1
|
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
|
2
|
Song C, Zhang Z, Leng D, He Z, Wang X, Liu W, Zhang W, Wu Q, Zhao Q, Chen G. ERK5 promotes autocrine expression to sustain mitogenic balance for cell fate specification in human pluripotent stem cells. Stem Cell Reports 2024; 19:1320-1335. [PMID: 39151429 PMCID: PMC11411316 DOI: 10.1016/j.stemcr.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 08/19/2024] Open
Abstract
The homeostasis of human pluripotent stem cells (hPSCs) requires the signaling balance of extracellular factors. Exogenous regulators from cell culture medium have been widely reported, but little attention has been paid to the autocrine factor from hPSCs themselves. In this report, we demonstrate that extracellular signal-related kinase 5 (ERK5) regulates endogenous autocrine factors essential for pluripotency and differentiation. ERK5 inhibition leads to erroneous cell fate specification in all lineages even under lineage-specific induction. hPSCs can self-renew under ERK5 inhibition in the presence of fibroblast growth factor 2 (FGF2) and transforming growth factor β (TGF-β), although NANOG expression is partially suppressed. Further analysis demonstrates that ERK5 promotes the expression of autocrine factors such as NODAL, FGF8, and WNT3. The addition of NODAL protein rescues NANOG expression and differentiation phenotypes under ERK5 inhibition. We demonstrate that constitutively active ERK5 pathway allows self-renewal even without essential growth factors FGF2 and TGF-β. This study highlights the essential contribution of autocrine pathways to proper maintenance and differentiation.
Collapse
Affiliation(s)
- Chengcheng Song
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Zhaoying Zhang
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Dongliang Leng
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Ziqing He
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Xuepeng Wang
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, China; CAM-SU Genomic Resource Center, Soochow University, Suzhou, Jiangsu, China
| | - Weiwei Liu
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Biological Imaging and Stem Cell Core Facility, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Wensheng Zhang
- CAM-SU Genomic Resource Center, Soochow University, Suzhou, Jiangsu, China
| | - Qiang Wu
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, China
| | - Qi Zhao
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Guokai Chen
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Faculty of Health Sciences, University of Macau, Taipa, Macau, China; Zhuhai UM Science & Technology Research Institute, Zhuhai, China.
| |
Collapse
|
3
|
Wu C, Liu H, Zhong D, Yang X, Liao Z, Chen Y, Zhang S, Su D, Zhang B, Li C, Tian L, Xu C, Su P. Mapk7 deletion in chondrocytes causes vertebral defects by reducing MEF2C/PTEN/AKT signaling. Genes Dis 2024; 11:964-977. [PMID: 37692479 PMCID: PMC10491872 DOI: 10.1016/j.gendis.2023.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/17/2023] [Accepted: 02/07/2023] [Indexed: 03/31/2023] Open
Abstract
Mutation of the MAPK7 gene was related to human scoliosis. Mapk7 regulated the development of limb bones and skulls in mice. However, the role of MAPK7 in vertebral development is still unclear. In this study, we constructed Col2a1-cre; Mapk7f/f transgenic mouse model to delete Mapk7 in cartilage, which displayed kyphosis and osteopenia. Mechanistically, Mapk7 loss decreased MEF2C expression and thus activated PTEN to oppose PI3K/AKT signaling in vertebral growth plate chondrocytes, which impaired chondrocyte hypertrophy and attenuated vertebral ossification. In vivo, systemic pharmacological activation of AKT rescued impaired chondrocyte hypertrophy and alleviated mouse vertebral defects caused by Mapk7 deficiency. Our study firstly clarified the mechanism by which MAPK7 was involved in vertebral development, which might contribute to understanding the pathology of spinal deformity and provide a basis for the treatment of developmental disorders of the spine.
Collapse
Affiliation(s)
- Chengzhi Wu
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Hengyu Liu
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Dongmei Zhong
- Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Xiaoming Yang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Zhiheng Liao
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Yuyu Chen
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Shun Zhang
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Deying Su
- Guangdong Provincial Key Laboratory of Proteomics and State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Baolin Zhang
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Chuan Li
- Research Center for Translational Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Liru Tian
- Research Center for Translational Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Caixia Xu
- Research Center for Translational Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Peiqiang Su
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| |
Collapse
|
4
|
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
|
5
|
Pan P, Geng T, Li Z, Ding X, Shi M, Li Y, Wang Y, Shi Y, Wu J, Zhong L, Ji D, Li Z, Meng X. Design, Synthesis, and Biological Evaluation of Proteolysis-Targeting Chimeras as Highly Selective and Efficient Degraders of Extracellular Signal-Regulated Kinase 5. J Med Chem 2023; 66:13568-13586. [PMID: 37751283 DOI: 10.1021/acs.jmedchem.3c00864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Extracellular signal-regulated kinase 5 (ERK5) is recognized as a key member of the mitogen-activated protein kinase family and is involved in tumor growth, migration, and angiogenesis. However, the results of ERK5 inhibition in multiple studies are controversial, and a highly specific ERK5-targeting agent is required to confirm physiological functions. Using proteolysis-targeting chimera technology, we designed the selective ERK5 degrader PPM-3 and examined its biological effect on cancer cells. Interestingly, the selective degradation of ERK5 with PPM-3 did not influence tumor cell growth directly. Based on proteomics analysis, the ERK5 deletion may be associated with tumor immunity. PPM-3 influences tumor development by affecting the differentiation of macrophages. Therefore, PPM-3 is an effective small-molecule tool for studying ERK5 and a promising immunotherapy drug candidate.
Collapse
Affiliation(s)
- Pengming Pan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Tongtong Geng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhongtang Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xuyang Ding
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Mengyuan Shi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yang Li
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yashuai Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yuanyuan Shi
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jiaojiao Wu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Liang Zhong
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Dengbo Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhongjun Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xiangbao Meng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
6
|
Le NT. The significance of ERK5 catalytic-independent functions in disease pathways. Front Cell Dev Biol 2023; 11:1235217. [PMID: 37601096 PMCID: PMC10436230 DOI: 10.3389/fcell.2023.1235217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/26/2023] [Indexed: 08/22/2023] Open
Abstract
Extracellular signal-regulated kinase 5 (ERK5), also known as BMK1 or MAPK7, represents a recent addition to the classical mitogen-activated protein kinase (MAPK) family. This family includes well-known members such as ERK1/2, c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase (p38 MAPK), as well as atypical MAPKs such as ERK3, ERK4, ERK7 (ERK8), and Nemo-like kinase (NLK). Comprehensive reviews available elsewhere provide detailed insights into ERK5, which interested readers can refer to for in-depth knowledge (Nithianandarajah-Jones et al., 2012; Monti et al., Cancers (Basel), 2022, 14). The primary aim of this review is to emphasize the essential characteristics of ERK5 and shed light on the intricate nature of its activation, with particular attention to the catalytic-independent functions in disease pathways.
Collapse
Affiliation(s)
- Nhat-Tu Le
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
7
|
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
|
8
|
Abe JI, Imanishi M, Li S, Zhang A, Ae Ko K, Samanthapudi VSK, Lee LL, Bojorges AP, Gi YJ, Hobbs BP, Deswal A, Herrmann J, Lin SH, Chini EN, Shen YH, Schadler KL, Nguyen THM, Gupte AA, Reyes-Gibby C, Yeung SCJ, Abe RJ, Olmsted-Davis EA, Krishnan S, Dantzer R, Palaskas NL, Cooke JP, Pownall HJ, Yoshimoto M, Fujiwara K, Hamilton DJ, Burks JK, Wang G, Le NT, Kotla S. An ERK5-NRF2 Axis Mediates Senescence-Associated Stemness and Atherosclerosis. Circ Res 2023; 133:25-44. [PMID: 37264926 PMCID: PMC10357365 DOI: 10.1161/circresaha.122.322017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 05/17/2023] [Indexed: 06/03/2023]
Abstract
BACKGROUND ERK5 (extracellular signal-regulated kinase 5) is a dual kinase transcription factor containing an N-terminal kinase domain and a C-terminal transcriptional activation domain. Many ERK5 kinase inhibitors have been developed and tested to treat cancer and inflammatory diseases. However, recent data have raised questions about the role of the catalytic activity of ERK5 in proliferation and inflammation. We aimed to investigate how ERK5 reprograms myeloid cells to the proinflammatory senescent phenotype, subsequently leading to atherosclerosis. METHODS A ERK5 S496A (dephosphorylation mimic) knock in (KI) mouse model was generated using CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat-associated 9), and atherosclerosis was characterized by hypercholesterolemia induction. The plaque phenotyping in homozygous ERK5 S496A KI and wild type (WT) mice was studied using imaging mass cytometry. Bone marrow-derived macrophages were isolated from hypercholesterolemic mice and characterized using RNA sequencing and functional in vitro approaches, including senescence, mitochondria reactive oxygen species, and inflammation assays, as well as by metabolic extracellular flux analysis. RESULTS We show that atherosclerosis was inhibited in ERK5 S496A KI mice. Furthermore, ERK5 S496 phosphorylation mediates both senescence-associated secretory phenotype and senescence-associated stemness by upregulating AHR (aryl hydrocarbon receptor) in plaque and bone marrow-derived macrophages isolated from hypercholesterolemic mice. We also discovered that ERK5 S496 phosphorylation could induce NRF2 (NFE2-related factor 2) SUMOylation at a novel K518 site to inhibit NRF2 transcriptional activity without altering ERK5 catalytic activity and mediates oxidized LDL (low-density lipoprotein)-induced senescence-associated secretory phenotype. Specific ERK5 kinase inhibitors (AX15836 and XMD8-92) also inhibited ERK5 S496 phosphorylation, suggesting the involvement of ERK5 S496 phosphorylation in the anti-inflammatory effects of these ERK5 kinase inhibitors. CONCLUSIONS We discovered a novel mechanism by which the macrophage ERK5-NRF2 axis develops a unique senescence-associated secretory phenotype/stemness phenotype by upregulating AHR to engender atherogenesis. The finding of senescence-associated stemness phenotype provides a molecular explanation to resolve the paradox of senescence in proliferative plaque by permitting myeloid cells to escape the senescence-induced cell cycle arrest during atherosclerosis formation.
Collapse
Affiliation(s)
- Jun-ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- These authors contributed equally to this work and were designated as co-first authors
| | - Masaki Imanishi
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- These authors contributed equally to this work and were designated as co-first authors
| | - Shengyu Li
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
- These authors contributed equally to this work and were designated as co-first authors
| | - Aijun Zhang
- Center for Bioenergetics, Houston Methodist Research Institute, Texas, and Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, Houston, Texas, USA
| | - Kyung Ae Ko
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Ling-Ling Lee
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Young Jin Gi
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Brian P. Hobbs
- Department of Population Health, The University of Texas at Austin, Austin, Texas, USA
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Joerg Herrmann
- Cardio Oncology Clinic, Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Steven H. Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Eduardo N. Chini
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Ying H. Shen
- Division of Cardiothoracic Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Keri L. Schadler
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Thi-Hong-Minh Nguyen
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Anisha A. Gupte
- Center for Bioenergetics, Houston Methodist Research Institute, Texas, and Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, Houston, Texas, USA
| | - Cielito Reyes-Gibby
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sai-Ching J. Yeung
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rei J. Abe
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | | | - Sunil Krishnan
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Robert Dantzer
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nicolas L. Palaskas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - John P. Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Henry J. Pownall
- Center for Bioenergetics, Houston Methodist Research Institute, Texas, and Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, Houston, Texas, USA
| | - Momoko Yoshimoto
- Center for Stem Cell & Regenerative Medicine, Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Keigi Fujiwara
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dale J. Hamilton
- Center for Bioenergetics, Houston Methodist Research Institute, Texas, and Department of Medicine, Houston Methodist, Weill Cornell Medicine Affiliate, Houston, Texas, USA
- These authors contributed equally to this work
| | - Jared K. Burks
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- These authors contributed equally to this work
| | - Guangyu Wang
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
- These authors were equivalent co-senior authors
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
- These authors were equivalent co-senior authors
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- These authors were equivalent co-senior authors
| |
Collapse
|
9
|
Im JY, Kang MJ, Kim BK, Won M. DDIAS, DNA damage-induced apoptosis suppressor, is a potential therapeutic target in cancer. Exp Mol Med 2023:10.1038/s12276-023-00974-6. [PMID: 37121974 DOI: 10.1038/s12276-023-00974-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/11/2023] [Accepted: 01/19/2023] [Indexed: 05/02/2023] Open
Abstract
Increasing evidence indicates that DNA damage-induced apoptosis suppressor (DDIAS) is an oncogenic protein that is highly expressed in a variety of cancers, including colorectal cancer, lung cancer, breast cancer, and hepatocellular carcinoma (HCC). The discovery of DDIAS as a novel therapeutic target and its role in human cancer biology is fascinating and noteworthy. Recent studies have shown that DDIAS is involved in tumorigenesis, metastasis, DNA repair and synthesis, and drug resistance and that it plays multiple roles with distinct binding partners in several human cancers. This review focuses on the function of DDIAS and its regulatory proteins in human cancer as potential targets for cancer therapy, as well as the development and future prospects of DDIAS inhibitors.
Collapse
Affiliation(s)
- Joo-Young Im
- Personalized Genomic Medicine Research Center, KRIBB, Daejeon, 34141, Republic of Korea.
| | - Mi-Jung Kang
- Personalized Genomic Medicine Research Center, KRIBB, Daejeon, 34141, Republic of Korea
| | - Bo-Kyung Kim
- Personalized Genomic Medicine Research Center, KRIBB, Daejeon, 34141, Republic of Korea
- University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
- R&D Center, OneCureGEN Co., Ltd., Daejeon, 34141, Republic of Korea
| | - Misun Won
- Personalized Genomic Medicine Research Center, KRIBB, Daejeon, 34141, Republic of Korea.
- University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
- R&D Center, OneCureGEN Co., Ltd., Daejeon, 34141, Republic of Korea.
| |
Collapse
|
10
|
Li T, Conroy KL, Kim AM, Halmai J, Gao K, Moreno E, Wang A, Passerini AG, Nolta JA, Zhou P. Role of MEF2C in the Endothelial Cells Derived from Human Induced Pluripotent Stem Cells. Stem Cells 2023; 41:341-353. [PMID: 36639926 PMCID: PMC10128960 DOI: 10.1093/stmcls/sxad005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 01/08/2023] [Indexed: 01/15/2023]
Abstract
Human induced pluripotent stem cells (hiPSCs) not only provide an abundant source of vascular cells for potential therapeutic applications in vascular disease but also constitute an excellent model for understanding the mechanisms that regulate the differentiation and the functionality of vascular cells. Here, we reported that myocyte enhancer factor 2C (MEF2C) transcription factor, but not any other members of the MEF2 family, was robustly upregulated during the differentiation of vascular progenitors and endothelial cells (ECs) from hiPSCs. Vascular endothelial growth factors (VEGF) strongly induced MEF2C expression in endothelial lineage cells. The specific upregulation of MEF2C during the commitment of endothelial lineage was dependent on the extracellular signal regulated kinase (ERK). Moreover, knockdown of MEF2C with shRNA in hiPSCs did not affect the differentiation of ECs from these hiPSCs, but greatly reduced the migration and tube formation capacity of the hiPSC-derived ECs. Through a chromatin immunoprecipitation-sequencing, genome-wide RNA-sequencing, quantitative RT-PCR, and immunostaining analyses of the hiPSC-derived endothelial lineage cells with MEF2C inhibition or knockdown compared to control hiPSC-derived ECs, we identified TNF-related apoptosis inducing ligand (TRAIL) and transmembrane protein 100 (TMEM100) as novel targets of MEF2C. This study demonstrates an important role for MEF2C in regulating human EC functions and highlights MEF2C and its downstream effectors as potential targets to treat vascular malfunction-associated diseases.
Collapse
Affiliation(s)
- Tao Li
- School of Medicine, Hunan Normal University, Changsha, Hunan, People’s Republic of China
- Stem Cell Program and Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA, USA
| | - Kelsey L Conroy
- Stem Cell Program and Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA, USA
| | - Amy M Kim
- Stem Cell Program and Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA, USA
| | - Julian Halmai
- Stem Cell Program and Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA, USA
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
- University of California Davis Gene Therapy Center, Sacramento, CA, USA
| | - Kewa Gao
- Department of Surgery, University of California Davis, Sacramento, CA, USA
| | - Emily Moreno
- Department of Biomedical Engineering, University of California Davis, Davis, CA, USA
| | - Aijun Wang
- Department of Surgery, University of California Davis, Sacramento, CA, USA
- Department of Biomedical Engineering, University of California Davis, Davis, CA, USA
| | - Anthony G Passerini
- Department of Biomedical Engineering, University of California Davis, Davis, CA, USA
| | - Jan A Nolta
- Stem Cell Program and Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA, USA
- University of California Davis Gene Therapy Center, Sacramento, CA, USA
| | - Ping Zhou
- Stem Cell Program and Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA, USA
- University of California Davis Gene Therapy Center, Sacramento, CA, USA
| |
Collapse
|
11
|
Tusa I, Menconi A, Tubita A, Rovida E. Pathophysiological Impact of the MEK5/ERK5 Pathway in Oxidative Stress. Cells 2023; 12:cells12081154. [PMID: 37190064 DOI: 10.3390/cells12081154] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/22/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Oxidative stress regulates many physiological and pathological processes. Indeed, a low increase in the basal level of reactive oxygen species (ROS) is essential for various cellular functions, including signal transduction, gene expression, cell survival or death, as well as antioxidant capacity. However, if the amount of generated ROS overcomes the antioxidant capacity, excessive ROS results in cellular dysfunctions as a consequence of damage to cellular components, including DNA, lipids and proteins, and may eventually lead to cell death or carcinogenesis. Both in vitro and in vivo investigations have shown that activation of the mitogen-activated protein kinase kinase 5/extracellular signal-regulated kinase 5 (MEK5/ERK5) pathway is frequently involved in oxidative stress-elicited effects. In particular, accumulating evidence identified a prominent role of this pathway in the anti-oxidative response. In this respect, activation of krüppel-like factor 2/4 and nuclear factor erythroid 2-related factor 2 emerged among the most frequent events in ERK5-mediated response to oxidative stress. This review summarizes what is known about the role of the MEK5/ERK5 pathway in the response to oxidative stress in pathophysiological contexts within the cardiovascular, respiratory, lymphohematopoietic, urinary and central nervous systems. The possible beneficial or detrimental effects exerted by the MEK5/ERK5 pathway in the above systems are also discussed.
Collapse
Affiliation(s)
- Ignazia Tusa
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Alessio Menconi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Alessandro Tubita
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| |
Collapse
|
12
|
Moustafa A, Hashemi S, Brar G, Grigull J, Ng SHS, Williams D, Schmitt-Ulms G, McDermott JC. The MEF2A transcription factor interactome in cardiomyocytes. Cell Death Dis 2023; 14:240. [PMID: 37019881 PMCID: PMC10076289 DOI: 10.1038/s41419-023-05665-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 12/02/2022] [Accepted: 02/08/2023] [Indexed: 04/07/2023]
Abstract
Transcriptional regulators encoded by the Myocyte Enhancer Factor 2 (MEF2) gene family play a fundamental role in cardiac development, homeostasis and pathology. Previous studies indicate that MEF2A protein-protein interactions serve as a network hub in several cardiomyocyte cellular processes. Based on the idea that interactions with regulatory protein partners underly the diverse roles of MEF2A in cardiomyocyte gene expression, we undertook a systematic unbiased screen of the MEF2A protein interactome in primary cardiomyocytes using an affinity purification-based quantitative mass spectrometry approach. Bioinformatic processing of the MEF2A interactome revealed protein networks involved in the regulation of programmed cell death, inflammatory responses, actin dynamics and stress signaling in primary cardiomyocytes. Further biochemical and functional confirmation of specific protein-protein interactions documented a dynamic interaction between MEF2A and STAT3 proteins. Integration of transcriptome level data from MEF2A and STAT3-depleted cardiomyocytes reveals that the balance between MEF2A and STAT3 activity exerts a level of executive control over the inflammatory response and cardiomyocyte cell survival and experimentally ameliorates Phenylephrine induced cardiomyocyte hypertrophy. Lastly, we identified several MEF2A/STAT3 co-regulated genes, including the MMP9 gene. Herein, we document the cardiomyocyte MEF2A interactome, which furthers our understanding of protein networks involved in the hierarchical control of normal and pathophysiological cardiomyocyte gene expression in the mammalian heart.
Collapse
Affiliation(s)
- Amira Moustafa
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada
| | - Sara Hashemi
- Analytical Sciences, Sanofi, Toronto, ON, M2R 3T4, Canada
- Seneca College, School of Health Sciences, King City, ON, L7B 1B3, Canada
| | - Gurnoor Brar
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada
| | - Jörg Grigull
- Department of Mathematics and Statistics, York University, Toronto, ON, M3J1P3, Canada
| | - Siemon H S Ng
- Analytical Sciences, Sanofi, Toronto, ON, M2R 3T4, Canada
- Analytical Development, Notch Therapeutics, Toronto, ON, M5G 1M1, Canada
| | - Declan Williams
- Tanz Centre for Research in Neurodegenerative Diseases, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5T 0S8, Canada
| | - Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5T 0S8, Canada
| | - John C McDermott
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada.
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada.
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada.
| |
Collapse
|
13
|
Martini AG, Smith JP, Medrano S, Sheffield NC, Sequeira-Lopez MLS, Gomez RA. Determinants of renin cell differentiation: a single cell epi-transcriptomics approach. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.18.524595. [PMID: 36711565 PMCID: PMC9882312 DOI: 10.1101/2023.01.18.524595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Rationale Renin cells are essential for survival. They control the morphogenesis of the kidney arterioles, and the composition and volume of our extracellular fluid, arterial blood pressure, tissue perfusion, and oxygen delivery. It is known that renin cells and associated arteriolar cells descend from FoxD1 + progenitor cells, yet renin cells remain challenging to study due in no small part to their rarity within the kidney. As such, the molecular mechanisms underlying the differentiation and maintenance of these cells remain insufficiently understood. Objective We sought to comprehensively evaluate the chromatin states and transcription factors (TFs) that drive the differentiation of FoxD1 + progenitor cells into those that compose the kidney vasculature with a focus on renin cells. Methods and Results We isolated single nuclei of FoxD1 + progenitor cells and their descendants from FoxD1 cre/+ ; R26R-mTmG mice at embryonic day 12 (E12) (n cells =1234), embryonic day 18 (E18) (n cells =3696), postnatal day 5 (P5) (n cells =1986), and postnatal day 30 (P30) (n cells =1196). Using integrated scRNA-seq and scATAC-seq we established the developmental trajectory that leads to the mosaic of cells that compose the kidney arterioles, and specifically identified the factors that determine the elusive, myo-endocrine adult renin-secreting juxtaglomerular (JG) cell. We confirm the role of Nfix in JG cell development and renin expression, and identified the myocyte enhancer factor-2 (MEF2) family of TFs as putative drivers of JG cell differentiation. Conclusions We provide the first developmental trajectory of renin cell differentiation as they become JG cells in a single-cell atlas of kidney vascular open chromatin and highlighted novel factors important for their stage-specific differentiation. This improved understanding of the regulatory landscape of renin expressing JG cells is necessary to better learn the control and function of this rare cell population as overactivation or aberrant activity of the RAS is a key factor in cardiovascular and kidney pathologies.
Collapse
|
14
|
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
|
15
|
The ERK5/NF-κB signaling pathway targets endometrial cancer proliferation and survival. Cell Mol Life Sci 2022; 79:524. [PMID: 36123565 PMCID: PMC9485191 DOI: 10.1007/s00018-022-04541-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/20/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022]
Abstract
Endometrial cancer (EC) is the most common type of gynecologic cancer in women of developed countries. Despite surgery combined with chemo-/radiotherapy regimens, overall survival of patients with high-risk EC tumors is poor, indicating a need for novel therapies. The MEK5-ERK5 pathway is activated in response to growth factors and to different stressors, including oxidative stress and cytokines. Previous evidence supports a role for the MEK5-ERK5 pathway in the pathology of several cancers. We investigated the role of ERK5 in EC. In silico analysis of the PanCancer Atlas dataset showed alterations in components of the MEK5-ERK5 pathway in 48% of EC patients. Here, we show that ERK5 inhibition or silencing decreased EGF-induced EC cell proliferation, and that genetic deletion of MEK5 resulted in EC impaired proliferation and reduced tumor growth capacity in nude mice. Pharmacologic inhibition or ERK5 silencing impaired NF-kB pathway in EC cells and xenografts. Furthermore, we found a positive correlation between ERK5 and p65/RELA protein levels in human EC tumor samples. Mechanistically, genetic or pharmacologic impairment of ERK5 resulted in downregulation of NEMO/IKKγ expression, leading to impaired p65/RELA activity and to apoptosis in EC cells and xenografts, which was rescued by NEMO/IKKγ overexpression. Notably, ERK5 inhibition, MEK5 deletion or NF-kB inhibition sensitized EC cells to standard EC chemotherapy (paclitaxel/carboplatin) toxicity, whereas ERK5 inhibition synergized with paclitaxel to reduce tumor xenograft growth in mice. Together, our results suggest that the ERK5-NEMO-NF-κB pathway mediates EC cell proliferation and survival. We propose the ERK5/NF-κB axis as new target for EC treatment.
Collapse
|
16
|
Li R, Shu M, Liu X, Nei Z, Ye B, Wang H, Gong Y. Genome-wide identification of mitogen-activated protein kinase (MAPK) gene family in yellow catfish (Pelteobagrus fulviadraco) and their expression profiling under the challenge of Aeromonas hydrophila. JOURNAL OF FISH BIOLOGY 2022; 101:699-710. [PMID: 35751135 DOI: 10.1111/jfb.15141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/19/2022] [Indexed: 06/15/2023]
Abstract
As serine/threonine protein kinases, mitogen-activated protein kinases (MAPK) take part in cellular metabolism. This work found 14 MAPK genes in the yellow catfish (Pelteobagrus fulviadraco) genome and evaluated their taxonomy, conserved domains and evolutionary linkages for a better understanding of the MAPK gene family's evolutionary relationship and antibacterial immune response. The findings revealed that several MAPK genes are activated in response to immunological and inflammatory responses. Collinearity research revealed that in yellow catfish and zebrafish, there are six pairs of highly similar MAPK genes, indicating that these genes have been more conserved throughout evolution. The MAPK gene quantification findings revealed that JNK1a, JNK1b, p38delta and p38alpha b expression levels were considerably upregulated, indicating that they act in fish innate immunity. The findings implied that MAPK genes may involve in defence against detrimental microbe in yellow catfish, which will help researchers better understand how MAPK genes work in the innate immune system.
Collapse
Affiliation(s)
- Ronghui Li
- Key Laboratory of Marine Biotechnology of Zhejiang Province, School of Marine Sciences, Ningbo University, Ningbo, China
| | - Mingyu Shu
- Key Laboratory of Marine Biotechnology of Zhejiang Province, School of Marine Sciences, Ningbo University, Ningbo, China
| | - Xuanxuan Liu
- Key Laboratory of Marine Biotechnology of Zhejiang Province, School of Marine Sciences, Ningbo University, Ningbo, China
| | - Zhiwei Nei
- Key Laboratory of Marine Biotechnology of Zhejiang Province, School of Marine Sciences, Ningbo University, Ningbo, China
| | - Ben Ye
- Key Laboratory of Marine Biotechnology of Zhejiang Province, School of Marine Sciences, Ningbo University, Ningbo, China
| | - Heyu Wang
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, China
| | - Yifu Gong
- Key Laboratory of Marine Biotechnology of Zhejiang Province, School of Marine Sciences, Ningbo University, Ningbo, China
| |
Collapse
|
17
|
Effect of Extracellular Signal-Regulated Protein Kinase 5 Inhibition in Clear Cell Renal Cell Carcinoma. Int J Mol Sci 2022; 23:ijms23158448. [PMID: 35955582 PMCID: PMC9369143 DOI: 10.3390/ijms23158448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 12/10/2022] Open
Abstract
(1) Background: Extracellular signal-regulating kinase 5 (ERK5) has been implicated in many cellular functions, including survival, proliferation, and vascularization. Our objectives were to examine the expression and effect of ERK5 in clear cell renal cell carcinoma (ccRCC). (2) Methods: The expressions of ERK5 and its regulating micro-RNA miR-143 were investigated using immunohistochemistry and quantitative reverse transcriptase PCR in surgical specimens of ccRCC patients. With invitro and in vivo studies, we used pharmacologic ERK5 inhibitor XMD8-92, RNA interference, pre-miR-143 transduction, Western blotting, MTS assay, apoptosis assay, and subcutaneous xenograft model. (3) Results: A strong ERK5 expression in surgical specimen was associated with high-grade (p = 0.01), high-recurrence free rate (p = 0.02), and high cancer-specific survival (p = 0.03). Expression levels of ERK5 and miR-143 expression level were correlated (p = 0.049). Pre-miR-143 transduction into ccRCC cell A498 suppressed ERK5 expression. ERK5 inhibition enhanced cyclin-dependent kinase inhibitor p21 expression and decreased anti-apoptotic molecules BCL2, resulting in decreased cell proliferation and survival both in ccRCC and endothelial cells. In the xenograft model, ERK5 inhibitor XMD8-92 suppressed tumor growth. (4) Conclusions: ERK5 is regulated by miR-143, and ERK5 inhibition is a promising target for ccRCC treatment.
Collapse
|
18
|
Shu C, Hou L, Chen Q, Zhu T, Yang J, Luo X, Su Y, Wang Y. Irradiation with a red light-emitting diode enhances the proliferation of stem cells of apical papilla via the ERK5 signalling pathway. Lasers Med Sci 2022; 37:2259-2268. [PMID: 35022873 DOI: 10.1007/s10103-021-03492-x] [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: 07/20/2021] [Accepted: 12/06/2021] [Indexed: 11/27/2022]
Abstract
This Querystudy aimed to investigate the effects of low-energy red light-emitting diode (LED) irradiation on the proliferation of stem cells from apical papilla (SCAPs) and preliminarily elucidated the underlying molecular mechanisms. SCAPs were isolated and identified in vitro. The light source was a 10 W red LED with continuous output and a wavelength of 600-700 nm. SCAPs were irradiated with 0 (control group), 0.5 J/cm2, 1 J/cm2, 3 J/cm2, or 5 J/cm2. Cell Counting Kit-8 (CCK-8) assays were used to analyze cell proliferation rates and determine the most effective concentration of extracellular signal-regulated kinase 5 (ERK5) blocker, BIX02189. A real-time polymerase chain reaction (RT-PCR) was carried out to determine the involvement of the ERK5 signalling pathway and proliferation-associated genes (C-Jun, Jun B, and Cyclin D1). 5-Ethynyl-2'-deoxyuridine (EDU) was used to analyze cell cycle kinetic parameters. CCK-8 assay results suggested that SCAPs in red LED groups exhibited a higher proliferation rate than those in the control group, and 10 μmol/L BIX02189 was the most effective blocker. The RT-PCR results demonstrate that red LEDs upregulated the expression of the ERK5, C-Jun, Jun B, and Cyclin D1 genes, and BIX02189 successfully blocked the ERK5 signalling pathway. The results of EdU staining indicated that red LED promoted DNA synthesis activity and that BIX02189 suppressed cells into S phase. Red LEDs irradiation enhances the proliferation of SCAPs via the ERK5 signalling pathway by upregulating the expression of C-Jun, Jun B, and Cyclin D1.
Collapse
Affiliation(s)
- Chunxia Shu
- School of Stomatology Southwest Medical University, Lu Zhou, 646000, China
- Suining First People's Hospital, Suining, 629000, China
| | - Lan Hou
- School of Stomatology Southwest Medical University, Lu Zhou, 646000, China
| | - Qiang Chen
- The TCM Hospital of Longquanyi District, Chengdu, 610100, China
| | - Tingting Zhu
- School of Stomatology of Qingdao University, Qingdao, 266003, China
| | - Juan Yang
- School of Stomatology Southwest Medical University, Lu Zhou, 646000, China
| | - Xiang Luo
- School of Stomatology Southwest Medical University, Lu Zhou, 646000, China
| | - Yutong Su
- School of Stomatology Southwest Medical University, Lu Zhou, 646000, China
| | - Yao Wang
- The Affiliated Stomatology Hospital of Southwest Medical University, Lu Zhou, 646000, China.
| |
Collapse
|
19
|
Glucose Starvation or Pyruvate Dehydrogenase Activation Induce a Broad, ERK5-Mediated, Metabolic Remodeling Leading to Fatty Acid Oxidation. Cells 2022; 11:cells11091392. [PMID: 35563698 PMCID: PMC9104157 DOI: 10.3390/cells11091392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/05/2022] [Accepted: 04/14/2022] [Indexed: 12/24/2022] Open
Abstract
Cells have metabolic flexibility that allows them to adapt to changes in substrate availability. Two highly relevant metabolites are glucose and fatty acids (FA), and hence, glycolysis and fatty acid oxidation (FAO) are key metabolic pathways leading to energy production. Both pathways affect each other, and in the absence of one substrate, metabolic flexibility allows cells to maintain sufficient energy production. Here, we show that glucose starvation or sustained pyruvate dehydrogenase (PDH) activation by dichloroacetate (DCA) induce large genetic remodeling to propel FAO. The extracellular signal-regulated kinase 5 (ERK5) is a key effector of this multistep metabolic remodeling. First, there is an increase in the lipid transport by expression of low-density lipoprotein receptor-related proteins (LRP), e.g., CD36, LRP1 and others. Second, an increase in the expression of members of the acyl-CoA synthetase long-chain (ACSL) family activates FA. Finally, the expression of the enzymes that catalyze the initial step in each cycle of FAO, i.e., the acyl-CoA dehydrogenases (ACADs), is induced. All of these pathways lead to enhanced cellular FAO. In summary, we show here that different families of enzymes, which are essential to perform FAO, are regulated by the signaling pathway, i.e., MEK5/ERK5, which transduces changes from the environment to genetic adaptations.
Collapse
|
20
|
Clinical Significance and Regulation of ERK5 Expression and Function in Cancer. Cancers (Basel) 2022; 14:cancers14020348. [PMID: 35053510 PMCID: PMC8773716 DOI: 10.3390/cancers14020348] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/08/2022] [Accepted: 01/08/2022] [Indexed: 02/06/2023] Open
Abstract
Extracellular signal-regulated kinase 5 (ERK5) is a unique kinase among MAPKs family members, given its large structure characterized by the presence of a unique C-terminal domain. Despite increasing data demonstrating the relevance of the ERK5 pathway in the growth, survival, and differentiation of normal cells, ERK5 has recently attracted the attention of several research groups given its relevance in inflammatory disorders and cancer. Accumulating evidence reported its role in tumor initiation and progression. In this review, we explore the gene expression profile of ERK5 among cancers correlated with its clinical impact, as well as the prognostic value of ERK5 and pERK5 expression levels in tumors. We also summarize the importance of ERK5 in the maintenance of a cancer stem-like phenotype and explore the major known contributions of ERK5 in the tumor-associated microenvironment. Moreover, although several questions are still open concerning ERK5 molecular regulation, different ERK5 isoforms derived from the alternative splicing process are also described, highlighting the potential clinical relevance of targeting ERK5 pathways.
Collapse
|
21
|
An ERK5-KLF2 signalling module regulates early embryonic gene expression and telomere rejuvenation in stem cells. Biochem J 2021; 478:4119-4136. [PMID: 34780645 PMCID: PMC8718266 DOI: 10.1042/bcj20210646] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/08/2021] [Accepted: 11/15/2021] [Indexed: 12/15/2022]
Abstract
The ERK5 MAP kinase signalling pathway drives transcription of naïve pluripotency genes in mouse Embryonic Stem Cells (mESCs). However, how ERK5 impacts on other aspects of mESC biology has not been investigated. Here, we employ quantitative proteomic profiling to identify proteins whose expression is regulated by the ERK5 pathway in mESCs. This reveals a function for ERK5 signalling in regulating dynamically expressed early embryonic 2-cell stage (2C) genes including the mESC rejuvenation factor ZSCAN4. ERK5 signalling and ZSCAN4 induction in mESCs increases telomere length, a key rejuvenative process required for prolonged culture. Mechanistically, ERK5 promotes ZSCAN4 and 2C gene expression via transcription of the KLF2 pluripotency transcription factor. Surprisingly, ERK5 also directly phosphorylates KLF2 to drive ubiquitin-dependent degradation, encoding negative feedback regulation of 2C gene expression. In summary, our data identify a regulatory module whereby ERK5 kinase and transcriptional activities bi-directionally control KLF2 levels to pattern 2C gene transcription and a key mESC rejuvenation process.
Collapse
|
22
|
Gámez-García A, Bolinaga-Ayala I, Yoldi G, Espinosa-Gil S, Diéguez-Martínez N, Megías-Roda E, Muñoz-Guardiola P, Lizcano JM. ERK5 Inhibition Induces Autophagy-Mediated Cancer Cell Death by Activating ER Stress. Front Cell Dev Biol 2021; 9:742049. [PMID: 34805151 PMCID: PMC8600073 DOI: 10.3389/fcell.2021.742049] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/19/2021] [Indexed: 01/18/2023] Open
Abstract
Autophagy is a highly conserved intracellular process that preserves cellular homeostasis by mediating the lysosomal degradation of virtually any component of the cytoplasm. Autophagy is a key instrument of cellular response to several stresses, including endoplasmic reticulum (ER) stress. Cancer cells have developed high dependency on autophagy to overcome the hostile tumor microenvironment. Thus, pharmacological activation or inhibition of autophagy is emerging as a novel antitumor strategy. ERK5 is a novel member of the MAP kinase family that is activated in response to growth factors and different forms of stress. Recent work has pointed ERK5 as a major player controlling cancer cell proliferation and survival. Therefore small-molecule inhibitors of ERK5 have shown promising therapeutic potential in different cancer models. Here, we report for the first time ERK5 as a negative regulator of autophagy. Thus, ERK5 inhibition or silencing induced autophagy in a panel of human cancer cell lines with different mutation patterns. As reported previously, ERK5 inhibitors (ERK5i) induced apoptotic cancer cell death. Importantly, we found that autophagy mediates the cytotoxic effect of ERK5i, since ATG5ˉ/ˉ autophagy-deficient cells viability was not affected by these compounds. Mechanistically, ERK5i stimulated autophagic flux independently of the canonical regulators AMPK or mTORC1. Moreover, ERK5 inhibition resulted in ER stress and activation of the Unfolded Protein Response (UPR) pathways. Specifically, ERK5i induced expression of the ER luminal chaperone BiP (a hallmark of ER stress), the UPR markers CHOP and ATF4, and the spliced form of XBP1. Pharmacological inhibition of UPR with chemical chaperone TUDC, or ATF4 silencing, resulted in impaired ERK5i-mediated UPR, autophagy and cytotoxicity. Overall, our results suggest that ERK5 inhibition induces autophagy-mediated cancer cell death by activating ER stress. Since ERK5 inhibition sensitizes cancer cells and tumors to chemotherapy, future work will determine the relevance of UPR and autophagy in the combined use of chemotherapy and ERK5i to tackle Cancer.
Collapse
Affiliation(s)
- Andrés Gámez-García
- Departament de Bioquímica i Biologia Molecular and Institut de Neurociències, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Idoia Bolinaga-Ayala
- Departament de Bioquímica i Biologia Molecular and Institut de Neurociències, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Protein Kinases in Cancer Research, Vall Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Guillermo Yoldi
- Departament de Bioquímica i Biologia Molecular and Institut de Neurociències, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Sergio Espinosa-Gil
- Departament de Bioquímica i Biologia Molecular and Institut de Neurociències, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Protein Kinases in Cancer Research, Vall Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Nora Diéguez-Martínez
- Departament de Bioquímica i Biologia Molecular and Institut de Neurociències, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Protein Kinases in Cancer Research, Vall Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Elisabet Megías-Roda
- Departament de Bioquímica i Biologia Molecular and Institut de Neurociències, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Protein Kinases in Cancer Research, Vall Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Pau Muñoz-Guardiola
- Departament de Bioquímica i Biologia Molecular and Institut de Neurociències, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Jose M Lizcano
- Departament de Bioquímica i Biologia Molecular and Institut de Neurociències, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.,Protein Kinases in Cancer Research, Vall Hebron Institut de Recerca (VHIR), Barcelona, Spain
| |
Collapse
|
23
|
Engineering digitizer circuits for chemical and genetic screens in human cells. Nat Commun 2021; 12:6150. [PMID: 34686672 PMCID: PMC8536748 DOI: 10.1038/s41467-021-26359-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/30/2021] [Indexed: 12/26/2022] Open
Abstract
Cell-based transcriptional reporters are invaluable in high-throughput compound and CRISPR screens for identifying compounds or genes that can impact a pathway of interest. However, many transcriptional reporters have weak activities and transient responses. This can result in overlooking therapeutic targets and compounds that are difficult to detect, necessitating the resource-consuming process of running multiple screens at various timepoints. Here, we present RADAR, a digitizer circuit for amplifying reporter activity and retaining memory of pathway activation. Reporting on the AP-1 pathway, our circuit identifies compounds with known activity against PKC-related pathways and shows an enhanced dynamic range with improved sensitivity compared to a classical reporter in compound screens. In the first genome-wide pooled CRISPR screen for the AP-1 pathway, RADAR identifies canonical genes from the MAPK and PKC pathways, as well as non-canonical regulators. Thus, our scalable system highlights the benefit and versatility of using genetic circuits in large-scale cell-based screening.
Collapse
|
24
|
Tusa I, Gagliardi S, Tubita A, Pandolfi S, Menconi A, Lulli M, Dello Sbarba P, Stecca B, Rovida E. The Hedgehog-GLI Pathway Regulates MEK5-ERK5 Expression and Activation in Melanoma Cells. Int J Mol Sci 2021; 22:11259. [PMID: 34681917 PMCID: PMC8538987 DOI: 10.3390/ijms222011259] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 12/28/2022] Open
Abstract
Malignant melanoma is the deadliest skin cancer, with a poor prognosis in advanced stages. We recently showed that the extracellular signal-regulated kinase 5 (ERK5), encoded by the MAPK7 gene, plays a pivotal role in melanoma by regulating cell functions necessary for tumour development, such as proliferation. Hedgehog-GLI signalling is constitutively active in melanoma and is required for proliferation. However, no data are available in literature about a possible interplay between Hedgehog-GLI and ERK5 pathways. Here, we show that hyperactivation of the Hedgehog-GLI pathway by genetic inhibition of the negative regulator Patched 1 increases the amount of ERK5 mRNA and protein. Chromatin immunoprecipitation showed that GLI1, the major downstream effector of Hedgehog-GLI signalling, binds to a functional non-canonical GLI consensus sequence at the MAPK7 promoter. Furthermore, we found that ERK5 is required for Hedgehog-GLI-dependent melanoma cell proliferation, and that the combination of GLI and ERK5 inhibitors is more effective than single treatments in reducing cell viability and colony formation ability in melanoma cells. Together, these findings led to the identification of a novel Hedgehog-GLI-ERK5 axis that regulates melanoma cell growth, and shed light on new functions of ERK5, paving the way for new therapeutic options in melanoma and other neoplasms with active Hedgehog-GLI and ERK5 pathways.
Collapse
Affiliation(s)
- Ignazia Tusa
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy; (I.T.); (A.T.); (A.M.); (M.L.); (P.D.S.)
| | - Sinforosa Gagliardi
- Core Research Laboratory-Institute for Cancer Research and Prevention (ISPRO), 50134 Florence, Italy; (S.G.); (S.P.)
| | - Alessandro Tubita
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy; (I.T.); (A.T.); (A.M.); (M.L.); (P.D.S.)
| | - Silvia Pandolfi
- Core Research Laboratory-Institute for Cancer Research and Prevention (ISPRO), 50134 Florence, Italy; (S.G.); (S.P.)
| | - Alessio Menconi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy; (I.T.); (A.T.); (A.M.); (M.L.); (P.D.S.)
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy; (I.T.); (A.T.); (A.M.); (M.L.); (P.D.S.)
| | - Persio Dello Sbarba
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy; (I.T.); (A.T.); (A.M.); (M.L.); (P.D.S.)
| | - Barbara Stecca
- Core Research Laboratory-Institute for Cancer Research and Prevention (ISPRO), 50134 Florence, Italy; (S.G.); (S.P.)
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy; (I.T.); (A.T.); (A.M.); (M.L.); (P.D.S.)
| |
Collapse
|
25
|
Ortega-Muelas M, Roche O, Fernández-Aroca DM, Encinar JA, Albandea-Rodríguez D, Arconada-Luque E, Pascual-Serra R, Muñoz I, Sánchez-Pérez I, Belandia B, Ruiz-Hidalgo MJ, Sánchez-Prieto R. ERK5 signalling pathway is a novel target of sorafenib: Implication in EGF biology. J Cell Mol Med 2021; 25:10591-10603. [PMID: 34655447 PMCID: PMC8581332 DOI: 10.1111/jcmm.16990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 09/10/2021] [Accepted: 09/30/2021] [Indexed: 12/16/2022] Open
Abstract
Sorafenib is a multikinase inhibitor widely used in cancer therapy with an antitumour effect related to biological processes as proliferation, migration or invasion, among others. Initially designed as a Raf inhibitor, Sorafenib was later shown to also block key molecules in tumour progression such as VEGFR and PDGFR. In addition, sorafenib has been connected with key signalling pathways in cancer such as EGFR/EGF. However, no definitive clue about the molecular mechanism linking sorafenib and EGF signalling pathway has been established so far. Our data in HeLa, U2OS, A549 and HEK293T cells, based on in silico, chemical and genetic approaches demonstrate that the MEK5/ERK5 signalling pathway is a novel target of sorafenib. In addition, our data show how sorafenib is able to block MEK5-dependent phosphorylation of ERK5 in the Ser218/Tyr220, affecting the transcriptional activation associated with ERK5. Moreover, we demonstrate that some of the effects of this kinase inhibitor onto EGF biological responses, such as progression through cell cycle or migration, are mediated through the effect exerted onto ERK5 signalling pathway. Therefore, our observations describe a novel target of sorafenib, the ERK5 signalling pathway, and establish new mechanistic insights for the antitumour effect of this multikinase inhibitor.
Collapse
Affiliation(s)
- Marta Ortega-Muelas
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas Universidad de Castilla-La Mancha, Unidad Asociada de Biomedicina UCLM, Unidad asociada al CSIC, Albacete, Spain
| | - Olga Roche
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas Universidad de Castilla-La Mancha, Unidad Asociada de Biomedicina UCLM, Unidad asociada al CSIC, Albacete, Spain.,Departamento de Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Diego M Fernández-Aroca
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas Universidad de Castilla-La Mancha, Unidad Asociada de Biomedicina UCLM, Unidad asociada al CSIC, Albacete, Spain
| | - José A Encinar
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología de Elche (IDiBE) e Instituto de Biología Molecular y Celular (IBMC), Universidad Miguel Hernández (UMH), Elche, Spain
| | - David Albandea-Rodríguez
- Departamento de Biología del Cáncer, Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), Unidad asociada de Biomedicina UCLM, Unidad asociada al CSIC, Madrid, Spain
| | - Elena Arconada-Luque
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas Universidad de Castilla-La Mancha, Unidad Asociada de Biomedicina UCLM, Unidad asociada al CSIC, Albacete, Spain
| | - Raquel Pascual-Serra
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas Universidad de Castilla-La Mancha, Unidad Asociada de Biomedicina UCLM, Unidad asociada al CSIC, Albacete, Spain
| | - Ismael Muñoz
- Departamento de Biología del Cáncer, Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), Unidad asociada de Biomedicina UCLM, Unidad asociada al CSIC, Madrid, Spain
| | - Isabel Sánchez-Pérez
- Departamento de Bioquímica, Facultad de Medicina, Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), Unidad asociada de Biomedicina UCLM, Unidad asociada al CSIC, Madrid, Spain
| | - Borja Belandia
- Departamento de Biología del Cáncer, Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), Unidad asociada de Biomedicina UCLM, Unidad asociada al CSIC, Madrid, Spain
| | - María J Ruiz-Hidalgo
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas Universidad de Castilla-La Mancha, Unidad Asociada de Biomedicina UCLM, Unidad asociada al CSIC, Albacete, Spain.,Área de Bioquímica y Biología Molecular. Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Ricardo Sánchez-Prieto
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas Universidad de Castilla-La Mancha, Unidad Asociada de Biomedicina UCLM, Unidad asociada al CSIC, Albacete, Spain.,Departamento de Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain.,Instituto de Investigaciones Biomédicas 'Alberto Sols', Consejo Superior de Investigaciones Científicas (IIBM-CSIC)-Universidad de Castilla-La Mancha (UCLM), Albacete, Spain
| |
Collapse
|
26
|
Subramaniam N, Nair R, Marsden PA. Epigenetic Regulation of the Vascular Endothelium by Angiogenic LncRNAs. Front Genet 2021; 12:668313. [PMID: 34512715 PMCID: PMC8427604 DOI: 10.3389/fgene.2021.668313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/17/2021] [Indexed: 12/15/2022] Open
Abstract
The functional properties of the vascular endothelium are diverse and heterogeneous between vascular beds. This is especially evident when new blood vessels develop from a pre-existing closed cardiovascular system, a process termed angiogenesis. Endothelial cells are key drivers of angiogenesis as they undergo a highly choreographed cascade of events that has both exogenous (e.g., hypoxia and VEGF) and endogenous regulatory inputs. Not surprisingly, angiogenesis is critical in health and disease. Diverse therapeutics target proteins involved in coordinating angiogenesis with varying degrees of efficacy. It is of great interest that recent work on non-coding RNAs, especially long non-coding RNAs (lncRNAs), indicates that they are also important regulators of the gene expression paradigms that underpin this cellular cascade. The protean effects of lncRNAs are dependent, in part, on their subcellular localization. For instance, lncRNAs enriched in the nucleus can act as epigenetic modifiers of gene expression in the vascular endothelium. Of great interest to genetic disease, they are undergoing rapid evolution and show extensive inter- and intra-species heterogeneity. In this review, we describe endothelial-enriched lncRNAs that have robust effects in angiogenesis.
Collapse
Affiliation(s)
- Noeline Subramaniam
- Marsden Lab, Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Marsden Lab, Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON, Canada
| | - Ranju Nair
- Marsden Lab, Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON, Canada
- Marsden Lab, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Philip A. Marsden
- Marsden Lab, Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Marsden Lab, Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON, Canada
- Marsden Lab, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
27
|
Small molecule ERK5 kinase inhibitors paradoxically activate ERK5 signalling: be careful what you wish for…. Biochem Soc Trans 2021; 48:1859-1875. [PMID: 32915196 PMCID: PMC7609025 DOI: 10.1042/bst20190338] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/15/2022]
Abstract
ERK5 is a protein kinase that also contains a nuclear localisation signal and a transcriptional transactivation domain. Inhibition of ERK5 has therapeutic potential in cancer and inflammation and this has prompted the development of ERK5 kinase inhibitors (ERK5i). However, few ERK5i programmes have taken account of the ERK5 transactivation domain. We have recently shown that the binding of small molecule ERK5i to the ERK5 kinase domain stimulates nuclear localisation and paradoxical activation of its transactivation domain. Other kinase inhibitors paradoxically activate their intended kinase target, in some cases leading to severe physiological consequences highlighting the importance of mitigating these effects. Here, we review the assays used to monitor ERK5 activities (kinase and transcriptional) in cells, the challenges faced in development of small molecule inhibitors to the ERK5 pathway, and classify the molecular mechanisms of paradoxical activation of protein kinases by kinase inhibitors.
Collapse
|
28
|
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
|
29
|
Loss of Mef2D function enhances TLR induced IL-10 production in macrophages. Biosci Rep 2021; 40:225925. [PMID: 32725155 PMCID: PMC7442974 DOI: 10.1042/bsr20201859] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/27/2020] [Accepted: 07/29/2020] [Indexed: 12/24/2022] Open
Abstract
Mef2 transcription factors comprise a family of four different isoforms that regulate a number of processes including neuronal and muscle development. While roles for Mef2C and Mef2D have been described in B-cell development their role in immunity has not been extensively studied. In innate immune cells such as macrophages, TLRs drive the production of both pro- and anti-inflammatory cytokines. IL-10 is an important anti-inflammatory cytokine produced by macrophages and it establishes an autocrine feedback loop to inhibit pro-inflammatory cytokine production. We show here that macrophages from Mef2D knockout mice have elevated levels of IL-10 mRNA induction compared with wild-type cells following LPS stimulation. The secretion of IL-10 was also higher from Mef2D knockout macrophages and this correlated to a reduction in the secretion of TNF, IL-6 and IL-12p40. The use of an IL-10 neutralising antibody showed that this reduction in pro-inflammatory cytokine production in the Mef2D knockouts was IL-10 dependent. As the IL-10 promoter has previously been reported to contain a potential binding site for Mef2D, it is possible that the binding of other Mef2 isoforms in the absence of Mef2D may result in a higher activation of the IL-10 gene. Further studies with compound Mef2 isoforms would be required to address this. We also show that Mef2D is highly expressed in the thymus, but that loss of Mef2D does not affect thymic T-cell development or the production of IFNγ from CD8 T cells.
Collapse
|
30
|
Lu YW, Martino N, Gerlach BD, Lamar JM, Vincent PA, Adam AP, Schwarz JJ. MEF2 (Myocyte Enhancer Factor 2) Is Essential for Endothelial Homeostasis and the Atheroprotective Gene Expression Program. Arterioscler Thromb Vasc Biol 2021; 41:1105-1123. [PMID: 33406884 DOI: 10.1161/atvbaha.120.314978] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Atherosclerosis predominantly forms in regions of oscillatory shear stress while regions of laminar shear stress are protected. This protection is partly through the endothelium in laminar flow regions expressing an anti-inflammatory and antithrombotic gene expression program. Several molecular pathways transmitting these distinct flow patterns to the endothelium have been defined. Our objective is to define the role of the MEF2 (myocyte enhancer factor 2) family of transcription factors in promoting an atheroprotective endothelium. Approach and Results: Here, we show through endothelial-specific deletion of the 3 MEF2 factors in the endothelium, Mef2a, -c, and -d, that MEF2 is a critical regulator of vascular homeostasis. MEF2 deficiency results in systemic inflammation, hemorrhage, thrombocytopenia, leukocytosis, and rapid lethality. Transcriptome analysis reveals that MEF2 is required for normal regulation of 3 pathways implicated in determining the flow responsiveness of the endothelium. Specifically, MEF2 is required for expression of Klf2 and Klf4, 2 partially redundant factors essential for promoting an anti-inflammatory and antithrombotic endothelium. This critical requirement results in phenotypic similarities between endothelial-specific deletions of Mef2a/c/d and Klf2/4. In addition, MEF2 regulates the expression of Notch family genes, Notch1, Dll1, and Jag1, which also promote an atheroprotective endothelium. In contrast to these atheroprotective pathways, MEF2 deficiency upregulates an atherosclerosis promoting pathway through increasing the amount of TAZ (transcriptional coactivator with PDZ-binding motif). CONCLUSIONS Our results implicate MEF2 as a critical upstream regulator of several transcription factors responsible for gene expression programs that affect development of atherosclerosis and promote an anti-inflammatory and antithrombotic endothelium. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Yao Wei Lu
- Department of Molecular and Cellular Physiology (Y.W.L., N.M., B.D.G., J.M.L., P.A.V., A.P.A., J.J.S.), Albany Medical College, NY
| | - Nina Martino
- Department of Molecular and Cellular Physiology (Y.W.L., N.M., B.D.G., J.M.L., P.A.V., A.P.A., J.J.S.), Albany Medical College, NY
| | - Brennan D Gerlach
- Department of Molecular and Cellular Physiology (Y.W.L., N.M., B.D.G., J.M.L., P.A.V., A.P.A., J.J.S.), Albany Medical College, NY
| | - John M Lamar
- Department of Molecular and Cellular Physiology (Y.W.L., N.M., B.D.G., J.M.L., P.A.V., A.P.A., J.J.S.), Albany Medical College, NY
| | - Peter A Vincent
- Department of Molecular and Cellular Physiology (Y.W.L., N.M., B.D.G., J.M.L., P.A.V., A.P.A., J.J.S.), Albany Medical College, NY
| | - Alejandro P Adam
- Department of Molecular and Cellular Physiology (Y.W.L., N.M., B.D.G., J.M.L., P.A.V., A.P.A., J.J.S.), Albany Medical College, NY.,Department of Ophthalmology (A.P.A.), Albany Medical College, NY
| | - John J Schwarz
- Department of Molecular and Cellular Physiology (Y.W.L., N.M., B.D.G., J.M.L., P.A.V., A.P.A., J.J.S.), Albany Medical College, NY
| |
Collapse
|
31
|
Patel P, Naik UP. Platelet MAPKs-a 20+ year history: What do we really know? J Thromb Haemost 2020; 18:2087-2102. [PMID: 32574399 DOI: 10.1111/jth.14967] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 01/01/2023]
Abstract
The existence of mitogen activated protein kinases (MAPKs) in platelets has been known for more than 20 years. Since that time hundreds of reports have been published describing the conditions that cause MAPK activation in platelets and their role in regulating diverse platelet functions from the molecular to physiological level. However, this cacophony of reports, with inconsistent and sometimes contradictory findings, has muddied the waters leading to great confusion. Since the last review of platelet MAPKs was published more than a decade ago, there have been more than 50 reports, including the description of novel knockout mouse models, that have furthered our knowledge. Therefore, we undertook an extensive literature review to delineate what is known about platelet MAPKs. We specifically discuss what is currently known about how MAPKs are activated and what signaling cascades they regulate in platelets incorporating recent findings from knockout mouse models. In addition, we will discuss the role each MAPK plays in regulating distinct platelet functions. In doing so, we hope to clarify the role for MAPKs and identify knowledge gaps in this field that await future researchers. In addition, we discuss the limitations of current studies with a particular focus on the off-target effects of commonly used MAPK inhibitors. We conclude with a look at the clinical utility of MAPK inhibitors as potential antithrombotic therapies with an analysis of current clinical trial data.
Collapse
Affiliation(s)
- Pravin Patel
- Department of Medicine, Cardeza Center for Hemostasis, Thrombosis, and Vascular Biology, Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ulhas P Naik
- Department of Medicine, Cardeza Center for Hemostasis, Thrombosis, and Vascular Biology, Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
32
|
Efficient Suppression of NRAS-Driven Melanoma by Co-Inhibition of ERK1/2 and ERK5 MAPK Pathways. J Invest Dermatol 2020; 140:2455-2465.e10. [PMID: 32376279 DOI: 10.1016/j.jid.2020.03.972] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/24/2020] [Accepted: 03/18/2020] [Indexed: 12/19/2022]
Abstract
Cutaneous melanoma is a highly malignant tumor typically driven by somatic mutation in the oncogenes BRAF or NRAS, leading to uncontrolled activation of the MEK/ERK MAPK pathway. Despite the availability of immunotherapy, MAPK pathway‒targeting regimens are still a valuable treatment option for BRAF-mutant melanoma. Unfortunately, patients with NRAS mutation do not benefit from such therapies owing to the lack of targetable BRAF mutations and a high degree of intrinsic and acquired resistance toward MEK inhibition. Here, we demonstrate that concomitant inhibition of ERK5 removes this constraint and effectively sensitizes NRAS-mutant melanoma cells for MAPK pathway‒targeting therapy. Using approved MEK inhibitors or a pharmacologic ERK inhibitor, we demonstrate that MAPK inhibition triggers a delayed activation of ERK5 through a PDGFR inhibitor-sensitive pathway in NRAS-mutant melanoma cells, resulting in sustained proliferation and survival. ERK5 phosphorylation also occurred naturally in NRAS-mutant melanoma cells and correlated with nuclear localization of its stem cell-associated effector KLF2. Importantly, MEK/ERK5 co-inhibition prevented long-term growth of human NRAS-mutant melanoma cells in vitro and effectively repressed tumor progression in a xenotransplant mouse model. Our findings suggest MEK/ERK5 cotargeting as a potential treatment option for NRAS-mutant melanoma, which currently is not amenable for targeted therapies.
Collapse
|
33
|
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
|
34
|
Lochhead PA, Tucker JA, Tatum NJ, Wang J, Oxley D, Kidger AM, Johnson VP, Cassidy MA, Gray NS, Noble MEM, Cook SJ. Paradoxical activation of the protein kinase-transcription factor ERK5 by ERK5 kinase inhibitors. Nat Commun 2020; 11:1383. [PMID: 32170057 PMCID: PMC7069993 DOI: 10.1038/s41467-020-15031-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 02/11/2020] [Indexed: 12/20/2022] Open
Abstract
The dual protein kinase-transcription factor, ERK5, is an emerging drug target in cancer and inflammation, and small-molecule ERK5 kinase inhibitors have been developed. However, selective ERK5 kinase inhibitors fail to recapitulate ERK5 genetic ablation phenotypes, suggesting kinase-independent functions for ERK5. Here we show that ERK5 kinase inhibitors cause paradoxical activation of ERK5 transcriptional activity mediated through its unique C-terminal transcriptional activation domain (TAD). Using the ERK5 kinase inhibitor, Compound 26 (ERK5-IN-1), as a paradigm, we have developed kinase-active, drug-resistant mutants of ERK5. With these mutants, we show that induction of ERK5 transcriptional activity requires direct binding of the inhibitor to the kinase domain. This in turn promotes conformational changes in the kinase domain that result in nuclear translocation of ERK5 and stimulation of gene transcription. This shows that both the ERK5 kinase and TAD must be considered when assessing the role of ERK5 and the effectiveness of anti-ERK5 therapeutics.
Collapse
Affiliation(s)
- Pamela A Lochhead
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.
| | - Julie A Tucker
- York Biomedical Research Institute and Department of Biology, University of York, York, YO10 5DD, UK
| | - Natalie J Tatum
- CRUK Newcastle Drug Discovery Unit, Newcastle University Centre for Cancer, Newcastle University, Newcastle, NE2 4HH, UK
| | - Jinhua Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - David Oxley
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Andrew M Kidger
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Victoria P Johnson
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
- Institute of Cancer Research, Chester Beatty Laboratories, 237 Fulham Road, London, SW3 6JB, UK
| | - Megan A Cassidy
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Martin E M Noble
- CRUK Newcastle Drug Discovery Unit, Newcastle University Centre for Cancer, Newcastle University, Newcastle, NE2 4HH, UK
| | - Simon J Cook
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.
| |
Collapse
|
35
|
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
|
36
|
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
|
37
|
A novel MEF2C mutation in lymphoid neoplasm diffuse large B-cell lymphoma promotes tumorigenesis by increasing c-JUN expression. Naunyn Schmiedebergs Arch Pharmacol 2020; 393:1549-1558. [PMID: 31900516 DOI: 10.1007/s00210-019-01764-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/01/2019] [Indexed: 12/14/2022]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most aggressive non-Hodgkin lymphoma (NHL), accounting for about 31% of the newly diagnosed NHL worldwide. Although approximately 60% of patients who initially received a standard R-CHOP treatment likely have a 3-year event-free survival, many patients become refractory or relapsed due to the genetic heterogeneity of this malignancy. Hence, new treatment strategies are urgently needed. MEF2C, a member of the MEF2 transcription factor family gene, plays great important roles involved in the development of various tissues and the pathogenesis of lymphoma. However, the exact functions and molecular mechanisms of MEF2C in DLBCL are not fully investigated. By Sanger sequencing, we identified a novel point mutation of MEF2C at the p.N389 site in DLBCL patient, which was further validated by several DLBCL cell lines. Intriguingly, we found that the p.N389S mutation did not influence MEF2C expression, protein stability, and subcellular distribution, but enhanced its transcriptional activity. Furthermore, we demonstrated that MEF2C p.N389S mutation promotes DLBCL cell proliferation, cellular adhesion, and tumor formation in nude mice. On mechanism, our data revealed that MEF2C p.N389S mutation increases c-JUN expression, and c-JUN regulation mediated the oncogenic function of MEF2C p.N389S mutation on DLBCL cells. Our finding may provide a significant insight into the DLBCL and a compelling therapy target for this disease treatment.
Collapse
|
38
|
Singh MV, Kotla S, Le NT, Ae Ko K, Heo KS, Wang Y, Fujii Y, Thi Vu H, McBeath E, Thomas TN, Jin Gi Y, Tao Y, Medina JL, Taunton J, Carson N, Dogra V, Doyley MM, Tyrell A, Lu W, Qiu X, Stirpe NE, Gates KJ, Hurley C, Fujiwara K, Maggirwar SB, Schifitto G, Abe JI. Senescent Phenotype Induced by p90RSK-NRF2 Signaling Sensitizes Monocytes and Macrophages to Oxidative Stress in HIV-Positive Individuals. Circulation 2019; 139:1199-1216. [PMID: 30586719 DOI: 10.1161/circulationaha.118.036232] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The incidence of cardiovascular disease is higher in HIV-positive (HIV+) patients than it is in the average population, and combination antiretroviral therapy (cART) is a recognized risk factor for cardiovascular disease. However, the molecular mechanisms that link cART and cardiovascular disease are currently unknown. Our study explores the role of the activation of p90RSK, a reactive oxygen species-sensitive kinase, in engendering senescent phenotype in macrophages and accelerating atherogenesis in patients undergoing cART. METHODS Peripheral whole blood from cART-treated HIV+ individuals and nontreated HIV-negative individuals was treated with H2O2 (200 µmol/L) for 4 minutes, and p90RSK activity in CD14+ monocytes was measured. Plaque formation in the carotids was also analyzed in these individuals. Macrophage senescence was determined by evaluating their efferocytotic ability, antioxidation-related molecule expression, telomere length, and inflammatory gene expression. The involvement of p90RSK-NRF2 signaling in cART-induced senescence was assessed by p90RSK-specific inhibitor (FMK-MEA) or dominant-negative p90RSK (DN-p90RSK) and NRF2 activator (NRF2A). Further, the severity of atherosclerosis was determined in myeloid cell-specific wild-type and DN-p90RSK transgenic mice. RESULTS Monocytes from HIV+ patients exhibited higher levels of p90RSK activity and were also more sensitive to reactive oxygen species than monocytes from HIV-negative individuals. A multiple linear regression analysis involving cART, Reynolds cardiovascular risk score, and basal p90RSK activity revealed that cART and basal p90RSK activity were the 2 significant determinants of plaque formation. Many of the antiretroviral drugs individually activated p90RSK, which simultaneously triggered all components of the macrophage senescent phenotype. cART inhibited antioxidant response element reporter activity via ERK5 S496 phosphorylation. NRF2A reversed the H2O2-induced overactivation of p90RSK in cART-treated macrophages by countering the induction of senescent phenotype. Last, the data obtained from our gain- or loss-of-function mice conclusively showed the crucial role of p90RSK in inducing senescent phenotype in macrophages and atherogenesis. CONCLUSIONS cART increased monocyte/macrophage sensitivity to reactive oxygen species- in HIV+ individuals by suppressing NRF2-ARE activity via p90RSK-mediated ERK5 S496 phosphorylation, which coordinately elicited senescent phenotypes and proinflammatory responses. As such, our report underscores the importance of p90RSK regulation in monocytes/macrophages as a viable biomarker and therapeutic target for preventing cardiovascular disease, especially in HIV+ patients treated with cART.
Collapse
Affiliation(s)
- Meera V Singh
- Departments of Microbiology and Immunology (M.V.S., N.E.S., K.J.G., S.B.M.), University of Rochester, NY
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston (S.K., N.-T.L., K.A.K., K.-S.H., Y.W., Y.F., H.T.V., E.M., T.N.T., Y.J.G., Y.T., J.L.M., K.F., J.-i.A.).,Radiology Research (S.K., N.-T.L., K.A.K.), Houston Methodist Research Institute, TX
| | - Nhat-Tu Le
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston (S.K., N.-T.L., K.A.K., K.-S.H., Y.W., Y.F., H.T.V., E.M., T.N.T., Y.J.G., Y.T., J.L.M., K.F., J.-i.A.).,Departments of Cardiovascular Sciences (N.-T.L.), Houston Methodist Research Institute, TX.,Radiology Research (S.K., N.-T.L., K.A.K.), Houston Methodist Research Institute, TX
| | - Kyung Ae Ko
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston (S.K., N.-T.L., K.A.K., K.-S.H., Y.W., Y.F., H.T.V., E.M., T.N.T., Y.J.G., Y.T., J.L.M., K.F., J.-i.A.).,Radiology Research (S.K., N.-T.L., K.A.K.), Houston Methodist Research Institute, TX
| | - Kyung-Sun Heo
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston (S.K., N.-T.L., K.A.K., K.-S.H., Y.W., Y.F., H.T.V., E.M., T.N.T., Y.J.G., Y.T., J.L.M., K.F., J.-i.A.).,Institute of Drug Research and Development, Chungnam National University, Daejeon, Republic of Korea (K.-S.H.)
| | - Yin Wang
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston (S.K., N.-T.L., K.A.K., K.-S.H., Y.W., Y.F., H.T.V., E.M., T.N.T., Y.J.G., Y.T., J.L.M., K.F., J.-i.A.)
| | - Yuka Fujii
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston (S.K., N.-T.L., K.A.K., K.-S.H., Y.W., Y.F., H.T.V., E.M., T.N.T., Y.J.G., Y.T., J.L.M., K.F., J.-i.A.)
| | - Hang Thi Vu
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston (S.K., N.-T.L., K.A.K., K.-S.H., Y.W., Y.F., H.T.V., E.M., T.N.T., Y.J.G., Y.T., J.L.M., K.F., J.-i.A.)
| | - Elena McBeath
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston (S.K., N.-T.L., K.A.K., K.-S.H., Y.W., Y.F., H.T.V., E.M., T.N.T., Y.J.G., Y.T., J.L.M., K.F., J.-i.A.)
| | - Tamlyn N Thomas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston (S.K., N.-T.L., K.A.K., K.-S.H., Y.W., Y.F., H.T.V., E.M., T.N.T., Y.J.G., Y.T., J.L.M., K.F., J.-i.A.)
| | - Young Jin Gi
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston (S.K., N.-T.L., K.A.K., K.-S.H., Y.W., Y.F., H.T.V., E.M., T.N.T., Y.J.G., Y.T., J.L.M., K.F., J.-i.A.)
| | - Yunting Tao
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston (S.K., N.-T.L., K.A.K., K.-S.H., Y.W., Y.F., H.T.V., E.M., T.N.T., Y.J.G., Y.T., J.L.M., K.F., J.-i.A.)
| | - Jan L Medina
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston (S.K., N.-T.L., K.A.K., K.-S.H., Y.W., Y.F., H.T.V., E.M., T.N.T., Y.J.G., Y.T., J.L.M., K.F., J.-i.A.)
| | - Jack Taunton
- Department of Cellular and Molecular Pharmacology, University of California-San Francisco (J.T.)
| | - Nancy Carson
- Imaging Sciences (N.C., V.D.), University of Rochester, NY
| | - Vikram Dogra
- Imaging Sciences (N.C., V.D.), University of Rochester, NY
| | - Marvin M Doyley
- Electrical and Computer Engineering (M.M.D.), University of Rochester, NY
| | - Alicia Tyrell
- Biostatistics and Computational Biology (A.T., W.L., X.Q.), University of Rochester, NY
| | - Wang Lu
- Biostatistics and Computational Biology (A.T., W.L., X.Q.), University of Rochester, NY
| | - Xing Qiu
- Biostatistics and Computational Biology (A.T., W.L., X.Q.), University of Rochester, NY
| | - Nicole E Stirpe
- Departments of Microbiology and Immunology (M.V.S., N.E.S., K.J.G., S.B.M.), University of Rochester, NY
| | - Kathleen J Gates
- Departments of Microbiology and Immunology (M.V.S., N.E.S., K.J.G., S.B.M.), University of Rochester, NY
| | - Christine Hurley
- Medicine, Infectious Disease (C.H.), University of Rochester, NY
| | - Keigi Fujiwara
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston (S.K., N.-T.L., K.A.K., K.-S.H., Y.W., Y.F., H.T.V., E.M., T.N.T., Y.J.G., Y.T., J.L.M., K.F., J.-i.A.)
| | - Sanjay B Maggirwar
- Departments of Microbiology and Immunology (M.V.S., N.E.S., K.J.G., S.B.M.), University of Rochester, NY
| | | | - Jun-Ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston (S.K., N.-T.L., K.A.K., K.-S.H., Y.W., Y.F., H.T.V., E.M., T.N.T., Y.J.G., Y.T., J.L.M., K.F., J.-i.A.)
| |
Collapse
|
39
|
Emerging roles for MEF2 in brain development and mental disorders. Curr Opin Neurobiol 2019; 59:49-58. [PMID: 31129473 DOI: 10.1016/j.conb.2019.04.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 04/18/2019] [Indexed: 12/26/2022]
Abstract
The MEF2 family of transcription factors regulate large programs of gene expression important for the development and maintenance of many tissues, including the brain. MEF2 proteins are regulated by neuronal synaptic activity, and they recruit several epigenetic enzymes to influence chromatin structure and gene expression during development and throughout adulthood. Here, we provide a brief review of the recent literature reporting important roles for MEF2 during early brain development and function, and we highlight emerging roles for MEF2 as a risk factor for multiple neurodevelopmental disorders and mental illnesses, such as autism, intellectual disability, and schizophrenia.
Collapse
|
40
|
Jiang W, Jin G, Cai F, Chen X, Cao N, Zhang X, Liu J, Chen F, Wang F, Dong W, Zhuang H, Hua ZC. Extracellular signal-regulated kinase 5 increases radioresistance of lung cancer cells by enhancing the DNA damage response. Exp Mol Med 2019; 51:1-20. [PMID: 30804322 PMCID: PMC6389946 DOI: 10.1038/s12276-019-0209-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 10/26/2018] [Accepted: 10/29/2018] [Indexed: 12/18/2022] Open
Abstract
Radiotherapy is a frequent mode of cancer treatment, although the development of radioresistance limits its effectiveness. Extensive investigations indicate the diversity of the mechanisms underlying radioresistance. Here, we aimed to explore the effects of extracellular signal-regulated kinase 5 (ERK5) on lung cancer radioresistance and the associated mechanisms. Our data showed that ERK5 is activated during solid lung cancer development, and ectopic expression of ERK5 promoted cell proliferation and G2/M cell cycle transition. In addition, we found that ERK5 is a potential regulator of radiosensitivity in lung cancer cells. Mechanistic investigations revealed that ERK5 could trigger IR-induced activation of Chk1, which has been implicated in DNA repair and cell cycle arrest in response to DNA double-strand breaks (DSBs). Subsequently, ERK5 knockdown or pharmacological inhibition selectively inhibited colony formation of lung cancer cells and enhanced IR-induced G2/M arrest and apoptosis. In vivo, ERK5 knockdown strongly radiosensitized A549 and LLC tumor xenografts to inhibition, with a higher apoptotic response and reduced tumor neovascularization. Taken together, our data indicate that ERK5 is a novel potential target for the treatment of lung cancer, and its expression might be used as a biomarker to predict radiosensitivity in NSCLC patients. Resistance to radiotherapy in patients with lung cancer may be countered by targeting a protein involved in promoting DNA repair. Radiotherapy causes DNA double-stranded breaks in lung cancer cells in order to kill them. However, cancer cells can show improved DNA repair and responses to damage, resulting in resistance to treatment. Zi-Chun Hua, Hongqin Zhuang at Nanjing University in China and co-workers examined the activity of the extracellular signal-related kinase 5 (ERK5) protein in response to the stress of ionizing radiation. They found that after radiation exposure ERK5 increased expression of another protein involved in DNA repair, facilitating cancer cell recovery. Knocking out ERK5 suppressed this resistance to radiotherapy. ERK5 could be a valuable target for treating lung cancer, and ERK5 expression level could be used as a biomarker for patient sensitivity to radiotherapy.
Collapse
Affiliation(s)
- Weiwei Jiang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Guanghui Jin
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China.,Department of Basic Medical Sciences, Medical College, Xiamen University, Xiamen, PR China
| | - Fangfang Cai
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Xiao Chen
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Nini Cao
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Xiangyu Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Jia Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Fei Chen
- Department of Nuclear Medicine, The Affiliated Nanjing First Hospital, Nanjing Medical University, Nanjing, PR China
| | - Feng Wang
- Department of Nuclear Medicine, The Affiliated Nanjing First Hospital, Nanjing Medical University, Nanjing, PR China
| | - Wei Dong
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Hongqin Zhuang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China.
| | - Zi-Chun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China. .,Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories Inc., Changzhou, 213164, PR China.
| |
Collapse
|
41
|
Nguyen D, Lemos C, Wortmann L, Eis K, Holton SJ, Boemer U, Moosmayer D, Eberspaecher U, Weiske J, Lechner C, Prechtl S, Suelzle D, Siegel F, Prinz F, Lesche R, Nicke B, Nowak-Reppel K, Himmel H, Mumberg D, von Nussbaum F, Nising CF, Bauser M, Haegebarth A. Discovery and Characterization of the Potent and Highly Selective (Piperidin-4-yl)pyrido[3,2- d]pyrimidine Based in Vitro Probe BAY-885 for the Kinase ERK5. J Med Chem 2019; 62:928-940. [PMID: 30563338 DOI: 10.1021/acs.jmedchem.8b01606] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The availability of a chemical probe to study the role of a specific domain of a protein in a concentration- and time-dependent manner is of high value. Herein, we report the identification of a highly potent and selective ERK5 inhibitor BAY-885 by high-throughput screening and subsequent structure-based optimization. ERK5 is a key integrator of cellular signal transduction, and it has been shown to play a role in various cellular processes such as proliferation, differentiation, apoptosis, and cell survival. We could demonstrate that inhibition of ERK5 kinase and transcriptional activity with a small molecule did not translate into antiproliferative activity in different relevant cell models, which is in contrast to the results obtained by RNAi technology.
Collapse
Affiliation(s)
- Duy Nguyen
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Clara Lemos
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Lars Wortmann
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Knut Eis
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Simon J Holton
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Ulf Boemer
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Dieter Moosmayer
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Uwe Eberspaecher
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Joerg Weiske
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Christian Lechner
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Stefan Prechtl
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Detlev Suelzle
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Franziska Siegel
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Florian Prinz
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Ralf Lesche
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Barbara Nicke
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | | | - Herbert Himmel
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Dominik Mumberg
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Franz von Nussbaum
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Carl F Nising
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Marcus Bauser
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Andrea Haegebarth
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| |
Collapse
|
42
|
Kang C, Kim JS, Kim CY, Kim EY, Chung HM. The Pharmacological Inhibition of ERK5 Enhances Apoptosis in Acute Myeloid Leukemia Cells. Int J Stem Cells 2018; 11:227-234. [PMID: 30343550 PMCID: PMC6285287 DOI: 10.15283/ijsc18053] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/13/2018] [Accepted: 08/14/2018] [Indexed: 12/26/2022] Open
Abstract
Acute myeloid leukemia (AML) is a fatal hematological malignancy which is resistant to a variety of chemotherapy drugs. Extracellular signal-regulated kinase 5 (ERK5) plays a novel role in chemoresistance in some cancer cells and this pathway is a central mediator of cell survival and apoptotic regulation. The aim of this study was to investigate the effect of ERK5 inhibitor, XMD8-92, on proliferation and apoptosis in AML cell lines. Findings showed that XMD8-92 inhibited the activation of ERK5 by G-CSF and decreased the expression of c-Myc and Cyclin D1. The treatment of XMD8-92 reduced the phosphorylation of ERK5 leading to a distinct inhibition of cell proliferation and increased apoptosis in Kasumi-1 and HL-60 cells. Taken together, our study suggests that the inhibition of ERK5 by XMD8-92 can trigger apoptosis and inhibit proliferation in AMLs. Therefore, the inhibition of ERK5 may be an effective adjuvant in AML chemotherapy.
Collapse
Affiliation(s)
- Changhee Kang
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Jong Soo Kim
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - C-Yoon Kim
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Eun-Young Kim
- Stem Cell Research Center, Jeju National University, Jeju, Korea.,Mirae Cell Bio Co. LTD, Seoul, Korea
| | - Hyung-Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| |
Collapse
|
43
|
Liu L, Levin DE. Intracellular mechanism by which genotoxic stress activates yeast SAPK Mpk1. Mol Biol Cell 2018; 29:2898-2909. [PMID: 30230955 PMCID: PMC6249863 DOI: 10.1091/mbc.e18-07-0441] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Stress-activated MAP kinases (SAPKs) respond to a wide variety of stressors. In most cases, the pathways through which specific stress signals are transmitted to the SAPKs are not known. The Saccharomyces cerevisiae SAPK Mpk1 (Slt2) is a well-characterized component of the cell-wall integrity (CWI) signaling pathway, which responds to physical and chemical challenges to the cell wall. However, Mpk1 is also activated in response to genotoxic stress through an unknown pathway. We show that, in contrast to cell-wall stress, the pathway for Mpk1 activation by genotoxic stress does not involve the stimulation of the MAP kinase kinases (MEKs) that function immediately upstream of Mpk1. Instead, DNA damage activates Mpk1 through induction of proteasomal degradation of Msg5, the dual-specificity protein phosphatase principally responsible for maintaining Mpk1 in a low-activity state in the absence of stress. Blocking Msg5 degradation in response to genotoxic stress prevented Mpk1 activation. This work raises the possibility that other Mpk1-activating stressors act intracellularly at different points along the canonical Mpk1 activation pathway.
Collapse
Affiliation(s)
- Li Liu
- Department of Molecular and Cell Biology, Boston University Goldman School of Dental Medicine, Boston, MA 02118
| | - David E Levin
- Department of Molecular and Cell Biology, Boston University Goldman School of Dental Medicine, Boston, MA 02118.,Department of Microbiology, Boston University School of Medicine, Boston, MA 02118
| |
Collapse
|
44
|
Abstract
Cerebral cavernous malformations (CCM) are manifested by microvascular lesions characterized by leaky endothelial cells with minimal intervening parenchyma predominantly in the central nervous system predisposed to hemorrhagic stroke, resulting in focal neurological defects. Till date, three proteins are implicated in this condition: CCM1 (KRIT1), CCM2 (MGC4607), and CCM3 (PDCD10). These multi-domain proteins form a protein complex via CCM2 that function as a docking site for the CCM signaling complex, which modulates many signaling pathways. Defects in the formation of this signaling complex have been shown to affect a wide range of cellular processes including cell-cell contact stability, vascular angiogenesis, oxidative damage protection and multiple biogenic events. In this review we provide an update on recent advances in structure and function of these CCM proteins, especially focusing on the signaling cascades involved in CCM pathogenesis and the resultant CCM cellular phenotypes in the past decade.
Collapse
Affiliation(s)
- Akhil Padarti
- Department of Biomedical Sciences, Texas Tech University Health Science Center El Paso, El Paso, TX 79905, USA
| | - Jun Zhang
- Department of Biomedical Sciences, Texas Tech University Health Science Center El Paso, El Paso, TX 79905, USA
| |
Collapse
|
45
|
Kashino Y, Obara Y, Okamoto Y, Saneyoshi T, Hayashi Y, Ishii K. ERK5 Phosphorylates K v4.2 and Inhibits Inactivation of the A-Type Current in PC12 Cells. Int J Mol Sci 2018; 19:ijms19072008. [PMID: 29996472 PMCID: PMC6073465 DOI: 10.3390/ijms19072008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 06/28/2018] [Accepted: 06/28/2018] [Indexed: 12/31/2022] Open
Abstract
Extracellular signal-regulated kinase 5 (ERK5) regulates diverse physiological responses such as proliferation, differentiation, and gene expression. Previously, we demonstrated that ERK5 is essential for neurite outgrowth and catecholamine biosynthesis in PC12 cells and sympathetic neurons. However, it remains unclear how ERK5 regulates the activity of ion channels, which are important for membrane excitability. Thus, we examined the effect of ERK5 on the ion channel activity in the PC12 cells that overexpress both ERK5 and the constitutively active MEK5 mutant. The gene and protein expression levels of voltage-dependent Ca2+ and K+ channels were determined by RT-qPCR or Western blotting. The A-type K+ current was recorded using the whole-cell patch clamp method. In these ERK5-activated cells, the gene expression levels of voltage-dependent L- and P/Q-type Ca2+ channels did not alter, but the N-type Ca2+ channel was slightly reduced. In contrast, those of Kv4.2 and Kv4.3, which are components of the A-type current, were significantly enhanced. Unexpectedly, the protein levels of Kv4.2 were not elevated by ERK5 activation, but the phosphorylation levels were increased by ERK5 activation. By electrophysiological analysis, the inactivation time constant of the A-type current was prolonged by ERK5 activation, without changes in the peak current. Taken together, ERK5 inhibits an inactivation of the A-type current by phosphorylation of Kv4.2, which may contribute to the neuronal differentiation process.
Collapse
Affiliation(s)
- Yurina Kashino
- Department of Pharmacology, Yamagata University School of Medicine, Yamagata 990-9585, Japan.
| | - Yutaro Obara
- Department of Pharmacology, Yamagata University School of Medicine, Yamagata 990-9585, Japan.
| | - Yosuke Okamoto
- Department of Pharmacology, Yamagata University School of Medicine, Yamagata 990-9585, Japan.
| | - Takeo Saneyoshi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan.
| | - Yasunori Hayashi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan.
| | - Kuniaki Ishii
- Department of Pharmacology, Yamagata University School of Medicine, Yamagata 990-9585, Japan.
| |
Collapse
|
46
|
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
|
47
|
Mitochondrial Complex I activity signals antioxidant response through ERK5. Sci Rep 2018; 8:7420. [PMID: 29743487 PMCID: PMC5943249 DOI: 10.1038/s41598-018-23884-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 03/21/2018] [Indexed: 11/29/2022] Open
Abstract
Oxidative phosphorylation (OXPHOS) generates ROS as a byproduct of mitochondrial complex I activity. ROS-detoxifying enzymes are made available through the activation of their antioxidant response elements (ARE) in their gene promoters. NRF2 binds to AREs and induces this anti-oxidant response. We show that cells from multiple origins performing OXPHOS induced NRF2 expression and its transcriptional activity. The NRF2 promoter contains MEF2 binding sites and the MAPK ERK5 induced MEF2-dependent NRF2 expression. Blocking OXPHOS in a mouse model decreased Erk5 and Nrf2 expression. Furthermore, fibroblasts derived from patients with mitochondrial disorders also showed low expression of ERK5 and NRF2 mRNAs. Notably, in cells lacking functional mitochondrial complex I activity OXPHOS did not induce ERK5 expression and failed to generate this anti-oxidant response. Complex I activity induces ERK5 expression through fumarate accumulation. Eukaryotic cells have evolved a genetic program to prevent oxidative stress directly linked to OXPHOS and not requiring ROS.
Collapse
|
48
|
Integration of human adipocyte chromosomal interactions with adipose gene expression prioritizes obesity-related genes from GWAS. Nat Commun 2018; 9:1512. [PMID: 29666371 PMCID: PMC5904163 DOI: 10.1038/s41467-018-03554-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 02/22/2018] [Indexed: 12/13/2022] Open
Abstract
Increased adiposity is a hallmark of obesity and overweight, which affect 2.2 billion people world-wide. Understanding the genetic and molecular mechanisms that underlie obesity-related phenotypes can help to improve treatment options and drug development. Here we perform promoter Capture Hi-C in human adipocytes to investigate interactions between gene promoters and distal elements as a transcription-regulating mechanism contributing to these phenotypes. We find that promoter-interacting elements in human adipocytes are enriched for adipose-related transcription factor motifs, such as PPARG and CEBPB, and contribute to heritability of cis-regulated gene expression. We further intersect these data with published genome-wide association studies for BMI and BMI-related metabolic traits to identify the genes that are under genetic cis regulation in human adipocytes via chromosomal interactions. This integrative genomics approach identifies four cis-eQTL-eGene relationships associated with BMI or obesity-related traits, including rs4776984 and MAP2K5, which we further confirm by EMSA, and highlights 38 additional candidate genes.
Collapse
|
49
|
Belkahla S, Haq Khan AU, Gitenay D, Alexia C, Gondeau C, Vo DN, Orecchioni S, Talarico G, Bertolini F, Cartron G, Hernandez J, Daujat-Chavanieu M, Allende-Vega N, Gonzalez MV. Changes in metabolism affect expression of ABC transporters through ERK5 and depending on p53 status. Oncotarget 2017; 9:1114-1129. [PMID: 29416681 PMCID: PMC5787424 DOI: 10.18632/oncotarget.23305] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 12/05/2017] [Indexed: 12/25/2022] Open
Abstract
Changes in metabolism require the efflux and influx of a diverse variety of metabolites. The ABC superfamily of transporters regulates the exchange of hundreds of substrates through the impermeable cell membrane. We show here that a metabolic switch to oxidative phosphorylation (OXPHOS), either by treating cells with dichloroacetate (DCA) or by changing the available substrates, reduced expression of ABCB1, ABCC1, ABCC5 and ABCG2 in wild-type p53-expressing cells. This metabolic change reduced histone changes associated to active promoters. Notably, DCA also inhibited expression of these genes in two animal models in vivo. In contrast, OXPHOS increased the expression of the same transporters in mutated (mut) or null p53-expressing cells. ABC transporters control the export of drugs from cancer cells and render tumors resistant to chemotherapy, playing an important role in multiple drug resistance (MDR). Wtp53 cells forced to perform OXPHOS showed impaired drug clearance. In contrast mutp53 cells increased drug clearance when performing OXPHOS. ABC transporter promoters contain binding sites for the transcription factors MEF2, NRF1 and NRF2 that are targets of the MAPK ERK5. OXPHOS induced expression of the MAPK ERK5. Decreasing ERK5 levels in wtp53 cells increased ABC expression whereas it inhibited expression in mutp53 cells. Our results showed that the ERK5/MEF2 pathway controlled ABC expression depending on p53 status.
Collapse
Affiliation(s)
- Sana Belkahla
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France
| | - Abrar Ul Haq Khan
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France.,Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), CHU Montpellier, Montpellier, France
| | - Delphine Gitenay
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France.,Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), CHU Montpellier, Montpellier, France
| | - Catherine Alexia
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France.,Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), CHU Montpellier, Montpellier, France
| | - Claire Gondeau
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France.,Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), CHU Montpellier, Montpellier, France.,Département d'Hépato-gastroentérologie A, Hôpital Saint Eloi, CHU Montpellier, Montpellier, France
| | - Dang-Nghiem Vo
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France
| | - Stefania Orecchioni
- Department of Oncology and Hemato-Oncology, European Institute of Oncology, Milan, Italy
| | - Giovanna Talarico
- Department of Oncology and Hemato-Oncology, European Institute of Oncology, Milan, Italy
| | - Francesco Bertolini
- Department of Oncology and Hemato-Oncology, European Institute of Oncology, Milan, Italy
| | - Guillaume Cartron
- Département d'Hématologie Clinique, CHU Montpellier, Université Montpellier I, Montpellier, France
| | - Javier Hernandez
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France
| | - Martine Daujat-Chavanieu
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France.,Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), CHU Montpellier, Montpellier, France
| | - Nerea Allende-Vega
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France.,Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), CHU Montpellier, Montpellier, France.,These two authors share senior authorship
| | - Martin Villalba Gonzalez
- Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), INSERM, Univ De Montpellier, Montpellier, France.,Department of Lymphocyte Differentiation, Tolerance and Metabolism: Basis for Immunotherapy, Institut De Médecine Régénératrice Et Biothérapie (IRMB), CHU Montpellier, Montpellier, France.,These two authors share senior authorship
| |
Collapse
|
50
|
Shen S, Huang D, Feng G, Zhu L, Zhang Y, Cao P, Zheng K, Zhang D, Feng X. MEF2 Transcription Factor Regulates Osteogenic Differentiation of Dental Pulp Stem Cells. Cell Reprogram 2017; 18:237-45. [PMID: 27459583 DOI: 10.1089/cell.2016.0016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The myocyte enhancer factor-2 (MEF2) is a member of the MADS-box family. It controls the expression of genes that are critical for biological processes such as proliferation, cell death, and differentiation. Some studies have shown that MEF2 expression is enhanced in osteogenic progenitor cells established from bone marrow stromal cells with other types of mesenchymal progenitor cells. However, the effect of MEF2 on dental pulp stem cells (DPSCs) is unclear. In this study, we investigate the effect of MEF2 on regulating osteogenic differentiation and proliferation of DPSCs. We find that MEF2 is stably expressed in DPSCs, and the expression is increased time-dependently along with cell osteogenic differentiation. MEF2 expression also increases the alkaline phosphatase (ALP), runt-related transcription factor 2 (Runx2) activity, and enhances mineralization in DPSCs. SB202190, inhibitor of p38, blocks the p38/MEF2 pathway and osteogenic differentiation. In addition, MEF2 overexpression inhibits DPSC proliferation. In summary, our data indicate that MEF2 not only regulates DPSCs as an inhibitor of cell proliferation but is also a promoter of osteogenic differentiation through the p38/MEF2 signaling pathway.
Collapse
Affiliation(s)
- Shuling Shen
- 1 The Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, China .,2 Department of Stomatology, Affiliated Hospital of Nantong University , Nantong, China
| | - Dan Huang
- 2 Department of Stomatology, Affiliated Hospital of Nantong University , Nantong, China
| | - Guijuan Feng
- 2 Department of Stomatology, Affiliated Hospital of Nantong University , Nantong, China
| | - Linhe Zhu
- 3 School of Science, Nanjing University of Aeronautics and Astronautics , Nanjing, China
| | - Ye Zhang
- 1 The Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, China .,2 Department of Stomatology, Affiliated Hospital of Nantong University , Nantong, China
| | - Peipei Cao
- 1 The Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, China .,2 Department of Stomatology, Affiliated Hospital of Nantong University , Nantong, China
| | - Ke Zheng
- 1 The Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, China .,2 Department of Stomatology, Affiliated Hospital of Nantong University , Nantong, China
| | - Dongmei Zhang
- 4 Department of Pathogen Biology, Medical College, Nantong University , Nantong, China
| | - Xingmei Feng
- 1 The Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University , Nantong, China .,2 Department of Stomatology, Affiliated Hospital of Nantong University , Nantong, China
| |
Collapse
|