1
|
Tuminello S, Ashebir YA, Schroff C, Ramaswami S, Durmus N, Chen Y, Snuderl M, Shao Y, Reibman J, Arslan AA. Genome-wide DNA methylation profiles and breast cancer among World Trade Center survivors. Environ Epidemiol 2024; 8:e313. [PMID: 38841706 PMCID: PMC11152787 DOI: 10.1097/ee9.0000000000000313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/08/2024] [Indexed: 06/07/2024] Open
Abstract
Background Increased incidence of cancer has been reported among World Trade Center (WTC)-exposed persons. Aberrant DNA methylation is a hallmark of cancer development. To date, only a few small studies have investigated the relationship between WTC exposure and DNA methylation. The main objective of this study was to assess the DNA methylation profiles of WTC-exposed community members who remained cancer free and those who developed breast cancer. Methods WTC-exposed women were selected from the WTC Environmental Health Center clinic, with peripheral blood collected during routine clinical monitoring visits. The reference group was selected from the NYU Women's Health Study, a prospective cohort study with blood samples collected before 9 November 2001. The Infinium MethylationEPIC array was used for global DNA methylation profiling, with adjustments for cell type composition and other confounders. Annotated probes were used for biological pathway and network analysis. Results A total of 64 WTC-exposed (32 cancer free and 32 with breast cancer) and 32 WTC-unexposed (16 cancer free and 16 with prediagnostic breast cancer) participants were included. Hypermethylated cytosine-phosphate-guanine probe sites (defined as β > 0.8) were more common among WTC-exposed versus unexposed participants (14.3% vs. 4.5%, respectively, among the top 5000 cytosine-phosphate-guanine sites). Cancer-related pathways (e.g., human papillomavirus infection, cGMP-PKG) were overrepresented in WTC-exposed groups (breast cancer patients and cancer-free subjects). Compared to the unexposed breast cancer patients, 47 epigenetically dysregulated genes were identified among WTC-exposed breast cancers. These genes formed a network, including Wnt/β-catenin signaling genes WNT4 and TCF7L2, and dysregulation of these genes contributes to cancer immune evasion. Conclusion WTC exposure likely impacts DNA methylation and may predispose exposed individuals toward cancer development, possibly through an immune-mediated mechanism.
Collapse
Affiliation(s)
- Stephanie Tuminello
- Department of Population Health, NYU Grossman School of Medicine, New York City, New York
| | - Yibeltal Arega Ashebir
- Department of Population Health, NYU Grossman School of Medicine, New York City, New York
| | - Chanel Schroff
- Department of Pathology, NYU Grossman School of Medicine, New York City, New York
| | - Sitharam Ramaswami
- Department of Pathology, NYU Grossman School of Medicine, New York City, New York
| | - Nedim Durmus
- Department of Medicine, NYU Grossman School of Medicine, New York City, New York
| | - Yu Chen
- Department of Population Health, NYU Grossman School of Medicine, New York City, New York
- NYU Perlmutter Comprehensive Cancer Center, New York City, New York
| | - Matija Snuderl
- Department of Pathology, NYU Grossman School of Medicine, New York City, New York
| | - Yongzhao Shao
- Department of Population Health, NYU Grossman School of Medicine, New York City, New York
- NYU Perlmutter Comprehensive Cancer Center, New York City, New York
| | - Joan Reibman
- Department of Medicine, NYU Grossman School of Medicine, New York City, New York
- Division of Environmental Medicine, Department of Medicine, NYU Grossman School of Medicine, New York City, New York
| | - Alan A. Arslan
- Department of Population Health, NYU Grossman School of Medicine, New York City, New York
- NYU Perlmutter Comprehensive Cancer Center, New York City, New York
- Department of Obstetrics and Gynecology, NYU Grossman School of Medicine, New York City, New York
| |
Collapse
|
2
|
Wen L, Liu Z, Zhou L, Liu Z, Li Q, Geng B, Xia Y. Bone and Extracellular Signal-Related Kinase 5 (ERK5). Biomolecules 2024; 14:556. [PMID: 38785963 PMCID: PMC11117709 DOI: 10.3390/biom14050556] [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: 03/26/2024] [Revised: 04/17/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
Bones are vital for anchoring muscles, tendons, and ligaments, serving as a fundamental element of the human skeletal structure. However, our understanding of bone development mechanisms and the maintenance of bone homeostasis is still limited. Extracellular signal-related kinase 5 (ERK5), a recently identified member of the mitogen-activated protein kinase (MAPK) family, plays a critical role in the pathogenesis and progression of various diseases, especially neoplasms. Recent studies have highlighted ERK5's significant role in both bone development and bone-associated pathologies. This review offers a detailed examination of the latest research on ERK5 in different tissues and diseases, with a particular focus on its implications for bone health. It also examines therapeutic strategies and future research avenues targeting ERK5.
Collapse
Affiliation(s)
- Lei Wen
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- Department of Orthopedics and Trauma Surgery, Affiliated Hospital of Yunnan University, Kunming 650032, China
| | - Zirui Liu
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Libo Zhou
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Zhongcheng Liu
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Qingda Li
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Bin Geng
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| | - Yayi Xia
- Department of Orthopedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; (L.W.); (Z.L.); (L.Z.); (Z.L.); (Q.L.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
| |
Collapse
|
3
|
Alharbi B, Alnajjar LI, Alhassan HH, Khan S, Jawaid T, Abdullaev BS, Alshammari N, Yadav DK, Adnan M, Shamsi A. Identification of mitogen-activated protein kinase 7 inhibitors from natural products: Combined virtual screening and dynamic simulation studies. J Mol Recognit 2024; 37:e3067. [PMID: 37956676 DOI: 10.1002/jmr.3067] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/22/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023]
Abstract
Mitogen-activated protein kinase 7 (MAPK7) is a serine/threonine protein kinase that belongs to the MAPK family and plays a vital role in various cellular processes such as cell proliferation, differentiation, gene transcription, apoptosis, metabolism, and cell survival. The elevated expression of MAPK7 has been associated with the onset and progression of multiple aggressive tumors in humans, underscoring the potential of targeting MAPK7 pathways in therapeutic research. This pursuit holds promise for the advancement of anticancer drug development by developing potential MAPK7 inhibitors. To look for potential MAPK7 inhibitors, we exploited structure-based virtual screening of natural products from the ZINC database. First, the Lipinski rule of five criteria was used to filter a large library of ~90,000 natural compounds, followed by ADMET and pan-assay interference compounds (PAINS) filters. Then, top hits were chosen based on their strong binding affinity as determined by molecular docking. Further, interaction analysis was performed to find effective and specific compounds that can precisely bind to the binding pocket of MAPK7. Consequently, two compounds, ZINC12296700 and ZINC02123081, exhibited significant binding affinity and demonstrated excellent drug-like properties. All-atom molecular dynamics simulations for 200 ns confirmed the stability of MAPK7-ZINC12296700 and MAPK7-ZINC02123081 docked complexes. According to the molecular mechanics Poisson-Boltzmann surface area investigation, the binding affinities of both complexes were considerable. Overall, the result suggests that ZINC12296700 and ZINC02123081 might be used as promising leads to develop novel MAPK7 inhibitors. Since these compounds would interfere with the kinase activity of MAPK7, therefore, may be implemented to control cell growth and proliferation in cancer after required validations.
Collapse
Affiliation(s)
- Bandar Alharbi
- Department of Medical Laboratory Science, College of Applied Medical Sciences, University of Hail, Hail, Saudi Arabia
| | - Lina I Alnajjar
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Hassan H Alhassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Shama Khan
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Science, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Talha Jawaid
- Department of Pharmacology, College of Medicine, Al Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Bekhzod S Abdullaev
- Department of Strategic Development, Innovation and Research, New Uzbekistan University, Tashkent, Uzbekistan
- Department of Oncology, School of Medicine, Central Asian University, Tashkent, Uzbekistan
| | - Nawaf Alshammari
- Department of Biology, College of Science, University of Ha'il, Ha'il, Saudi Arabia
| | - Dharmendra Kumar Yadav
- College of Pharmacy, Gachon University of Medicine and Science, Incheon, South Korea
- Arontier Co., Seoul, Republic of Korea
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, Ha'il, Saudi Arabia
| | - Anas Shamsi
- Center for Medical and Bio-Allied Health Sciences Research Research, Ajman University, Ajman, United Arab Emirates
| |
Collapse
|
4
|
Hwang J, Moon H, Kim H, Kim KY. Identification of a Novel ERK5 (MAPK7) Inhibitor, MHJ-627, and Verification of Its Potent Anticancer Efficacy in Cervical Cancer HeLa Cells. Curr Issues Mol Biol 2023; 45:6154-6169. [PMID: 37504304 PMCID: PMC10377775 DOI: 10.3390/cimb45070388] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 07/29/2023] Open
Abstract
Extracellular signal-regulated kinase 5 (ERK5), a member of the mitogen-activated protein kinase (MAPK) family, is involved in key cellular processes. However, overexpression and upregulation of ERK5 have been reported in various cancers, and ERK5 is associated with almost every biological characteristic of cancer cells. Accordingly, ERK5 has become a novel target for the development of anticancer drugs as inhibition of ERK5 shows suppressive effects of the deleterious properties of cancer cells. Herein, we report the synthesis and identification of a novel ERK5 inhibitor, MHJ-627, and verify its potent anticancer efficacy in a yeast model and the cervical cancer HeLa cell line. MHJ-627 successfully inhibited the kinase activity of ERK5 (IC50: 0.91 μM) and promoted the mRNA expression of tumor suppressors and anti-metastatic genes. Moreover, we observed significant cancer cell death, accompanied by a reduction in mRNA levels of the cell proliferation marker, proliferating cell nuclear antigen (PCNA), following ERK5 inhibition due to MHJ-627 treatment. We expect this finding to serve as a lead compound for further identification of inhibitors for ERK5-directed novel approaches for oncotherapy with increased specificity.
Collapse
Affiliation(s)
- Jeonghye Hwang
- Department of Genetics and Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Hyejin Moon
- Department of Applied Chemistry, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Hakwon Kim
- Department of Applied Chemistry, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Ki-Young Kim
- Department of Genetics and Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, Republic of Korea
| |
Collapse
|
5
|
Li M, Liu X. Pitavastatin maintains MAPK7 expression and alleviates angiotensin II-induced vascular endothelial cell inflammation and injury. Exp Ther Med 2022; 23:132. [PMID: 35069813 PMCID: PMC8756435 DOI: 10.3892/etm.2021.11055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/25/2021] [Indexed: 12/13/2022] Open
Abstract
Statins have been reported to suppress vascular remodeling in rats with spontaneous hypertension. However, the possible effects of the statin pitavastatin on hypertension-induced endothelial inflammation and injury remain to be fully elucidated. The present study aimed to evaluate the effects of pitavastatin on HUVEC injury and inflammation. HUVECs were exposed to angiotensin (Ang) II with or without pitavastatin co-treatment, after which MAPK7 expression was detected via reverse transcription-quantitative PCR and western blotting. MAPK7 expression was additionally silenced in HUVECs via transfection with short hairpin RNA, followed by Ang II treatment with or without pitavastatin. Cell viability, inflammation, reactive oxygen species (ROS) production, nitric oxide (NO) production and cell apoptosis were then measured by using Cell Conting Kit-8, ELISA, commercial corresponding kits and TUNEL staining, respectively. Western blotting was also used to determine the protein expression of endothelial NO synthase and endothelin 1, and the proteins involved in apoptosis. Results of the present study demonstrated that the expression levels of MAPK7 in Ang II-induced endothelial cells were decreased, which was reversed following treatment with pitavastatin. Pitavastatin reversed the Ang II-induced reduction in cell viability and reversed the Ang II-induced increase in inflammatory factor and ROS levels and apoptosis in HUVECs by activating MAPK7. Treatment with pitavastatin also increased the production of NO in addition to increasing the expression of endothelial NO synthase and endothelin-1 in Ang II-induced HUVECs through MAPK7 activation. Collectively, results from the present study demonstrated that treatment with pitavastatin preserves MAPK7 expression to alleviate Ang II-induced vascular endothelial cell inflammation and injury. Therefore, findings of the present study may help to elucidate the mechanisms underlying the effects of pitavastatin on vascular endothelial cell inflammation and injury.
Collapse
Affiliation(s)
- Min Li
- Department of Outpatient Nursing, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, P.R. China
| | - Xiaohua Liu
- Department of Nursing, Chengdu Shuangliu District Maternal and Child Health Hospital, Chengdu, Sichuan 610200, P.R. China
| |
Collapse
|
6
|
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: 6] [Impact Index Per Article: 2.0] [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
|
7
|
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
|
8
|
Sánchez-Fdez A, Re-Louhau MF, Rodríguez-Núñez P, Ludeña D, Matilla-Almazán S, Pandiella A, Esparís-Ogando A. Clinical, genetic and pharmacological data support targeting the MEK5/ERK5 module in lung cancer. NPJ Precis Oncol 2021; 5:78. [PMID: 34404896 PMCID: PMC8371118 DOI: 10.1038/s41698-021-00218-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 07/20/2021] [Indexed: 11/09/2022] Open
Abstract
Despite advances in its treatment, lung cancer still represents the most common and lethal tumor. Because of that, efforts to decipher the pathophysiological actors that may promote lung tumor generation/progression are being made, with the final aim of establishing new therapeutic options. Using a transgenic mouse model, we formerly demonstrated that the sole activation of the MEK5/ERK5 MAPK route had a pathophysiological role in the onset of lung adenocarcinomas. Given the prevalence of that disease and its frequent dismal prognosis, our findings opened the possibility of targeting the MEK5/ERK5 route with therapeutic purposes. Here we have explored such possibility. We found that increased levels of MEK5/ERK5 correlated with poor patient prognosis in lung cancer. Moreover, using genetic as well as pharmacological tools, we show that targeting the MEK5/ERK5 route is therapeutically effective in lung cancer. Not only genetic disruption of ERK5 by CRISPR/Cas9 caused a relevant inhibition of tumor growth in vitro and in vivo; such ERK5 deficit augmented the antitumoral effect of agents normally used in the lung cancer clinic. The clinical correlation studies together with the pharmacological and genetic results establish the basis for considering the targeting of the MEK5/ERK5 route in the therapy for lung cancer.
Collapse
Affiliation(s)
- Adrián Sánchez-Fdez
- Institute of Molecular and Cellular Biology of Cancer (IBMCC)-CSIC, Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Cancer Network Research (CIBERONC), Salamanca, Spain
| | - María Florencia Re-Louhau
- Institute of Molecular and Cellular Biology of Cancer (IBMCC)-CSIC, Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Pablo Rodríguez-Núñez
- Institute of Molecular and Cellular Biology of Cancer (IBMCC)-CSIC, Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Dolores Ludeña
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Pathology Service, University Hospital, Salamanca, Spain
| | - Sofía Matilla-Almazán
- Institute of Molecular and Cellular Biology of Cancer (IBMCC)-CSIC, Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Cancer Network Research (CIBERONC), Salamanca, Spain
| | - Atanasio Pandiella
- Institute of Molecular and Cellular Biology of Cancer (IBMCC)-CSIC, Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Cancer Network Research (CIBERONC), Salamanca, Spain
| | - Azucena Esparís-Ogando
- Institute of Molecular and Cellular Biology of Cancer (IBMCC)-CSIC, Salamanca, Spain. .,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain. .,Cancer Network Research (CIBERONC), Salamanca, Spain.
| |
Collapse
|
9
|
Sreedurgalakshmi K, Srikar R, Rajkumari R. CRISPR-Cas deployment in non-small cell lung cancer for target screening, validations, and discoveries. Cancer Gene Ther 2020; 28:566-580. [PMID: 33191402 DOI: 10.1038/s41417-020-00256-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/14/2020] [Accepted: 10/29/2020] [Indexed: 12/24/2022]
Abstract
Continued advancements in CRISPR-Cas systems have accelerated genome research. Use of CRISPR-Cas in cancer research has been of great interest that is resulting in development of orthogonal methods for drug target validations and discovery of new therapeutic targets through genome-wide screens of cancer cells. CRISPR-based screens have also revealed several new cancer drivers through alterations in tumor suppressor genes (TSGs) and oncogenes inducing resistance to targeted therapies via activation of alternate signaling pathways. Given such dynamic status of cancer, we review the application of CRISPR-Cas in non-small cell lung cancer (NSCLC) for development of mutant models, drug screening, target validation, novel target discoveries, and other emerging potential applications. In addition, CRISPR-based approach for development of novel anticancer combination therapies is also discussed in this review.
Collapse
Affiliation(s)
- K Sreedurgalakshmi
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamilnadu, India.,Division of Biosimilars and Gene Therapy, R&D, Levim Biotech LLP, Chennai, Tamilnadu, India
| | - R Srikar
- Division of Biosimilars and Gene Therapy, R&D, Levim Biotech LLP, Chennai, Tamilnadu, India.
| | - Reena Rajkumari
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamilnadu, India.
| |
Collapse
|
10
|
Proteomic analysis reveals the molecular mechanism of Hippophae rhamnoides polysaccharide intervention in LPS-induced inflammation of IPEC-J2 cells in piglets. Int J Biol Macromol 2020; 164:3294-3304. [PMID: 32888998 DOI: 10.1016/j.ijbiomac.2020.08.235] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/17/2020] [Accepted: 08/30/2020] [Indexed: 01/15/2023]
Abstract
Early weaning can cause intestinal disorders and dysfunction in piglets, and may induce intestinal diseases. Hippophae rhamnoides polysaccharide (HRP) has anti-inflammatory and immune promotion function. However, few studies have explored the change of differentially protein expression by lipopolysaccharide (LPS)-induced porcine intestinal epithelial cell (IPEC-J2) after HRP pre-treatment. In this study, proteomic analysis was used to explore the essential proteins and immune-related pathways that can be regulated by LPS-induced IPEC-J2 cells after HRP pre-treatment. The results indicate that by searching the Sus scrofa database, a total of 18,768 proteins was identified. Among recognized proteins, there are 2052 (1720 up-regulated and 332 down-regulated), 358 (262 up-regulated and 96 down-regulated) and1532 (314 up-regulated and 1218 down-regulated) proteins expressed differently in C vs. L, C vs. H6-L and L vs. H6-L, respectively. The Cluster of Orthologous Groups (COG) analysis divided the identified proteins into 23 categories. Gene Ontology (GO) enrichment analysis revealed that cellular process, cell, cell part, organelle and binding were the most enriched pathways for differentially expressed proteins. KEGG enrichment analysis indicated that the top 20 pathways in the L-vs-H6-L group related to immunity were the Tight junction, MAPK signaling pathway, PI3K-Akt signaling pathway, rap1 signaling pathway, HIF-1 signaling pathway, Ras signaling pathway and Fc gamma R-mediated phagocytosis. Moreover, we also found 42 key proteins related to these immune pathways in this study. Additionally, western blotting analyses confirmed that LPS reduced the levels of claudin2 (CLDN2), insulin-like growth factor 2 (IGF2) and increased phosphorylated mitogen-activated protein kinase 7 (MAPK7), phosphorylated transcription factor p65 (RELA), phosphorylated nuclear factor NF-kappa-B p105 subunit (NF-κB1) and phosphorylated nuclear factor of kappa light polypeptide gene enhancer in B-cells 2 (NF-κB2). Pre-treatment with HRP increased the levels of CLDN2, IGF2 and reduced the levels of the phosphorylated MAPK7, phosphorylated RELA, phosphorylated NF-κB1 and phosphorylated NF-κB2 in cells. These results also showed that HRP alleviated LPS-induced inflammation in IPEC-J2 cells by inhibiting the MAPK/NF-κB signaling pathway and its related differentially expressed proteins.
Collapse
|
11
|
Hoang VT, Matossian MD, Ucar DA, Elliott S, La J, Wright MK, Burks HE, Perles A, Hossain F, King CT, Browning VE, Bursavich J, Fang F, Del Valle L, Bhatt AB, Cavanaugh JE, Flaherty PT, Anbalagan M, Rowan BG, Bratton MR, Nephew KP, Miele L, Collins-Burow BM, Martin EC, Burow ME. ERK5 Is Required for Tumor Growth and Maintenance Through Regulation of the Extracellular Matrix in Triple Negative Breast Cancer. Front Oncol 2020; 10:1164. [PMID: 32850332 PMCID: PMC7416559 DOI: 10.3389/fonc.2020.01164] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 06/09/2020] [Indexed: 12/16/2022] Open
Abstract
Conventional mitogen-activated protein kinase (MAPK) family members regulate diverse cellular processes involved in tumor initiation and progression, yet the role of ERK5 in cancer biology is not fully understood. Triple-negative breast cancer (TNBC) presents a clinical challenge due to the aggressive nature of the disease and a lack of targeted therapies. ERK5 signaling contributes to drug resistance and metastatic progression through distinct mechanisms, including activation of epithelial-to-mesenchymal transition (EMT). More recently a role for ERK5 in regulation of the extracellular matrix (ECM) has been proposed, and here we investigated the necessity of ERK5 in TNBC tumor formation. Depletion of ERK5 expression using the CRISPR/Cas9 system in MDA-MB-231 and Hs-578T cells resulted in loss of mesenchymal features, as observed through gene expression profile and cell morphology, and suppressed TNBC cell migration. In vivo xenograft experiments revealed ERK5 knockout disrupted tumor growth kinetics, which was restored using high concentration Matrigel™ and ERK5-ko reduced expression of the angiogenesis marker CD31. These findings implicated a role for ERK5 in the extracellular matrix (ECM) and matrix integrity. RNA-sequencing analyses demonstrated downregulation of matrix-associated genes, integrins, and pro-angiogenic factors in ERK5-ko cells. Tissue decellularization combined with cryo-SEM and interrogation of biomechanical properties revealed that ERK5-ko resulted in loss of key ECM fiber alignment and mechanosensing capabilities in breast cancer xenografts compared to parental wild-type cells. In this study, we identified a novel role for ERK5 in tumor growth kinetics through modulation of the ECM and angiogenesis axis in breast cancer.
Collapse
Affiliation(s)
- Van T. Hoang
- Section of Hematology & Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Margarite D. Matossian
- Section of Hematology & Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Deniz A. Ucar
- Department of Genetics, Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Steven Elliott
- Section of Hematology & Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Jacqueline La
- Section of Hematology & Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Maryl K. Wright
- Section of Hematology & Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Hope E. Burks
- Section of Hematology & Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Aaron Perles
- Section of Hematology & Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Fokhrul Hossain
- Department of Genetics, Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Connor T. King
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Valentino E. Browning
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Jacob Bursavich
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Fang Fang
- Medical Sciences, School of Medicine, Indiana University Bloomington, Bloomington, IN, United States
| | - Luis Del Valle
- Department of Pathology, Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Akshita B. Bhatt
- Department of Pharmacology, School of Pharmacy, Duquesne University, Pittsburgh, PA, United States
| | - Jane E. Cavanaugh
- Department of Pharmacology, School of Pharmacy, Duquesne University, Pittsburgh, PA, United States
| | - Patrick T. Flaherty
- Department of Medicinal Chemistry, School of Pharmacy, Duquesne University, Pittsburgh, PA, United States
| | - Muralidharan Anbalagan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Brian G. Rowan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Melyssa R. Bratton
- Cellular and Molecular Biology Core, Xavier University, New Orleans, LA, United States
| | - Kenneth P. Nephew
- Medical Sciences, School of Medicine, Indiana University Bloomington, Bloomington, IN, United States
| | - Lucio Miele
- Department of Genetics, Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Bridgette M. Collins-Burow
- Section of Hematology & Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
- Tulane Cancer Center, New Orleans, LA, United States
| | - Elizabeth C. Martin
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Matthew E. Burow
- Section of Hematology & Medical Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
12
|
Bucher-Johannessen C, Page CM, Haugen TB, Wojewodzic MW, Fosså SD, Grotmol T, Haugnes HS, Rounge TB. Cisplatin treatment of testicular cancer patients introduces long-term changes in the epigenome. Clin Epigenetics 2019; 11:179. [PMID: 31796056 PMCID: PMC6892132 DOI: 10.1186/s13148-019-0764-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 10/15/2019] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Cisplatin-based chemotherapy (CBCT) is part of standard treatment of several cancers. In testicular cancer (TC) survivors, an increased risk of developing metabolic syndrome (MetS) is observed. In this epigenome-wide association study, we investigated if CBCT relates to epigenetic changes (DNA methylation) and if epigenetic changes render individuals susceptible for developing MetS later in life. We analyzed methylation profiles, using the MethylationEPIC BeadChip, in samples collected ~ 16 years after treatment from 279 Norwegian TC survivors with known MetS status. Among the CBCT treated (n = 176) and non-treated (n = 103), 61 and 34 developed MetS, respectively. We used two linear regression models to identify if (i) CBCT results in epigenetic changes and (ii) epigenetic changes play a role in development of MetS. Then we investigated if these changes in (i) and (ii) links to genes, functional networks, and pathways related to MetS symptoms. RESULTS We identified 35 sites that were differentially methylated when comparing CBCT treated and untreated TC survivors. The PTK6-RAS-MAPk pathway was significantly enriched with these sites and infers a gene network of 13 genes with CACNA1D (involved in insulin release) as a network hub. We found nominal MetS-associations and a functional gene network with ABCG1 and NCF2 as network hubs. CONCLUSION Our results suggest that CBCT has long-term effects on the epigenome. We could not directly link the CBCT effects to the risk of developing MetS. Nevertheless, since we identified differential methylation occurring in genes associated with conditions pertaining to MetS, we hypothesize that epigenomic changes may also play a role in the development of MetS in TC survivors. Further studies are needed to validate this hypothesis.
Collapse
Affiliation(s)
| | - Christian M Page
- Oslo Centre for Biostatistics and Epidemiology, Section for Research Support, Oslo University Hospital, Oslo, Norway.,Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Trine B Haugen
- Faculty of Health Sciences, OsloMet - Oslo Metropolitan University, Oslo, Norway
| | | | - Sophie D Fosså
- Department of Research, Cancer Registry of Norway, Oslo, Norway.,Department of Oncology, The Norwegian Radium Hospital/Oslo University Hospital, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Tom Grotmol
- Department of Research, Cancer Registry of Norway, Oslo, Norway
| | - Hege S Haugnes
- Department of Oncology, University Hospital of North Norway, Tromsø, Norway.,Institute of Clinical Medicine, UIT The Arctic University of Norway, Tromsø, Norway
| | - Trine B Rounge
- Department of Research, Cancer Registry of Norway, Oslo, Norway. .,Department of Informatics, University of Oslo, Oslo, Norway.
| |
Collapse
|
13
|
Hou Y, Feng H, Jiao J, Qian L, Sun B, Chen P, Li Q, Liang Z. Mechanism of miR-143-3p inhibiting proliferation, migration and invasion of osteosarcoma cells by targeting MAPK7. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2065-2071. [PMID: 31126193 DOI: 10.1080/21691401.2019.1620252] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Objective: To investigate the effects of miR-143-3p and MAPK7 on the proliferation, migration, and invasion of U2OS human osteosarcoma (OS) cells. Methods: The expression of miR-143-3p and MAPK7 in U2OS cells were detected by qRT-PCR, and the protein level of MAPK7 was measured by western blot assay. The targeting relationship between miR-143-3p and MAPK7 was predicted by TargetScan and verified by dual luciferase reporter assay. MTT and Transwell assays were used to detect cell viability, migrated cells and invaded cells of U2OS cells. Results: Compared with hFOB1.19 cells, the expression of miR-143-3p was down-regulated and MAPK7 was up-regulated in U2OS cells. Cell viability, migration and invasion ability significantly decreased induced by miR-143-3p overexpression or MAPK7 knockdown in U2OS cells. The results of dual luciferase reporter assay indicated that miR-143-3p interacted with MAPK7. Furthermore, overexpression of MAPK7 could reverse the inhibitory effects on cell proliferation, migration and invasion in U2OS cells induced by miR-143-3p mimics. Conclusion: miR-143-3p could inhibit proliferation, migration and invasion of U2OS cells by targeting MAPK7.
Collapse
Affiliation(s)
- Yu Hou
- a Department of Orthopedics, Affiliated Hospital of Hebei University , Baoding , Hebei , China
| | - Helin Feng
- b Department of Orthopedics, Fourth Hospital of Hebei Medical University , Shijiazhuang , Hebei , China
| | - Jianbao Jiao
- a Department of Orthopedics, Affiliated Hospital of Hebei University , Baoding , Hebei , China
| | - Ligang Qian
- a Department of Orthopedics, Affiliated Hospital of Hebei University , Baoding , Hebei , China
| | - Bo Sun
- a Department of Orthopedics, Affiliated Hospital of Hebei University , Baoding , Hebei , China
| | - Pingtao Chen
- a Department of Orthopedics, Affiliated Hospital of Hebei University , Baoding , Hebei , China
| | - Qinggui Li
- a Department of Orthopedics, Affiliated Hospital of Hebei University , Baoding , Hebei , China
| | - Zhixing Liang
- a Department of Orthopedics, Affiliated Hospital of Hebei University , Baoding , Hebei , China
| |
Collapse
|
14
|
Xue Z, Wang J, Yu W, Li D, Zhang Y, Wan F, Kou X. Biochanin A protects against PM 2.5-induced acute pulmonary cell injury by interacting with the target protein MEK5. Food Funct 2019; 10:7188-7203. [PMID: 31608342 DOI: 10.1039/c9fo01382b] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Epidemiological studies have shown that exposure to ambient fine particulate matter (PM2.5) is associated with an increased risk for cardiopulmonary diseases. The MEK5/ERK5 and NF-κB signaling pathways are closely related to the regulation of acute pulmonary cell injury (APCI) and may play an important role in the underlying pathophysiological mechanisms. Related studies have shown that Biochanin A (BCA) effectively interferes with APCI, but the underlying mechanism through which this occurs is not fully understood. Previously, based on proteomic and bioinformatic research, we found the indispensable role of MEK5 in mediating remission effects of BCA against PM2.5-induced lung toxicity. Therefore, using A549 adenocarcinoma human alveolar basal epithelial cells (A549 cells), we combined western blot and qRT-PCR to study the protective signaling pathways induced by BCA, indicating that MEK5/ERK5 and NF-κB are both involved in mediating APCI in response to PM2.5, and MEK5/ERK5 positively activated NF-κB and its downstream cellular regulatory factors. BCA significantly suppressed PM2.5-induced upregulation of MEK5/ERK5 expression and phosphorylation and activation of NF-κB. Furthermore, due to the specificity of the MEK5/ERK5 protein structure, the binding sites and binding patterns of BCA and MEK5 were analyzed using molecular docking correlation techniques, which showed that there are stable hydrogen bonds between BCA and the PB1 domain of MEK5 as well as its kinase domain. BCA forms a stable complex with MEK5, which has potential effects on MEKK2/3-MEK5-ERK5 ternary interactions, p62/αPKC-mediated NF-κB regulation, and inhibition of MEK5 target protein phosphorylation. Therefore, our study suggests that MEK5 is an important regulator of intracellular signaling of APCI in response to PM2.5 exposure. BCA may exert anti-APCI activity by targeting MEK5 to inhibit activation of the MEK5/ERK5/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Zhaohui Xue
- Department of Food Science, School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China.
| | - Junyu Wang
- Department of Food Science, School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China.
| | - Wancong Yu
- Tianjin Academy of Agricultural Science, 300381, Tianjin, China
| | - Dan Li
- Department of Food Science, School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China.
| | - Yixia Zhang
- Department of Food Science, School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China.
| | - Fang Wan
- Department of Food Science, School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China.
| | - Xiaohong Kou
- Department of Food Science, School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China.
| |
Collapse
|
15
|
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
|
16
|
Wang Y, Wang G, Ma Y, Teng J, Wang Y, Cui Y, Dong Y, Shao S, Zhan Q, Liu X. FAT1, a direct transcriptional target of E2F1, suppresses cell proliferation, migration and invasion in esophageal squamous cell carcinoma. Chin J Cancer Res 2019; 31:609-619. [PMID: 31564804 PMCID: PMC6736659 DOI: 10.21147/j.issn.1000-9604.2019.04.05] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Objective Growing evidence indicates that FAT atypical cadherin 1 (FAT1) has aberrant genetic alterations and exhibits potential tumor suppressive function in esophageal squamous cell carcinoma (ESCC). However, the role of FAT1 in ESCC tumorigenesis remains not well elucidated. The aim of this study was to further investigate genetic alterations and biological functions of FAT1, as well as to explore its transcriptional regulation and downstream targets in ESCC. Methods The mutations of FAT1 in ESCC were achieved by analyzing a combined study from seven published genomic data, while the copy number variants of FAT1 were obtained from an analysis of our previous data as well as of The Cancer Genome Atlas (TCGA) and Cancer Cell Line Encyclopedia (CCLE) databases using the cBioPortal. The transcriptional regulation of FAT1 expression was investigated by chromatin immunoprecipitation (ChIP) and the luciferase reporter assays. In-cell western, Western blot and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were used to assess the indicated gene expression. In addition, colony formation and Transwell migration/invasion assays were employed to test cell proliferation, migration and invasion. Finally, RNA sequencing was used to study the transcriptomes. Results FAT1 was frequently mutated in ESCC and was deleted in multiple cancers. Furthermore, the transcription factor E2F1 occupied the promoter region of FAT1, and depletion of E2F1 led to a decrease in transcription activity and mRNA levels of FAT1. Moreover, we found that knockdown of FAT1 promoted KYSE30 and KYSE150 cell proliferation, migration and invasion; while overexpression of FAT1 inhibited KYSE30 and KYSE410 cell proliferation, migration and invasion. In addition, knockdown of FAT1 led to enrichment of the mitogen-activated protein kinase (MAPK) signaling pathway and cell adhesion process. Conclusions Our data provided evidence for the tumor suppressive function of FAT1 in ESCC cells and elucidated the transcriptional regulation of FAT1 by E2F1, which may facilitate the understanding of molecular mechanisms of the progression of ESCC.
Collapse
Affiliation(s)
- Yu Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Guangchao Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yunping Ma
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Jinglei Teng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Yan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yongping Cui
- Shenzhen Peking University-The Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Yan Dong
- College of Stomatology, Dalian Medical University, Dalian 116044, China
| | - Shujuan Shao
- Key Laboratory of Proteomics, Dalian Medical University, Dalian 116044, China
| | - Qimin Zhan
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xuefeng Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
17
|
Ma Q, Tan Y, Chen X, Chen S, Sun Y, Zhou B. Regulation of the MAPK signaling pathway by miR-421-5p in rats under light pollution. Int J Mol Med 2018; 42:3329-3343. [PMID: 30221682 PMCID: PMC6202112 DOI: 10.3892/ijmm.2018.3874] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 09/11/2018] [Indexed: 01/14/2023] Open
Abstract
The present study aimed to explore the difference in the expression profiles of ovarian microRNA sequences in rats in a light pollution environment and rats in a normal light environment. Rats in the control group were exposed to 12‑h light/dark cycles, while rats in the model group were continuously exposed to 24‑h light. The ovaries were extracted from the two groups of rats, and Illumina HiSeq 2500 high‑throughput sequencing technology was used to detect the differences in microRNA (miRNA) expression among the two groups. Fluorescence quantitative reverse transcription‑polymerase chain reaction was used to verify the differential expression of miRNA. The present study was designed to experimentally validate the interaction between miR‑421‑5p and mitogen‑activated protein kinase (MAPK) 7 by using the dual‑luciferase reporter system, and to explore the expression of proteins in the MAPK signaling pathway with a lentiviral vector‑mediated small hairpin RNA interference against microRNA‑421‑5p. The expression of 45 miRNAs was significantly different. In total, 13 miRNAs were upregulated, of which 5 miRNA sequences were known and 8 were predicted. Furthermore, 32 miRNAs were downregulated, of which 11 miRNA sequences were known and 21 were predicted. The results of the luciferase reporter assay confirmed the targeting association between miR‑421‑5p and MAPK7. The expression levels of MAPK and genes in its downstream signaling pathways, including c‑Fos, CREB and c‑Myc, were downregulated when miR‑421‑5p was overexpressed and upregulated when miR‑421‑5p was silenced. The differential expression of miRNAs may serve an important role in the development of the ovary in a light pollution environment. miR‑421‑5p may regulate ovarian growth and development by targeting the MAPK signaling pathway in light polluted rat ovaries.
Collapse
Affiliation(s)
- Qianwen Ma
- Zhenjiang Hospital Affiliated to Nanjing University of Chinese Medicine, Zhenjiang Hospital of Traditional Chinese Medicine, Zhenjiang, Jiangsu 212000, P.R. China
| | - Yong Tan
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Xuan Chen
- Zhenjiang Hospital Affiliated to Nanjing University of Chinese Medicine, Zhenjiang Hospital of Traditional Chinese Medicine, Zhenjiang, Jiangsu 212000, P.R. China
| | - Shuping Chen
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Yuying Sun
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Beibei Zhou
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| |
Collapse
|
18
|
Bhushan A, Singh A, Kapur S, Borthakar BB, Sharma J, Rai AK, Kataki AC, Saxena S. Identification and Validation of Fibroblast Growth Factor 12 Gene as a Novel Potential Biomarker in Esophageal Cancer Using Cancer Genomic Datasets. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2018; 21:616-631. [PMID: 29049013 DOI: 10.1089/omi.2017.0116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) has a complex, multifactorial etiology in which environmental, geographical, and genetic factors play major roles. It is the second most common cancer among men and the fourth most common among women in India, with a particularly high prevalence in Northeast India. In this study, an integrative in silico [DAVID, NCG5.0, Oncomine, Cancer Cell Line Encyclopedia, and The Cancer Genome Atlas (TCGA)] approach was used to identify the potential biomarkers by using the available three genomic datasets on ESCC from Northeast India followed by its in vitro functional validation. Fibroblast Growth Factor 12 (FGF12) gene was overexpressed in ESCC. The upregulation of FGF12 was also observed on ESCC of TCGA OncoPrint portal, whereas very low expression of FGF12 gene was mapped in normal esophageal tissue on the GTEx database. Silencing of FGF12 showed significant inhibition in activity of tumor cell proliferation, colony formation, and cell migration. The upregulation of FGF12 showed significantly reduced survival in ESCC patients. The protein interaction analysis of FGF12 found the binding with MAPK8IP2 and MAPK13. High expression of FGF12 along with MAPK8IP2, and MAPK13 proteins correlate with poor survival in ESCC patients. Tissue microarray also showed expression of these proteins in patients with ESCC. These results indicate that FGF12 has a potential role in ESCC and suggest that cancer genomic datasets with application of in silico approaches are instrumental for biomarker discovery research broadly and specifically, for the identification of FGF12 as a putative biomarker in ESCC.
Collapse
Affiliation(s)
- Ashish Bhushan
- 1 National Institute of Pathology (ICMR) , New Delhi, India .,2 Faculty of Health and Biomedical Sciences, Symbiosis International University , Pune, India
| | - Avninder Singh
- 1 National Institute of Pathology (ICMR) , New Delhi, India
| | - Sujala Kapur
- 1 National Institute of Pathology (ICMR) , New Delhi, India
| | | | | | - Avdhesh K Rai
- 3 Dr. B. Borooah Cancer Institute (BBCI) , Guwahati, India
| | - Amal C Kataki
- 3 Dr. B. Borooah Cancer Institute (BBCI) , Guwahati, India
| | - Sunita Saxena
- 1 National Institute of Pathology (ICMR) , New Delhi, India
| |
Collapse
|
19
|
Dong X, Lv B, Li Y, Cheng Q, Su C, Yin G. MiR-143 regulates the proliferation and migration of osteosarcoma cells through targeting MAPK7. Arch Biochem Biophys 2017; 630:47-53. [PMID: 28734729 DOI: 10.1016/j.abb.2017.07.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 07/04/2017] [Accepted: 07/18/2017] [Indexed: 12/12/2022]
Abstract
Accumulating documents have been suggested that microRNA-143 (miR-143) function as a tumor suppressor, involved in many biological processes including tumor initiation and progression. However, the biological function and molecular mechanism of miR-143 in Osteosarcoma (OS) still remains to be further investigated. Despite many efforts have been made, the prognosis of OS is still unsatisfied. Thus, exploring the underlying mechanism of OS and finding new treatment targets is essential for improving the survival rate of OS patients. In our study, we determined the level of miR-143 in clinical OS tissues and cells, and explored its function and underlying mechanisms in the tumorigenesis of OS. Our findings revealed that miR-143 expression was significantly downregulated in OS tissues and cell lines. Gain-of-function assays indicated that forced expression of miR-143 in OS cells inhibited cell proliferation and migration/invasion. Bioinformatics and luciferase reporter assays confirmed that MAPK7 was targets gene of miR-143. The results of the present study indicated that miR-143 could be a potential target for treating OS.
Collapse
Affiliation(s)
- Xiancheng Dong
- Nanjing Medical University, Nanjing, PR China; Department of Orthopedics, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, PR China
| | - Bin Lv
- Nanjing Medical University, Nanjing, PR China
| | - Yusong Li
- Department of Orthopedics, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, PR China
| | - Qinghua Cheng
- Department of Orthopedics, People's Hospital of Lishui District in Nanjing City, Nanjing, PR China
| | - Chuan Su
- Nanjing Medical University, Nanjing, PR China; Department of Pathogen Biology & Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, PR China.
| | - Guoyong Yin
- Nanjing Medical University, Nanjing, PR China; Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China.
| |
Collapse
|
20
|
Loveridge CJ, Mui EJ, Patel R, Tan EH, Ahmad I, Welsh M, Galbraith J, Hedley A, Nixon C, Blyth K, Sansom O, Leung HY. Increased T-cell Infiltration Elicited by Erk5 Deletion in a Pten-Deficient Mouse Model of Prostate Carcinogenesis. Cancer Res 2017; 77:3158-3168. [PMID: 28515147 PMCID: PMC5474317 DOI: 10.1158/0008-5472.can-16-2565] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/09/2016] [Accepted: 04/12/2017] [Indexed: 12/13/2022]
Abstract
Prostate cancer does not appear to respond to immune checkpoint therapies where T-cell infiltration may be a key limiting factor. Here, we report evidence that ablating the growth regulatory kinase Erk5 can increase T-cell infiltration in an established Pten-deficient mouse model of human prostate cancer. Mice that were doubly mutant in prostate tissue for Pten and Erk5 (prostate DKO) exhibited a markedly increased median survival with reduced tumor size and proliferation compared with control Pten-mutant mice, the latter of which exhibited increased Erk5 mRNA expression. A comparative transcriptomic analysis revealed upregulation in prostate DKO mice of the chemokines Ccl5 and Cxcl10, two potent chemoattractants for T lymphocytes. Consistent with this effect, we observed a relative increase in a predominantly CD4+ T-cell infiltrate in the prostate epithelial and stroma of tumors from DKO mice. Collectively, our results offer a preclinical proof of concept for ERK5 as a target to enhance T-cell infiltrates in prostate cancer, with possible implications for leveraging immune therapy in this disease. Cancer Res; 77(12); 3158-68. ©2017 AACR.
Collapse
Affiliation(s)
- Carolyn J Loveridge
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden, Glasgow, United Kingdom
- CRUK Beatson Institute, Bearsden, Glasgow, United Kingdom
| | - Ernest J Mui
- CRUK Beatson Institute, Bearsden, Glasgow, United Kingdom
| | - Rachana Patel
- CRUK Beatson Institute, Bearsden, Glasgow, United Kingdom
| | - Ee Hong Tan
- CRUK Beatson Institute, Bearsden, Glasgow, United Kingdom
| | - Imran Ahmad
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden, Glasgow, United Kingdom
- CRUK Beatson Institute, Bearsden, Glasgow, United Kingdom
| | - Michelle Welsh
- CRUK Beatson Institute, Bearsden, Glasgow, United Kingdom
| | - Julie Galbraith
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden, Glasgow, United Kingdom
| | - Ann Hedley
- CRUK Beatson Institute, Bearsden, Glasgow, United Kingdom
| | - Colin Nixon
- CRUK Beatson Institute, Bearsden, Glasgow, United Kingdom
| | - Karen Blyth
- CRUK Beatson Institute, Bearsden, Glasgow, United Kingdom
| | - Owen Sansom
- CRUK Beatson Institute, Bearsden, Glasgow, United Kingdom
| | - Hing Y Leung
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden, Glasgow, United Kingdom.
- CRUK Beatson Institute, Bearsden, Glasgow, United Kingdom
| |
Collapse
|
21
|
Zhao XK, Mao YM, Meng H, Song X, Hu SJ, Lv S, Cheng R, Zhang TJ, Han XN, Ren JL, Qi YJ, Wang LD. Shared susceptibility loci at 2q33 region for lung and esophageal cancers in high-incidence areas of esophageal cancer in northern China. PLoS One 2017; 12:e0177504. [PMID: 28542283 PMCID: PMC5436667 DOI: 10.1371/journal.pone.0177504] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 04/30/2017] [Indexed: 11/19/2022] Open
Abstract
Background Cancers from lung and esophagus are the leading causes of cancer-related deaths in China and share many similarities in terms of histological type, risk factors and genetic variants. Recent genome-wide association studies (GWAS) in Chinese esophageal cancer patients have demonstrated six high-risk candidate single nucleotide polymorphisms (SNPs). Thus, the present study aimed to determine the risk of these SNPs predisposing to lung cancer in Chinese population. Methods A total of 1170 lung cancer patients and 1530 normal subjects were enrolled in this study from high-incidence areas for esophageal cancer in Henan, northern China. Five milliliters of blood were collected from all subjects for genotyping. Genotyping of 20 high-risk SNP loci identified from genome-wide association studies (GWAS) on esophageal, lung and gastric cancers was performed using TaqMan allelic discrimination assays. Polymorphisms were examined for deviation from Hardy-Weinberg equilibrium (HWE) using Х2 test. Bonferroni correction was performed to correct the statistical significance of 20 SNPs with the risk of lung cancer. The Pearson’s Х2 test was used to compare the distributions of gender, TNM stage, histopathological type, smoking and family history by lung susceptibility genotypes. Kaplan-Meier and Cox regression analyses were carried out to evaluate the associations between genetic variants and overall survival. Results Four of the 20 SNPs identified as high-risk SNPs in Chinese esophageal cancer showed increased risk for Chinese lung cancer, which included rs3769823 (OR = 1.26; 95% CI = 1.107–1.509; P = 0.02), rs10931936 (OR = 1.283; 95% CI = 1.100–1.495; P = 0.04), rs2244438 (OR = 1.294; 95% CI = 1.098–1.525; P = 0.04) and rs13016963 (OR = 1.268; 95% CI = 1.089–1.447; P = 0.04). All these SNPs were located at 2q33 region harboringgenes of CASP8, ALS2CR12 and TRAK2. However, none of these susceptibility SNPs was observed to be significantly associated with gender, TNM stage, histopathological type, smoking, family history and overall survival. Conclusions The present study identified four high-risk SNPs at 2q33 locus for Chinese lung cancer and demonstrated the shared susceptibility loci at 2q33 region for Chinese lung and esophageal cancers.
Collapse
Affiliation(s)
- Xue Ke Zhao
- Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Min Mao
- Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hui Meng
- Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xin Song
- Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shou Jia Hu
- Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuang Lv
- Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Rang Cheng
- Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tang Juan Zhang
- Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xue Na Han
- Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jing Li Ren
- Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Jun Qi
- Henan Key Laboratory of Cancer Epigenetic, Cancer Institute, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Li Dong Wang
- Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- * E-mail:
| |
Collapse
|
22
|
Zhang X, Maity T, Kashyap MK, Bansal M, Venugopalan A, Singh S, Awasthi S, Marimuthu A, Charles Jacob HK, Belkina N, Pitts S, Cultraro CM, Gao S, Kirkali G, Biswas R, Chaerkady R, Califano A, Pandey A, Guha U. Quantitative Tyrosine Phosphoproteomics of Epidermal Growth Factor Receptor (EGFR) Tyrosine Kinase Inhibitor-treated Lung Adenocarcinoma Cells Reveals Potential Novel Biomarkers of Therapeutic Response. Mol Cell Proteomics 2017; 16:891-910. [PMID: 28331001 PMCID: PMC5417828 DOI: 10.1074/mcp.m117.067439] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 02/24/2017] [Indexed: 02/05/2023] Open
Abstract
Mutations in the Epidermal growth factor receptor (EGFR) kinase domain, such as the L858R missense mutation and deletions spanning the conserved sequence 747LREA750, are sensitive to tyrosine kinase inhibitors (TKIs). The gatekeeper site residue mutation, T790M accounts for around 60% of acquired resistance to EGFR TKIs. The first generation EGFR TKIs, erlotinib and gefitinib, and the second generation inhibitor, afatinib are FDA approved for initial treatment of EGFR mutated lung adenocarcinoma. The predominant biomarker of EGFR TKI responsiveness is the presence of EGFR TKI-sensitizing mutations. However, 30-40% of patients with EGFR mutations exhibit primary resistance to these TKIs, underscoring the unmet need of identifying additional biomarkers of treatment response. Here, we sought to characterize the dynamics of tyrosine phosphorylation upon EGFR TKI treatment of mutant EGFR-driven human lung adenocarcinoma cell lines with varying sensitivity to EGFR TKIs, erlotinib and afatinib. We employed stable isotope labeling with amino acids in cell culture (SILAC)-based quantitative mass spectrometry to identify and quantify tyrosine phosphorylated peptides. The proportion of tyrosine phosphorylated sites that had reduced phosphorylation upon erlotinib or afatinib treatment correlated with the degree of TKI-sensitivity. Afatinib, an irreversible EGFR TKI, more effectively inhibited tyrosine phosphorylation of a majority of the substrates. The phosphosites with phosphorylation SILAC ratios that correlated with the TKI-sensitivity of the cell lines include sites on kinases, such as EGFR-Y1197 and MAPK7-Y221, and adaptor proteins, such as SHC1-Y349/350, ERRFI1-Y394, GAB1-Y689, STAT5A-Y694, DLG3-Y705, and DAPP1-Y139, suggesting these are potential biomarkers of TKI sensitivity. DAPP1, is a novel target of mutant EGFR signaling and Y-139 is the major site of DAPP1 tyrosine phosphorylation. We also uncovered several off-target effects of these TKIs, such as MST1R-Y1238/Y1239 and MET-Y1252/1253. This study provides unique insight into the TKI-mediated modulation of mutant EGFR signaling, which can be applied to the development of biomarkers of EGFR TKI response.
Collapse
Affiliation(s)
- Xu Zhang
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Tapan Maity
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Manoj K Kashyap
- §Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205
| | - Mukesh Bansal
- ¶Department of System Biology, Columbia University, New York, New York, 10032
- ‖PsychoGenics Inc., Tarrytown, New York, 10591
| | - Abhilash Venugopalan
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Sahib Singh
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Shivangi Awasthi
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | | | | | - Natalya Belkina
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Stephanie Pitts
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Constance M Cultraro
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Shaojian Gao
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Guldal Kirkali
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Romi Biswas
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Raghothama Chaerkady
- §Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205
- **Medimmune LLC, Gaithersburg, Maryland, 20878
| | - Andrea Califano
- ¶Department of System Biology, Columbia University, New York, New York, 10032
| | - Akhilesh Pandey
- §Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205
| | - Udayan Guha
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892;
| |
Collapse
|
23
|
Hoang VT, Yan TJ, Cavanaugh JE, Flaherty PT, Beckman BS, Burow ME. Oncogenic signaling of MEK5-ERK5. Cancer Lett 2017; 392:51-59. [PMID: 28153789 PMCID: PMC5901897 DOI: 10.1016/j.canlet.2017.01.034] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 01/17/2017] [Accepted: 01/23/2017] [Indexed: 12/17/2022]
Abstract
Mitogen-activated protein kinases (MAPKs) regulate diverse cellular processes including proliferation, cell survival, differentiation, and apoptosis. While conventional MAPK constituents have well-defined roles in oncogenesis, the MEK5 pathway has only recently emerged in cancer research. In this review, we consider the MEK5 signaling cascade, focusing specifically on its involvement in drug resistance and regulation of aggressive cancer phenotypes. Moreover, we explore the role of MEK5/ERK5 in tumorigenesis and metastatic progression, discussing the discrepancies in preclinical studies and assessing its viability as a therapeutic target for anti-cancer agents.
Collapse
Affiliation(s)
- Van T Hoang
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA, USA
| | - Thomas J Yan
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA, USA
| | - Jane E Cavanaugh
- Department of Pharmacological Sciences, Division of Medicinal Chemistry, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | - Patrick T Flaherty
- Department of Pharmacological Sciences, Division of Medicinal Chemistry, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | | | - Matthew E Burow
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA, USA; Department of Pharmacology, Tulane University, New Orleans, LA, USA; Tulane Cancer Center, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
24
|
Shalaby SY, Chitragari G, Sumpio BJ, Sumpio BE. Shear Stress Induces Change in Extracellular Signal-Regulated Kinase 5 Levels with Sustained Activation under Disturbed and Continuous Laminar Flow. Int J Angiol 2017; 26:109-115. [PMID: 28566937 DOI: 10.1055/s-0037-1599057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Extracellular signal-regulated kinase 5 (ERK5) has been reported to regulate endothelial integrity and protect from vascular dysfunction under laminar flow. Previously reported research indicates that under laminar flow ERK5 is activated with production of atheroprotective molecules. However, the characterization of ERK5 activation and levels under different flow patterns has not been investigated. Confluent HUVECs were serum-starved then seeded on glass slides. HUVECs incubated in 1% FBS were exposed to continuous laminar flow (CLF), to-and-fro flow (TFF), or pulsatile forward flow (PFF) in a parallel plate flow chamber. At the end of experimentation, cell lysates were immunoblotted with antibodies to phospho-ERK5 and total ERK5. ERK5 activation was assessed by the levels of phosphorylated ERK5. The densitometric mean ± SEM is calculated and analyzed by ANOVA. p < 0.05 is considered significant. Levels of ERK5 decreased with all flow conditions with the largest decrease in TFF flow condition. TFF and CLF exhibited sustained ERK5 phosphorylation in HUVECs stimulated for up to 4 hours. PFF had transient phosphorylation of ERK5 at 2 hours, which then became undetectable at 4 hours of exposure to flow. Also, TFF and CLF both showed decreased levels at 4 hours, suggesting a decrease in activation for these flow conditions. Exposure of HUVEC to different types of shear stress results in varying patterns of activation of ERK5. Activation of ERK5 with TFF suggests a role in the pathogenesis of atherosclerosis and vascular remodeling under disturbed flow conditions.
Collapse
Affiliation(s)
- S Y Shalaby
- Section of Vascular Surgery, Yale University, New Haven, Connecticut
| | - G Chitragari
- Section of Vascular Surgery, Yale University, New Haven, Connecticut
| | - B J Sumpio
- Section of Vascular Surgery, Yale University, New Haven, Connecticut
| | - B E Sumpio
- Section of Vascular Surgery, Yale University, New Haven, Connecticut
| |
Collapse
|
25
|
Simões AES, Rodrigues CMP, Borralho PM. The MEK5/ERK5 signalling pathway in cancer: a promising novel therapeutic target. Drug Discov Today 2016; 21:1654-1663. [PMID: 27320690 DOI: 10.1016/j.drudis.2016.06.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/18/2016] [Accepted: 06/08/2016] [Indexed: 12/18/2022]
Abstract
Conventional mitogen-activated protein kinase (MAPK) family members are among the most sought-after oncogenic effectors for the development of novel human cancer treatment strategies. MEK5/ERK5 has been the less-studied MAPK subfamily, despite its increasingly demonstrated relevance in the growth, survival, and differentiation of normal cells. MEK5/ERK5 signalling has already been proposed to have pivotal roles in several cancer hallmarks, and to mediate the effects of a range of oncogenes. Accumulating evidence indicates the contribution of MEK5/ERK5 signalling to therapy resistance and the benefits of using MEK5/ERK5 inhibitory strategies in the treatment of human cancer. Here, we explore the major known contributions of MEK5/ERK5 signalling to the onset and progression of several types of cancer, and highlight the potential clinical relevance of targeting MEK5/ERK5 pathways.
Collapse
Affiliation(s)
- André E S Simões
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal.
| | - Pedro M Borralho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal.
| |
Collapse
|
26
|
Chen H, Tucker J, Wang X, Gavine PR, Phillips C, Augustin MA, Schreiner P, Steinbacher S, Preston M, Ogg D. Discovery of a novel allosteric inhibitor-binding site in ERK5: comparison with the canonical kinase hinge ATP-binding site. Acta Crystallogr D Struct Biol 2016; 72:682-93. [PMID: 27139631 PMCID: PMC4854315 DOI: 10.1107/s2059798316004502] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 03/16/2016] [Indexed: 01/20/2023] Open
Abstract
MAP kinases act as an integration point for multiple biochemical signals and are involved in a wide variety of cellular processes such as proliferation, differentiation, regulation of transcription and development. As a member of the MAP kinase family, ERK5 (MAPK7) is involved in the downstream signalling pathways of various cell-surface receptors, including receptor tyrosine kinases and G protein-coupled receptors. In the current study, five structures of the ERK5 kinase domain co-crystallized with ERK5 inhibitors are reported. Interestingly, three of the compounds bind at a novel allosteric binding site in ERK5, while the other two bind at the typical ATP-binding site. Binding of inhibitors at the allosteric site is accompanied by displacement of the P-loop into the ATP-binding site and is shown to be ATP-competitive in an enzymatic assay of ERK5 kinase activity. Kinase selectivity data show that the most potent allosteric inhibitor exhibits superior kinase selectivity compared with the two inhibitors that bind at the canonical ATP-binding site. An analysis of these structures and comparison with both a previously published ERK5-inhibitor complex structure (PDB entry 4b99) and the structures of three other kinases (CDK2, ITK and MEK) in complex with allosteric inhibitors are presented.
Collapse
Affiliation(s)
- Hongming Chen
- Chemistry Innovation Centre, Discovery Sciences, AstraZeneca R&D Mölndal, 431 83 Mölndal, Sweden
| | - Julie Tucker
- Structure and Biophysics, Discovery Sciences, AstraZeneca R&D Alderley Park, Macclesfield SK10 4TG, England
| | - Xiaotao Wang
- Innovation Centre China, AstraZeneca Asia and Emerging Markets iMed, Shanghai 201203, People’s Republic of China
| | - Paul R. Gavine
- Innovation Centre China, AstraZeneca Asia and Emerging Markets iMed, Shanghai 201203, People’s Republic of China
| | - Chris Phillips
- Structure and Biophysics, Discovery Sciences, AstraZeneca R&D Alderley Park, Macclesfield SK10 4TG, England
| | - Martin A. Augustin
- Proteros biostructures GmbH, Bunsenstrasse 7a, 82152 Martinsried, Germany
| | - Patrick Schreiner
- Proteros biostructures GmbH, Bunsenstrasse 7a, 82152 Martinsried, Germany
| | - Stefan Steinbacher
- Proteros biostructures GmbH, Bunsenstrasse 7a, 82152 Martinsried, Germany
| | - Marian Preston
- Screening Sciences, Discovery Sciences, AstraZeneca R&D Alderley Park, Macclesfield SK10 4TG, England
| | - Derek Ogg
- Structure and Biophysics, Discovery Sciences, AstraZeneca R&D Alderley Park, Macclesfield SK10 4TG, England
| |
Collapse
|
27
|
Lochhead PA, Clark J, Wang LZ, Gilmour L, Squires M, Gilley R, Foxton C, Newell DR, Wedge SR, Cook SJ. Tumor cells with KRAS or BRAF mutations or ERK5/MAPK7 amplification are not addicted to ERK5 activity for cell proliferation. Cell Cycle 2016; 15:506-18. [PMID: 26959608 PMCID: PMC5056618 DOI: 10.1080/15384101.2015.1120915] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 10/08/2015] [Accepted: 11/12/2015] [Indexed: 10/24/2022] Open
Abstract
ERK5, encoded by MAPK7, has been proposed to play a role in cell proliferation, thus attracting interest as a cancer therapeutic target. While oncogenic RAS or BRAF cause sustained activation of the MEK1/2-ERK1/2 pathway, ERK5 is directly activated by MEK5. It has been proposed that RAS and RAF proteins can also promote ERK5 activation. Here we investigated the interplay between RAS-RAF-MEK-ERK and ERK5 signaling and studied the role of ERK5 in tumor cell proliferation in 2 disease-relevant cell models. We demonstrate that although an inducible form of CRAF (CRAF:ER*) can activate ERK5 in fibroblasts, the response is delayed and reflects feed-forward signaling. Additionally, oncogenic KRAS and BRAF do not activate ERK5 in epithelial cells. Although KRAS and BRAF do not couple directly to MEK5-ERK5, ERK5 signaling might still be permissive for proliferation. However, neither the selective MEK5 inhibitor BIX02189 or ERK5 siRNA inhibited proliferation of colorectal cancer cells harbouring KRAS(G12C/G13D) or BRAF(V600E). Furthermore, there was no additive or synergistic effect observed when BIX02189 was combined with the MEK1/2 inhibitor Selumetinib (AZD6244), suggesting that ERK5 was neither required for proliferation nor a driver of innate resistance to MEK1/2 inhibitors. Finally, even cancer cells with MAPK7 amplification were resistant to BIX02189 and ERK5 siRNA, showing that ERK5 amplification does not confer addiction to ERK5 for cell proliferation. Thus ERK5 signaling is unlikely to play a role in tumor cell proliferation downstream of KRAS or BRAF or in tumor cells with ERK5 amplification. These results have important implications for the role of ERK5 as an anti-cancer drug target.
Collapse
Affiliation(s)
| | - Jonathan Clark
- Biological Chemistry Facility; The Babraham Institute; Cambridge, UK
| | - Lan-Zhen Wang
- The Northern Institute for Cancer Research; University of Newcastle upon Tyne, Newcastle, UK
| | - Lesley Gilmour
- Cancer Research Technology; The Beatson Institute for Cancer Research; Garscube Estate; Glasgow, UK
- Current address: Translational Radiation Biology; The Beatson Institute for Cancer Research; Garscube Estate; Glasgow, UK
| | - Matthew Squires
- Signalling Laboratory; The Babraham Institute; Cambridge, UK
- Current address: Novartis; Basel, Switzerland
| | - Rebecca Gilley
- Signalling Laboratory; The Babraham Institute; Cambridge, UK
| | - Caroline Foxton
- Cancer Research Technology; CRT Discovery Laboratories; London Bioscience Innovation Centre; London, UK
- Current address: Centre for Drug Development; Cancer Research UK; London, UK
| | - David R. Newell
- The Northern Institute for Cancer Research; University of Newcastle upon Tyne, Newcastle, UK
| | - Stephen R. Wedge
- The Northern Institute for Cancer Research; University of Newcastle upon Tyne, Newcastle, UK
| | - Simon J. Cook
- Signalling Laboratory; The Babraham Institute; Cambridge, UK
| |
Collapse
|