ERK5 to promote cell proliferation. Although some ERK5 kinase inhibitors have shown antiproliferative activity it is likely that those tumors expressing kinase-inactive nuclear ERK5 will not respond to these inhibitors.
Collapse
Affiliation(s)
| | | | - Jose M. Lizcano
- Protein Kinases and Signal Transduction Laboratory, Institut de Neurociencies and Departament de Bioquimica i Biologia Molecular, Facultat de Medicina, Universitat Autonoma de BarcelonaBarcelona, Spain
| |
Collapse
|
34
|
Tsioumpekou M, Papadopoulos N, Burovic F, Heldin CH, Lennartsson J. Platelet-derived growth factor (PDGF)-induced activation of Erk5 MAP-kinase is dependent on Mekk2, Mek1/2, PKC and PI3-kinase, and affects BMP signaling. Cell Signal 2016; 28:1422-1431. [PMID: 27339033 DOI: 10.1016/j.cellsig.2016.06.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 06/16/2016] [Accepted: 06/16/2016] [Indexed: 02/09/2023]
Abstract
Platelet-derived growth factor-BB (PDGF-BB) binds to its tyrosine kinase receptors (PDGFRs) and stimulates mitogenicity and survival of cells of mesenchymal origin. Activation of PDGFRs initiates a number of downstream signaling pathways, including phosphatidyl 3'-inositol kinase (PI3-kinase), phospholipase Cγ and MAP kinase pathways. In this report, we show that Erk5 MAP kinase is activated in response to PDGF-BB in the smooth muscle cell line MOVAS in a manner dependent on Mekk2, Mek1/2, Mek5, PI3-kinase and protein kinase C (PKC). The co-operation of Mek1/2 and Mekk2 in the activation of Erk5, suggests a close co-regulation between the Erk1/2 and Erk5 MAP kinase pathways. Furthermore, we found that classical PKCs are important for Erk5 activation. In addition, we found that PKCζ interacts with Erk5 and may exert a negative feed-back effect. We observed no nuclear accumulation of Erk5 in response to PDGF-BB stimulation, however, we identified a mechanism by which cytoplasmic Erk5 influences gene expression; Erk5 was essential for PDGF-BB-mediated Smad1/5/8 signaling by stimulating release and/or activation of bone morphogenetic protein(s) (BMPs). Thus, PDGF-BB-induced Erk5 activation involves parallel stimulatory and inhibitory pathways and promotes Smad1/5/8 signaling.
Collapse
Affiliation(s)
- Maria Tsioumpekou
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE-751 24 Uppsala, Sweden
| | - Natalia Papadopoulos
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE-751 24 Uppsala, Sweden
| | - Fatima Burovic
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE-751 24 Uppsala, Sweden
| | - Carl-Henrik Heldin
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE-751 24 Uppsala, Sweden
| | - Johan Lennartsson
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE-751 24 Uppsala, Sweden.
| |
Collapse
|
35
|
Koliou X, Fedonidis C, Kalpachidou T, Mangoura D. Nuclear import mechanism of neurofibromin for localization on the spindle and function in chromosome congression. J Neurochem 2015; 136:78-91. [PMID: 26490262 DOI: 10.1111/jnc.13401] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 10/12/2015] [Accepted: 10/15/2015] [Indexed: 12/28/2022]
Abstract
Neurofibromatosis type-1 (NF-1) is caused by mutations in the tumor suppressor gene NF1; its protein product neurofibromin is a RasGTPase-activating protein, a property that has yet to explain aneuploidy, most often observed in astrocytes in NF-1. Here, we provide a mechanistic model for the regulated nuclear import of neurofibromin during the cell cycle and for a role in chromosome congression. Specifically, we demonstrate that neurofibromin, phosphorylated on Ser2808, a residue adjacent to a nuclear localization signal in the C-terminal domain (CTD), by Protein Kinase C-epsilon (PKC-ε), accumulates in a Ran-dependent manner and through binding to lamin in the nucleus at G2 in glioblastoma cells. Furthermore, we identify CTD as a tubulin-binding domain and show that a phosphomimetic substitution of its Ser2808 results in a predominantly nuclear localization. Confocal analysis shows that endogenous neurofibromin localizes on the centrosomes at interphase, as well as on the mitotic spindle, through direct associations with tubulins, in glioblastoma cells and primary astrocytes. More importantly, analysis of mitotic phenotypes after siRNA-mediated depletion shows that acute loss of this tumor suppressor protein leads to aberrant chromosome congression at the metaphase plate. Therefore, neurofibromin protein abundance and nuclear import are mechanistically linked to an error-free chromosome congression. Concerned with neurofibromin's, a tumor suppressor, mechanism of action, we demonstrate in astrocytic cells that its synthesis, phosphorylation by Protein Kinase C-ε on Ser2808 (a residue adjacent to a nuclear localization sequence), and nuclear import are cell cycle-dependent, being maximal at G2. During mitosis, neurofibromin is an integral part of the spindle, while its depletion leads to aberrant chromosome congression, possibly explaining the development of chromosomal instability in Neurofibromatosis type-1. Read the Editorial Highlight for this article on page 11. Cover Image for this issue: doi: 10.1111/jnc.13300.
Collapse
Affiliation(s)
- Xeni Koliou
- Basic Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Constantinos Fedonidis
- Basic Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Theodora Kalpachidou
- Basic Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Dimitra Mangoura
- Basic Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
36
|
Chu UB, Duellman T, Weaver SJ, Tao Y, Yang J. Endothelial protective genes induced by statin are mimicked by ERK5 activation as triggered by a drug combination of FTI-277 and GGTI-298. Biochim Biophys Acta Gen Subj 2015; 1850:1415-25. [PMID: 25829196 DOI: 10.1016/j.bbagen.2015.03.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 03/08/2015] [Accepted: 03/23/2015] [Indexed: 01/12/2023]
Abstract
BACKGROUND Statins are potent inhibitors of cholesterol biosynthesis and are clinically beneficial in preventing cardiovascular diseases, however, the therapeutic utility of these drugs is limited by myotoxicity. Here, we explored the mechanism of statin-mediated activation of ERK5 in the human endothelium with the goal of identifying compounds that confer endothelial protection but are nontoxic to muscle. METHODS An ERK5-one hybrid luciferase reporter transfected into COS-7 cells with pharmacological and molecular manipulations dissected the signaling pathway leading to statin activation of ERK5. qRT-PCR of HUVEC cells documented the transcriptional activation of endothelial-protective genes. Lastly, morphological and cellular ATP analysis, and induction of atrogin-1 in C2C12 myotubes were used to assess statin-induced myopathy. RESULTS Statin activation of ERK5 is dependent on the cellular reduction of GGPPs. Furthermore, we found that the combination of FTI-277 (inhibitor of farnesyl transferase) and GGTI-298 (inhibitor of geranylgeranyl transferase I) mimicked the statin-mediated activation of ERK5. FTI-277 and GGTI-298 together recapitulated the beneficial effects of statins by transcriptionally upregulating anti-inflammatory mediators such as eNOS, THBD, and KLF2. Finally, C2C12 skeletal myotubes treated with both FTI-277 and GGTI-298 evoked less morphological and cellular changes recognized as biomarkers of statin-associated myopathy. CONCLUSIONS Statin-induced endothelial protection and myopathy are mediated by distinct metabolic intermediates and co-inhibition of farnesyl transferase and geranylgeranyl transferase I confer endothelial protection without myopathy. GENERAL SIGNIFICANCE The combinatorial FTI-277 and GGTI-298 drug regimen provides a promising alternative avenue for endothelial protection without myopathy.
Collapse
Affiliation(s)
- Uyen B Chu
- Department of Anesthesiology, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53706 USA
| | - Tyler Duellman
- Department of Anesthesiology, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53706 USA; Training Program in Translational Cardiovascular Medicine, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53706 USA
| | - Sara J Weaver
- Department of Anesthesiology, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53706 USA
| | - Yunting Tao
- Department of Anesthesiology, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53706 USA
| | - Jay Yang
- Department of Anesthesiology, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53706 USA; Training Program in Translational Cardiovascular Medicine, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53706 USA.
| |
Collapse
|
37
|
Gocek E, Studzinski GP. The Potential of Vitamin D-Regulated Intracellular Signaling Pathways as Targets for Myeloid Leukemia Therapy. J Clin Med 2015; 4:504-34. [PMID: 26239344 PMCID: PMC4470153 DOI: 10.3390/jcm4040504] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 01/06/2015] [Accepted: 03/06/2015] [Indexed: 02/06/2023] Open
Abstract
The current standard regimens for the treatment of acute myeloid leukemia (AML) are curative in less than half of patients; therefore, there is a great need for innovative new approaches to this problem. One approach is to target new treatments to the pathways that are instrumental to cell growth and survival with drugs that are less harmful to normal cells than to neoplastic cells. In this review, we focus on the MAPK family of signaling pathways and those that are known to, or potentially can, interact with MAPKs, such as PI3K/AKT/FOXO and JAK/STAT. We exemplify the recent studies in this field with specific relevance to vitamin D and its derivatives, since they have featured prominently in recent scientific literature as having anti-cancer properties. Since microRNAs also are known to be regulated by activated vitamin D, this is also briefly discussed here, as are the implications of the emerging acquisition of transcriptosome data and potentiation of the biological effects of vitamin D by other compounds. While there are ongoing clinical trials of various compounds that affect signaling pathways, more studies are needed to establish the clinical utility of vitamin D in the treatment of cancer.
Collapse
Affiliation(s)
- Elzbieta Gocek
- Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland.
| | - George P Studzinski
- Department of Pathology, New Jersey Medical School, Rutgers, The State University of New Jersey, 185 South Orange Ave., Newark, NJ 17101, USA.
| |
Collapse
|
38
|
Honda T, Obara Y, Yamauchi A, Couvillon AD, Mason JJ, Ishii K, Nakahata N. Phosphorylation of ERK5 on Thr732 is associated with ERK5 nuclear localization and ERK5-dependent transcription. PLoS One 2015; 10:e0117914. [PMID: 25689862 PMCID: PMC4331489 DOI: 10.1371/journal.pone.0117914] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 01/05/2015] [Indexed: 12/20/2022] Open
Abstract
Extracellular signal-regulated kinases (ERKs) play critical roles in numerous cellular processes, including proliferation and differentiation. ERK5 contains a kinase domain at the N-terminal, and the unique extended C-terminal includes multiple autophosphorylation sites that enhance ERK5-dependent transcription. However, the impact of phosphorylation at the various sites remain unclear. In this study, we examined the role of phosphorylation at the ERK5 C-terminal. We found that a constitutively active MEK5 mutant phosphorylated ERK5 at the TEY motif, resulting in the sequential autophosphorylation of multiple C-terminal residues, including Thr732 and Ser769/773/775. However, when ERK1/2 was selectively activated by an oncogenic RAS mutant, ERK5 phosphorylation at Thr732 was induced without affecting the phosphorylation status at TEY or Ser769/773/775. The Thr732 phosphorylation was U0126-sensitive and was observed in a kinase-dead mutant of ERK5 as well, suggesting that ERK1/2 can phosphorylate ERK5 at Thr732. This phosphorylation was also promoted by epidermal growth factor and nerve growth factor in HEK293 and PC12 cells, respectively. The ERK5–T732A mutant was localized in the cytosol under basal conditions. In contrast, ERK5 phosphorylated at Thr732 via the RAS-ERK1/2 pathway and ERK5–T732E, which mimics the phosphorylated form, were localized in both the nucleus and cytosol. Finally, ER–32A and U0126 blocked ERK5-dependent MEF2C transcriptional activity. Based on these findings, we propose a novel cross-talk mechanism in which ERK1/2, following activation by growth factor stimulation, phosphorylates ERK5 at Thr732. This phosphorylation event is responsible for ERK5 nuclear localization and ERK5-dependent transcription.
Collapse
Affiliation(s)
- Takuto Honda
- Department of Cellular Signaling, Graduate School of Pharmaceutical Sciences, Tohoku University, 6–3 Aoba, Aramaki, Aoba-ku, Sendai 980–8578, Japan
| | - Yutaro Obara
- Department of Cellular Signaling, Graduate School of Pharmaceutical Sciences, Tohoku University, 6–3 Aoba, Aramaki, Aoba-ku, Sendai 980–8578, Japan
- Department of Pharmacology, Yamagata University School of Medicine, 2–2–2 Iida-Nishi, Yamagata, 990–9585, Japan
- * E-mail:
| | - Arata Yamauchi
- Department of Cellular Signaling, Graduate School of Pharmaceutical Sciences, Tohoku University, 6–3 Aoba, Aramaki, Aoba-ku, Sendai 980–8578, Japan
| | - Anthony D. Couvillon
- Cell Signaling Technology, 3 Trask Lane, Danvers, MA 01923, United States of America
| | - Justin J. Mason
- Cell Signaling Technology, 3 Trask Lane, Danvers, MA 01923, United States of America
| | - Kuniaki Ishii
- Department of Pharmacology, Yamagata University School of Medicine, 2–2–2 Iida-Nishi, Yamagata, 990–9585, Japan
| | - Norimichi Nakahata
- Department of Cellular Signaling, Graduate School of Pharmaceutical Sciences, Tohoku University, 6–3 Aoba, Aramaki, Aoba-ku, Sendai 980–8578, Japan
| |
Collapse
|
39
|
Umapathy G, El Wakil A, Witek B, Chesler L, Danielson L, Deng X, Gray NS, Johansson M, Kvarnbrink S, Ruuth K, Schönherr C, Palmer RH, Hallberg B. The kinase ALK stimulates the kinase ERK5 to promote the expression of the oncogene MYCN in neuroblastoma. Sci Signal 2014; 7:ra102. [PMID: 25351247 DOI: 10.1126/scisignal.2005470] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Anaplastic lymphoma kinase (ALK) is an important molecular target in neuroblastoma. Although tyrosine kinase inhibitors abrogating ALK activity are currently in clinical use for the treatment of ALK-positive (ALK(+)) disease, monotherapy with ALK tyrosine kinase inhibitors may not be an adequate solution for ALK(+) neuroblastoma patients. Increased expression of the gene encoding the transcription factor MYCN is common in neuroblastomas and correlates with poor prognosis. We found that the kinase ERK5 [also known as big mitogen-activated protein kinase (MAPK) 1 (BMK1)] is activated by ALK through a pathway mediated by phosphoinositide 3-kinase (PI3K), AKT, MAPK kinase kinase 3 (MEKK3), and MAPK kinase 5 (MEK5). ALK-induced transcription of MYCN and stimulation of cell proliferation required ERK5. Pharmacological or RNA interference-mediated inhibition of ERK5 suppressed the proliferation of neuroblastoma cells in culture and enhanced the antitumor efficacy of the ALK inhibitor crizotinib in both cells and xenograft models. Together, our results indicate that ERK5 mediates ALK-induced transcription of MYCN and proliferation of neuroblastoma, suggesting that targeting both ERK5 and ALK may be beneficial in neuroblastoma patients.
Collapse
Affiliation(s)
- Ganesh Umapathy
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Göteborg, Sweden
| | - Abeer El Wakil
- Department of Molecular Biology, Building 6L, Umeå University, 901 87 Umeå, Sweden
| | - Barbara Witek
- Department of Molecular Biology, Building 6L, Umeå University, 901 87 Umeå, Sweden
| | - Louis Chesler
- The Institute of Cancer Research, The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5NG, UK
| | - Laura Danielson
- The Institute of Cancer Research, The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5NG, UK
| | - Xianming Deng
- School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China. Dana-Farber Cancer Institute, Harvard Medical School, Seeley G. Mudd Building, 628A, 250 Longwood Avenue, Boston, MA 02115, USA
| | - Nathanael S Gray
- Dana-Farber Cancer Institute, Harvard Medical School, Seeley G. Mudd Building, 628A, 250 Longwood Avenue, Boston, MA 02115, USA
| | - Mikael Johansson
- Department of Radiation Sciences, Oncology, Umeå University, 901 87 Umea, Sweden
| | - Samuel Kvarnbrink
- Department of Radiation Sciences, Oncology, Umeå University, 901 87 Umea, Sweden
| | - Kristina Ruuth
- Department of Molecular Biology, Building 6L, Umeå University, 901 87 Umeå, Sweden
| | - Christina Schönherr
- Department of Molecular Biology, Building 6L, Umeå University, 901 87 Umeå, Sweden
| | - Ruth H Palmer
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Göteborg, Sweden. Department of Molecular Biology, Building 6L, Umeå University, 901 87 Umeå, Sweden
| | - Bengt Hallberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-405 30 Göteborg, Sweden.
| |
Collapse
|
40
|
Shalaby S, Chitragari G, Sumpio BJ, Sumpio BE. Characterization of extracellular signal-regulated kinase 5 levels in human umbilical vein endothelial cells exposed to disturbed and uniform flow. Int J Angiol 2014; 23:187-92. [PMID: 25317031 DOI: 10.1055/s-0034-1378136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Extracellular signal-regulated kinase 5 (ERK5) has been reported to regulate endothelial cell integrity and protect from vascular dysfunction under continuous laminar flow. However, the effect of flow on ERK5 levels has not been determined. Confluent human umbilical vein endothelial cells (HUVECs) were seeded on fibronectin coated glass slides and serum starved for 2 hours with 1% fetal bovine serum (FBS). HUVECs were then exposed to to and fro flow (TFF), pulsatile forward flow (PFF), or continuous laminar flow (CLF) in a parallel plate flow chamber for up to 2 hours. At the end of experiment, cell lysates were prepared and immunoblotted with antibodies to total ERK5. Both CLF and TFF exhibited a decrease in ERK5 after levels after 2-hour exposure. However, the level of ERK5 for PFF remained the same. Disturbed, but not uniform pulsatile, flow decreases ERK5 levels in HUVECs.
Collapse
Affiliation(s)
- Sherif Shalaby
- Department of Vascular Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Gautham Chitragari
- Department of Vascular Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Brandon J Sumpio
- Department of Vascular Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Bauer E Sumpio
- Department of Vascular Surgery, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
41
|
Eger G, Papadopoulos N, Lennartsson J, Heldin CH. NR4A1 promotes PDGF-BB-induced cell colony formation in soft agar. PLoS One 2014; 9:e109047. [PMID: 25269081 PMCID: PMC4182636 DOI: 10.1371/journal.pone.0109047] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 08/30/2014] [Indexed: 01/04/2023] Open
Abstract
The fibroblast mitogen platelet-derived growth factor -BB (PDGF-BB) induces a transient expression of the orphan nuclear receptor NR4A1 (also named Nur77, TR3 or NGFIB). The aim of the present study was to investigate the pathways through which NR4A1 is induced by PDGF-BB and its functional role. We demonstrate that in PDGF-BB stimulated NIH3T3 cells, the MEK1/2 inhibitor CI-1040 strongly represses NR4A1 expression, whereas Erk5 downregulation delays the expression, but does not block it. Moreover, we report that treatment with the NF-κB inhibitor BAY11-7082 suppresses NR4A1 mRNA and protein expression. The majority of NR4A1 in NIH3T3 was found to be localized in the cytoplasm and only a fraction was translocated to the nucleus after continued PDGF-BB treatment. Silencing NR4A1 slightly increased the proliferation rate of NIH3T3 cells; however, it did not affect the chemotactic or survival abilities conferred by PDGF-BB. Moreover, overexpression of NR4A1 promoted anchorage-independent growth of NIH3T3 cells and the glioblastoma cell lines U-105MG and U-251MG. Thus, whereas NR4A1, induced by PDGF-BB, suppresses cell growth on a solid surface, it increases anchorage-independent growth.
Collapse
MESH Headings
- Agar
- Animals
- Becaplermin
- Benzamides/pharmacology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Chemotaxis/drug effects
- Gene Expression Regulation
- Humans
- MAP Kinase Kinase 1/antagonists & inhibitors
- MAP Kinase Kinase 1/genetics
- MAP Kinase Kinase 1/metabolism
- MAP Kinase Kinase 2/antagonists & inhibitors
- MAP Kinase Kinase 2/genetics
- MAP Kinase Kinase 2/metabolism
- Mice
- Mitogen-Activated Protein Kinase 7/antagonists & inhibitors
- Mitogen-Activated Protein Kinase 7/genetics
- Mitogen-Activated Protein Kinase 7/metabolism
- NF-kappa B/antagonists & inhibitors
- NF-kappa B/genetics
- NF-kappa B/metabolism
- NIH 3T3 Cells
- Neuroglia/drug effects
- Neuroglia/metabolism
- Neuroglia/pathology
- Nitriles/pharmacology
- Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Protein Kinase Inhibitors/pharmacology
- Proto-Oncogene Proteins c-sis/pharmacology
- RNA, Messenger/antagonists & inhibitors
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction
- Sulfones/pharmacology
Collapse
Affiliation(s)
- Glenda Eger
- Ludwig Institute for Cancer Research, Uppsala University, Uppsala, Sweden
| | | | - Johan Lennartsson
- Ludwig Institute for Cancer Research, Uppsala University, Uppsala, Sweden
- * E-mail:
| | - Carl-Henrik Heldin
- Ludwig Institute for Cancer Research, Uppsala University, Uppsala, Sweden
| |
Collapse
|
42
|
Intracellular mobility and nuclear trafficking of the stress-activated kinase JNK1 are impeded by hyperosmotic stress. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:253-64. [DOI: 10.1016/j.bbamcr.2013.10.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 10/15/2013] [Accepted: 10/21/2013] [Indexed: 12/22/2022]
|
43
|
Beta-like importins mediate the nuclear translocation of mitogen-activated protein kinases. Mol Cell Biol 2013; 34:259-70. [PMID: 24216760 DOI: 10.1128/mcb.00799-13] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The rapid nuclear translocation of signaling proteins upon stimulation is important for the regulation of de novo gene expression. We have studied the stimulated nuclear shuttling of c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinases (MAPKs) and found that they translocate into the nucleus in a Ran-dependent, but NLS- or NTS-independent, manner, unrelated to their catalytic activity. We show that this translocation involves three β-like importins, importins 3, 7, and 9 (Imp3/7/9). Knockdown of these importins inhibits the nuclear translocation of the MAPKs and, thereby, activation of their transcription factor targets. We further demonstrate that the translocation requires the stimulated formation of heterotrimers composed of Imp3/Imp7/MAPK or Imp3/Imp9/MAPK. JNK1/2 and p38α/β bind to either Imp7 or Imp9 upon stimulated posttranslational modification of the two Imps, while Imp3 joins the complex after its stimulation-induced phosphorylation. Once formed, these heterotrimers move to the nuclear envelope, where importin 3 remains, while importins 7 and 9 escort the MAPKs into the nucleus. These results suggest that β-like importins are central mediators of stimulated nuclear translocation of signaling proteins and therefore add a central level of regulation to stimulated transcription.
Collapse
|
44
|
Abstract
Classic nuclear shuttling is mediated by an importin-α∙β heterodimer that binds to cargoes containing a nuclear localization signal, and shuttles most nuclear proteins immediately after their translation. Aside from this canonical mechanism, kariopheryn-βs or β-like importins operate by binding to non-canonical nuclear localization signals to mediate translocation without the assistance of importin-α. The mechanism by which these components operate is much less understood and is currently under investigation. Recently, several β-like importins have been implicated in the stimulated nuclear translocation of signaling proteins. Here, we propose that this group of importins might be responsible for the swift nuclear shuttling of many proteins following various stimuli.
Collapse
|
45
|
Wierk JK, Langbehn A, Kamper M, Richter S, Burda PC, Heussler VT, Deschermeier C. Plasmodium berghei MAPK1 displays differential and dynamic subcellular localizations during liver stage development. PLoS One 2013; 8:e59755. [PMID: 23544094 PMCID: PMC3609774 DOI: 10.1371/journal.pone.0059755] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Accepted: 02/18/2013] [Indexed: 11/18/2022] Open
Abstract
Mitogen-activated protein kinases (MAPKs) regulate key signaling events in eukaryotic cells. In the genomes of protozoan Plasmodium parasites, the causative agents of malaria, two genes encoding kinases with significant homology to other eukaryotic MAPKs have been identified (mapk1, mapk2). In this work, we show that both genes are transcribed during Plasmodium berghei liver stage development, and analyze expression and subcellular localization of the PbMAPK1 protein in liver stage parasites. Live cell imaging of transgenic parasites expressing GFP-tagged PbMAPK1 revealed a nuclear localization of PbMAPK1 in the early schizont stage mediated by nuclear localization signals in the C-terminal domain. In contrast, a distinct localization of PbMAPK1 in comma/ring-shaped structures in proximity to the parasite's nuclei and the invaginating parasite membrane was observed during the cytomere stage of parasite development as well as in immature blood stage schizonts. The PbMAPK1 localization was found to be independent of integrity of a motif putatively involved in ATP binding, integrity of the putative activation motif and the presence of a predicted coiled-coil domain in the C-terminal domain. Although PbMAPK1 knock out parasites showed normal liver stage development, the kinase may still fulfill a dual function in both schizogony and merogony of liver stage parasites regulated by its dynamic and stage-dependent subcellular localization.
Collapse
Affiliation(s)
- Jannika Katharina Wierk
- Department of Molecular Parasitology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Annette Langbehn
- Department of Molecular Parasitology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Maria Kamper
- Department of Molecular Parasitology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Stefanie Richter
- Department of Molecular Parasitology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | | | - Christina Deschermeier
- Department of Molecular Parasitology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| |
Collapse
|
46
|
Canonical and kinase activity-independent mechanisms for extracellular signal-regulated kinase 5 (ERK5) nuclear translocation require dissociation of Hsp90 from the ERK5-Cdc37 complex. Mol Cell Biol 2013; 33:1671-86. [PMID: 23428871 DOI: 10.1128/mcb.01246-12] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The mitogen-activated protein (MAP) kinase extracellular signal-regulated kinase 5 (ERK5) plays a crucial role in cell proliferation, regulating gene transcription. ERK5 has a unique C-terminal tail which contains a transcriptional activation domain, and activates transcription by phosphorylating transcription factors and acting itself as a transcriptional coactivator. However, the molecular mechanisms that regulate its nucleocytoplasmatic traffic are unknown. We have used tandem affinity purification to identify proteins that interact with ERK5. We show that ERK5 interacts with the Hsp90-Cdc37 chaperone in resting cells, and that inhibition of Hsp90 or Cdc37 results in ERK5 ubiquitylation and proteasomal degradation. Interestingly, activation of cellular ERK5 induces Hsp90 dissociation from the ERK5-Cdc37 complex, leading to ERK5 nuclear translocation and activation of transcription, by a mechanism which requires the autophosphorylation at its C-terminal tail. Consequently, active ERK5 is no longer sensitive to Hsp90 or Cdc37 inhibitors. Cdc37 overexpression also induces Hsp90 dissociation and the nuclear translocation of a kinase-inactive form of ERK5 which retains transcriptional activity. This is the first example showing that ERK5 transcriptional activity does not require kinase activity. Since Cdc37 cooperates with ERK5 to promote cell proliferation, Cdc37 overexpression (as happens in some cancers) might represent a new, noncanonical mechanism by which ERK5 regulates tumor proliferation.
Collapse
|
47
|
Glatz G, Gógl G, Alexa A, Reményi A. Structural mechanism for the specific assembly and activation of the extracellular signal regulated kinase 5 (ERK5) module. J Biol Chem 2013; 288:8596-8609. [PMID: 23382384 PMCID: PMC3605678 DOI: 10.1074/jbc.m113.452235] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Mitogen-activated protein kinase (MAPK) activation depends on a linear binding motif found in all MAPK kinases (MKK). In addition, the PB1 (Phox and Bem1) domain of MKK5 is required for extracellular signal regulated kinase 5 (ERK5) activation. We present the crystal structure of ERK5 in complex with an MKK5 construct comprised of the PB1 domain and the linear binding motif. We show that ERK5 has distinct protein-protein interaction surfaces compared with ERK2, which is the closest ERK5 paralog. The two MAPKs have characteristically different physiological functions and their distinct protein-protein interaction surface topography enables them to bind different sets of activators and substrates. Structural and biochemical characterization revealed that the MKK5 PB1 domain cooperates with the MAPK binding linear motif to achieve substrate specific binding, and it also enables co-recruitment of the upstream activating enzyme and the downstream substrate into one signaling competent complex. Studies on present day MAPKs and MKKs hint on the way protein kinase networks may evolve. In particular, they suggest how paralogous enzymes with similar catalytic properties could acquire novel signaling roles by merely changing the way they make physical links to other proteins.
Collapse
Affiliation(s)
- Gábor Glatz
- Department of Biochemistry, Eötvös Loránd University, Budapest H-1117, Hungary
| | - Gergő Gógl
- Department of Biochemistry, Eötvös Loránd University, Budapest H-1117, Hungary
| | - Anita Alexa
- Department of Biochemistry, Eötvös Loránd University, Budapest H-1117, Hungary
| | - Attila Reményi
- Department of Biochemistry, Eötvös Loránd University, Budapest H-1117, Hungary.
| |
Collapse
|
48
|
Prévost M, Chamousset D, Nasa I, Freele E, Morrice N, Moorhead G, Trinkle-Mulcahy L. Quantitative fragmentome mapping reveals novel, domain-specific partners for the modular protein RepoMan (recruits PP1 onto mitotic chromatin at anaphase). Mol Cell Proteomics 2013; 12:1468-86. [PMID: 23362328 DOI: 10.1074/mcp.m112.023291] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
RepoMan is a protein phosphatase 1 (PP1) regulatory subunit that targets the phosphatase to key substrates throughout the cell cycle. Most work to date has focused on the mitotic roles of RepoMan/PP1, although equally important interphase role(s) have been demonstrated. Initial mapping of the interactome of nuclear RepoMan, both endogenous and tagged, was complicated by various factors, including antibody cross-reactivity and low sensitivity of the detection of chromatin-associated partners above the high background of proteins that bind nonspecifically to affinity matrices. We therefore adapted the powerful combination of fluorescence imaging with labeling-based quantitative proteomics to map the "fragmentomes" of specific regions of RepoMan. These regions demonstrate distinct localization patterns and turnover dynamics that reflect underlying binding events. The increased sensitivity and signal-to-noise ratio provided by this unique approach facilitated identification of a large number of novel RepoMan interactors, several of which were rigorously validated in follow-up experiments, including the association of RepoMan/PP1 with a specific PP2A-B56γ complex, interaction with ribosomal proteins and import factors involved in their nucleocytoplasmic transport and interaction with proteins involved in the response to DNA damage. This same strategy can be used to investigate the cellular roles of other modular proteins.
Collapse
Affiliation(s)
- Michèle Prévost
- Department of Cellular and Molecular Medicine and Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
49
|
Yang SH, Sharrocks AD, Whitmarsh AJ. MAP kinase signalling cascades and transcriptional regulation. Gene 2012; 513:1-13. [PMID: 23123731 DOI: 10.1016/j.gene.2012.10.033] [Citation(s) in RCA: 328] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Revised: 10/08/2012] [Accepted: 10/18/2012] [Indexed: 02/06/2023]
Abstract
The MAP kinase (MAPK) signalling pathways play fundamental roles in a wide range of cellular processes and are often deregulated in disease states. One major mode of action for these pathways is in controlling gene expression, in particular through regulating transcription. In this review, we discuss recent significant advances in this area. In particular we focus on the mechanisms by which MAPKs are targeted to the nucleus and chromatin, and once there, how they impact on chromatin structure and subsequent gene regulation. We also discuss how systems biology approaches have contributed to our understanding of MAPK signaling networks, and also how the MAPK pathways intersect with other regulatory pathways in the nucleus. Finally, we summarise progress in studying the physiological functions of key MAPK transcriptional targets.
Collapse
Affiliation(s)
- Shen-Hsi Yang
- Faculty of Life Sciences, University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | | | | |
Collapse
|
50
|
Cytoskeletal reorganization mediates fluid shear stress-induced ERK5 activation in osteoblastic cells. Cell Biol Int 2012; 36:229-36. [PMID: 21954859 DOI: 10.1042/cbi20110113] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Mechanotransduction is a complicated process, of which mechanosensation is the first step. Previous studies have shown that the cytoskeleton plays a crucial role in mechanosensation and the mediation of intracellular signal transduction. However, the mechanism of mechanotransduction in the bone remains elusive. Here, we investigated the potential involvement of a novel MAPK (mitogen-activated protein kinase) member, ERK5 (extracellular-signal-regulated kinase 5), in the response of osteoblastic cells to FSS (fluid shear stress). Our results demonstrated that ERK5 was rapidly phosphorylated in pre-osteoblastic MC3T3-E1 cells upon FSS, and the integrity and reorganization of the cytoskeleton were critical in this process, in which the cytoskeleton-dependent activation of FAK (focal adhesion kinase) may be involved in the activation of ERK5 induced by FSS. Moreover, we found that cytoskeletal disruption led to significant down-regulation of ERK5 phosphorylation, but had no effect on ERK5 nuclear localization. Furthermore, the cytoskeleton rapidly reorganized in response to FSS, but long-time fluid load, even at a physiological level, led to cytoskeletal disruption, suggesting that other pathways may be involved in long-term mechanotransduction. Taken together, our data provide new insight into the mechanisms of mechanosensation by highlighting the link between ERK5 activation and cytoskeletal reorganization in osteoblasts undergoing FSS.
Collapse
|