1
|
Mondru AK, Wilkinson B, Aljasir MA, Alrumayh A, Greaves G, Emmett M, Albohairi S, Pritchard-Jones R, Cross MJ. The ERK5 pathway in BRAFV600E melanoma cells plays a role in development of acquired resistance to dabrafenib but not vemurafenib. FEBS Lett 2024; 598:2011-2027. [PMID: 38977937 DOI: 10.1002/1873-3468.14960] [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: 01/23/2024] [Revised: 04/28/2024] [Accepted: 05/14/2024] [Indexed: 07/10/2024]
Abstract
Malignant melanoma, an aggressive skin cancer with a poor prognosis, frequently features BRAFV600E mutation resulting in activation of the MAPK pathway and melanocyte proliferation and survival. BRAFV600E inhibitors like vemurafenib and dabrafenib have enhanced patient survival, yet drug resistance remains a significant challenge. We investigated the role of the ERK5 pathway in BRAFV600E melanoma cells and cells with acquired resistance to PLX4720 (vemurafenib) and dabrafenib. In BRAFV600E melanoma, ERK5 inhibition minimally affected viability compared to ERK1/2 inhibition. In vemurafenib-resistant cells, ERK5 inhibition alone didn't impact viability or restore drug sensitivity to vemurafenib. However, in dabrafenib-resistant cells, ERK5 inhibition reduced viability and enhanced the anti-proliferative effect of MEK1/2 inhibition. Targeting the ERK5 pathway may represent a therapeutic opportunity in dabrafenib-resistant melanoma.
Collapse
Affiliation(s)
- Anil Kumar Mondru
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, UK
| | - Beth Wilkinson
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, UK
| | - Mohammad A Aljasir
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, UK
| | - Ahmed Alrumayh
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, UK
| | - Georgia Greaves
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, UK
| | - Maxine Emmett
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, UK
| | - Saad Albohairi
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, UK
| | - Rowan Pritchard-Jones
- Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, UK
| | - Michael J Cross
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, UK
| |
Collapse
|
2
|
Sumiyoshi S, Shiozaki A, Kosuga T, Simizu H, Kudo M, Kiuchi J, Arita T, Konishi H, Komatsu S, Kuriu Y, Kubota T, Fujiwara H, Morinaga Y, Konishi E, Otsuji E. Functional Analysis and Clinical Importance of ATP1A1 in Colon Cancer. Ann Surg Oncol 2023; 30:6898-6910. [PMID: 37407874 DOI: 10.1245/s10434-023-13779-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/04/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Na+/K+-ATPase α1 subunit (ATP1A1) exhibits aberrant expression in various types of cancer. Moreover, its levels in specific tissues are associated with the development of cancer. Nevertheless, the mechanism and signaling pathways underlying the effects of ATP1A1 in colon cancer (CC) have not been elucidated, and its prognostic impact remains unknown. METHODS Knockdown of ATP1A1 expression was performed in human CC cell lines HT29 and Caco2 using small interfering RNA. The roles of ATP1A1 in various biological processes of cells (i.e., proliferation, cell cycle, apoptosis, migration, and invasion) were assessed. Microarray analysis was utilized for gene expression profiling. Samples obtained from 200 patients with CC who underwent curative colectomy were analyzed through immunohistochemistry. RESULTS ATP1A1 knockdown suppressed cell proliferation, migration, and invasion and induced apoptosis. The results of the microarray analysis revealed that the upregulated or downregulated gene expression in ATP1A1-depleted cells was related to the extracellular signal-regulated kinase 5 (ERK5) signaling pathway [epidermal growth factor receptor (EGFR), mitogen-activated protein kinase kinase 5 (MAP2K5), mitogen-activated protein kinase 7 (MAPK7), FOS, MYC, and BCL2 associated agonist of cell death (BAD)]. Immunohistochemical analysis demonstrated a correlation between ATP1A1 expression and pathological T stage (p = 0.0054), and multivariate analysis identified high ATP1A1 expression as an independent predictor of poor recurrence-free survival in patients with CC (p = 0.0040, hazard ratio: 2.807, 95% confidence interval 1.376-6.196). CONCLUSIONS ATP1A1 regulates tumor progression through the ERK5 signaling pathway. High ATP1A1 expression is associated with poor long-term outcomes in patients with CC.
Collapse
Affiliation(s)
- Shutaro Sumiyoshi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Toshiyuki Kosuga
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroki Simizu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Michihiro Kudo
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Jun Kiuchi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomohiro Arita
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshiaki Kuriu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takeshi Kubota
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hitoshi Fujiwara
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yukiko Morinaga
- Department of Surgical Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eiichi Konishi
- Department of Surgical Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
3
|
Jain A, Casanova D, Padilla AV, Paniagua Bojorges A, Kotla S, Ko KA, Samanthapudi VSK, Chau K, Nguyen MTH, Wen J, Hernandez Gonzalez SL, Rodgers SP, Olmsted-Davis EA, Hamilton DJ, Reyes-Gibby C, Yeung SCJ, Cooke JP, Herrmann J, Chini EN, Xu X, Yusuf SW, Yoshimoto M, Lorenzi PL, Hobbs B, Krishnan S, Koutroumpakis E, Palaskas NL, Wang G, Deswal A, Lin SH, Abe JI, Le NT. Premature senescence and cardiovascular disease following cancer treatments: mechanistic insights. Front Cardiovasc Med 2023; 10:1212174. [PMID: 37781317 PMCID: PMC10540075 DOI: 10.3389/fcvm.2023.1212174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/03/2023] [Indexed: 10/03/2023] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality, especially among the aging population. The "response-to-injury" model proposed by Dr. Russell Ross in 1999 emphasizes inflammation as a critical factor in atherosclerosis development, with atherosclerotic plaques forming due to endothelial cell (EC) injury, followed by myeloid cell adhesion and invasion into the blood vessel walls. Recent evidence indicates that cancer and its treatments can lead to long-term complications, including CVD. Cellular senescence, a hallmark of aging, is implicated in CVD pathogenesis, particularly in cancer survivors. However, the precise mechanisms linking premature senescence to CVD in cancer survivors remain poorly understood. This article aims to provide mechanistic insights into this association and propose future directions to better comprehend this complex interplay.
Collapse
Affiliation(s)
- Ashita Jain
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Diego Casanova
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | | | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kyung Ae Ko
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | - Khanh Chau
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Minh T. H. Nguyen
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Jake Wen
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | | | - Shaefali P. Rodgers
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | | | - Dale J. Hamilton
- Department of Medicine, Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Cielito Reyes-Gibby
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sai-Ching J. Yeung
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - John P. Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Joerg Herrmann
- Cardio Oncology Clinic, Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Eduardo N. Chini
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Xiaolei Xu
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
| | - Syed Wamique Yusuf
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Momoko Yoshimoto
- Center for Stem Cell & Regenerative Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Philip L. Lorenzi
- Department of Bioinformatics and Computational Biology, Division of VP Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Brain Hobbs
- Department of Population Health, The University of Texas at Austin, Austin, TX, United States
| | - Sunil Krishnan
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Efstratios Koutroumpakis
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nicolas L. Palaskas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Guangyu Wang
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Steven H. Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jun-ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
4
|
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
|
5
|
Proteogenomics of diffuse gliomas reveal molecular subtypes associated with specific therapeutic targets and immune-evasion mechanisms. Nat Commun 2023; 14:505. [PMID: 36720864 PMCID: PMC9889805 DOI: 10.1038/s41467-023-36005-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 01/12/2023] [Indexed: 02/02/2023] Open
Abstract
Diffuse gliomas are devastating brain tumors. Here, we perform a proteogenomic profiling of 213 retrospectively collected glioma tumors. Proteogenomic analysis reveals the downstream biological events leading by EGFR-, IDH1-, TP53-mutations. The comparative analysis illustrates the distinctive features of GBMs and LGGs, indicating CDK2 inhibitor might serve as a promising drug target for GBMs. Further proteogenomic integrative analysis combined with functional experiments highlight the cis-effect of EGFR alterations might lead to glioma tumor cell proliferation through ERK5 medicates nucleotide synthesis process. Proteome-based stratification of gliomas defines 3 proteomic subgroups (S-Ne, S-Pf, S-Im), which could serve as a complement to WHO subtypes, and would provide the essential framework for the utilization of specific targeted therapies for particular glioma subtypes. Immune clustering identifies three immune subtypes with distinctive immune cell types. Further analysis reveals higher EGFR alteration frequencies accounts for elevation of immune check point protein: PD-L1 and CD70 in T-cell infiltrated tumors.
Collapse
|
6
|
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
|
7
|
OXTR High stroma fibroblasts control the invasion pattern of oral squamous cell carcinoma via ERK5 signaling. Nat Commun 2022; 13:5124. [PMID: 36045118 PMCID: PMC9433374 DOI: 10.1038/s41467-022-32787-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 08/17/2022] [Indexed: 11/20/2022] Open
Abstract
The Pattern Of Invasion (POI) of tumor cells into adjacent normal tissues clinically predicts postoperative tumor metastasis/recurrence of early oral squamous cell carcinoma (OSCC), but the mechanisms underlying the development of these subtypes remain unclear. Focusing on the highest score of POIs (Worst POI, WPOI) present within each tumor, we observe a disease progression-driven shift of WPOI towards the high-risk type 4/5, associated with a mesenchymal phenotype in advanced OSCC. WPOI 4-5-derived cancer-associated fibroblasts (CAFsWPOI4-5), characterized by high oxytocin receptor expression (OXTRHigh), contribute to local-regional metastasis. OXTRHigh CAFs induce a desmoplastic stroma and CCL26 is required for the invasive phenotype of CCR3+ tumors. Mechanistically, OXTR activates nuclear ERK5 transcription signaling via Gαq and CDC37 to maintain high levels of OXTR and CCL26. ERK5 ablation reprograms the pro-invasive phenotype of OXTRHigh CAFs. Therefore, targeting ERK5 signaling in OXTRHigh CAFs is a potential therapeutic strategy for OSCC patients with WPOI 4-5. Worst pattern of invasion (WPOI) is a parameter used to quantify tumor invasiveness of oral squamous cell carcinoma (OSCC). Here the authors show that a fibroblast subset characterized by the expression of the oxytocin receptor is enriched in highly invasive WPOI 4-5 OSCC tumors and can be targeted to reduce the desmoplastic stroma and tumor metastasis.
Collapse
|
8
|
Cook SJ, Lochhead PA. ERK5 Signalling and Resistance to ERK1/2 Pathway Therapeutics: The Path Less Travelled? Front Cell Dev Biol 2022; 10:839997. [PMID: 35903549 PMCID: PMC9315226 DOI: 10.3389/fcell.2022.839997] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 06/13/2022] [Indexed: 12/01/2022] Open
Abstract
The RAS-regulated RAF-MEK1/2-ERK1/2 signalling pathway is frequently de-regulated in human cancer. Melanoma in particular exhibits a high incidence of activating BRAFV600E/K and NRASQ61L/K mutations and such cells are addicted to the activity of these mutant oncoproteins. As a result three different BRAF inhibitors (BRAFi) have now been approved for BRAFV600E/K- mutant melanoma and have transformed the treatment of this disease. Despite this, clinical responses are typically transient as tumour cells develop resistance. These resistance mechanisms frequently involve reinstatement of ERK1/2 signalling and BRAFi are now deployed in combination with one of three approved MEK1/2 inhibitors (MEKi) to provide more durable, but still transient, clinical responses. Furthermore, inhibitors to ERK1/2 (ERK1/2i) have also been developed to counteract ERK1/2 signalling. However, recent studies have suggested that BRAFi/MEKi and ERK1/2i resistance can arise through activation of a parallel signalling pathway leading to activation of ERK5, an unusual protein kinase that contains both a kinase domain and a transcriptional transactivation domain. Here we review the evidence supporting ERK5 as a mediator of BRAFi/MEKi and ERK1/2i resistance. We also review the challenges in targeting ERK5 signalling with small molecules, including paradoxical activation of the transcriptional transactivation domain, and discuss new therapeutic modalities that could be employed to target ERK5.
Collapse
Affiliation(s)
- Simon J. Cook
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | - Pamela A. Lochhead
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
- Mechanistic and Structural Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, United Kingdom
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
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
|
12
|
High ELK3 expression is associated with the VEGF-C/VEGFR-3 axis and gastric tumorigenesis and enhances infiltration of M2 macrophages. Future Med Chem 2020; 12:2209-2224. [PMID: 33191789 DOI: 10.4155/fmc-2019-0337] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: To assess the expression and effect of ELK3 in gastric cancer, along with its associations with the VEGF-C/VEGFR-3 axis, IC50 of the VEGFR-3 inhibitor axitinib and immune infiltration of M2-polarized macrophages in gastric cancer, and to analyze the possible epigenetic regulation mechanism. Materials & methods: Expression profiles and methylation data from 1645 samples were obtained and examined from multi-institutional public datasets. The associations were assessed by multiple analysis methods. Results: Elevated ELK3 is associated with the VEGF-C/VEGFR-3 axis and tumorigenesis, reduces the effect of axitinib in vitro, enhances immune infiltration of M2 macrophages and affects clinical outcomes. Hypomethylation contributes to the upregulation of ELK3 in gastric cancer. Conclusions: ELK3 is a potential therapeutic target which reduces the effect of axitinib and enhances infiltration of M2-polarized macrophages.
Collapse
|
13
|
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: 22] [Impact Index Per Article: 4.4] [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
|
14
|
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
|
15
|
Cristea S, Coles GL, Hornburg D, Gershkovitz M, Arand J, Cao S, Sen T, Williamson SC, Kim JW, Drainas AP, He A, Cam LL, Byers LA, Snyder MP, Contrepois K, Sage J. The MEK5-ERK5 Kinase Axis Controls Lipid Metabolism in Small-Cell Lung Cancer. Cancer Res 2020; 80:1293-1303. [PMID: 31969375 PMCID: PMC7073279 DOI: 10.1158/0008-5472.can-19-1027] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 12/13/2019] [Accepted: 01/13/2020] [Indexed: 12/31/2022]
Abstract
Small-cell lung cancer (SCLC) is an aggressive form of lung cancer with dismal survival rates. While kinases often play key roles driving tumorigenesis, there are strikingly few kinases known to promote the development of SCLC. Here, we investigated the contribution of the MAPK module MEK5-ERK5 to SCLC growth. MEK5 and ERK5 were required for optimal survival and expansion of SCLC cell lines in vitro and in vivo. Transcriptomics analyses identified a role for the MEK5-ERK5 axis in the metabolism of SCLC cells, including lipid metabolism. In-depth lipidomics analyses showed that loss of MEK5/ERK5 perturbs several lipid metabolism pathways, including the mevalonate pathway that controls cholesterol synthesis. Notably, depletion of MEK5/ERK5 sensitized SCLC cells to pharmacologic inhibition of the mevalonate pathway by statins. These data identify a new MEK5-ERK5-lipid metabolism axis that promotes the growth of SCLC. SIGNIFICANCE: This study is the first to investigate MEK5 and ERK5 in SCLC, linking the activity of these two kinases to the control of cell survival and lipid metabolism.
Collapse
Affiliation(s)
- Sandra Cristea
- Department of Pediatrics, Stanford University, Stanford, California
- Department of Genetics, Stanford University, Stanford, California
| | - Garry L Coles
- Department of Pediatrics, Stanford University, Stanford, California
- Department of Genetics, Stanford University, Stanford, California
| | - Daniel Hornburg
- Department of Genetics, Stanford University, Stanford, California
| | - Maya Gershkovitz
- Department of Pediatrics, Stanford University, Stanford, California
- Department of Genetics, Stanford University, Stanford, California
| | - Julia Arand
- Department of Pediatrics, Stanford University, Stanford, California
- Department of Genetics, Stanford University, Stanford, California
| | - Siqi Cao
- Department of Pediatrics, Stanford University, Stanford, California
- Department of Genetics, Stanford University, Stanford, California
| | - Triparna Sen
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stuart C Williamson
- Department of Pediatrics, Stanford University, Stanford, California
- Department of Genetics, Stanford University, Stanford, California
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Jun W Kim
- Department of Pediatrics, Stanford University, Stanford, California
- Department of Genetics, Stanford University, Stanford, California
| | - Alexandros P Drainas
- Department of Pediatrics, Stanford University, Stanford, California
- Department of Genetics, Stanford University, Stanford, California
| | - Andrew He
- Department of Pediatrics, Stanford University, Stanford, California
- Department of Genetics, Stanford University, Stanford, California
| | - Laurent Le Cam
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut Régional du Cancer de Montpellier, Montpellier, France
| | - Lauren Averett Byers
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael P Snyder
- Department of Genetics, Stanford University, Stanford, California
| | - Kévin Contrepois
- Department of Genetics, Stanford University, Stanford, California
| | - Julien Sage
- Department of Pediatrics, Stanford University, Stanford, California.
- Department of Genetics, Stanford University, Stanford, California
| |
Collapse
|
16
|
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
|
17
|
Myers SM, Miller DC, Molyneux L, Arasta M, Bawn RH, Blackburn TJ, Cook SJ, Edwards N, Endicott JA, Golding BT, Griffin RJ, Hammonds T, Hardcastle IR, Harnor SJ, Heptinstall AB, Lochhead PA, Martin MP, Martin NC, Newell DR, Owen PJ, Pang LC, Reuillon T, Rigoreau LJM, Thomas HD, Tucker JA, Wang LZ, Wong AC, Noble MEM, Wedge SR, Cano C. Identification of a novel orally bioavailable ERK5 inhibitor with selectivity over p38α and BRD4. Eur J Med Chem 2019; 178:530-543. [PMID: 31212132 DOI: 10.1016/j.ejmech.2019.05.057] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/30/2019] [Accepted: 05/20/2019] [Indexed: 11/18/2022]
Abstract
Extracellular regulated kinase 5 (ERK5) signalling has been implicated in driving a number of cellular phenotypes including endothelial cell angiogenesis and tumour cell motility. Novel ERK5 inhibitors were identified using high throughput screening, with a series of pyrrole-2-carboxamides substituted at the 4-position with an aroyl group being found to exhibit IC50 values in the micromolar range, but having no selectivity against p38α MAP kinase. Truncation of the N-substituent marginally enhanced potency (∼3-fold) against ERK5, but importantly attenuated inhibition of p38α. Systematic variation of the substituents on the aroyl group led to the selective inhibitor 4-(2-bromo-6-fluorobenzoyl)-N-(pyridin-3-yl)-1H-pyrrole-2-carboxamide (IC50 0.82 μM for ERK5; IC50 > 120 μM for p38α). The crystal structure (PDB 5O7I) of this compound in complex with ERK5 has been solved. This compound was orally bioavailable and inhibited bFGF-driven Matrigel plug angiogenesis and tumour xenograft growth. The selective ERK5 inhibitor described herein provides a lead for further development into a tool compound for more extensive studies seeking to examine the role of ERK5 signalling in cancer and other diseases.
Collapse
Affiliation(s)
- Stephanie M Myers
- Newcastle Drug Discovery, Northern Institute for Cancer Research, School of Chemistry, Bedson Building, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| | - Duncan C Miller
- Newcastle Drug Discovery, Northern Institute for Cancer Research, School of Chemistry, Bedson Building, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| | - Lauren Molyneux
- Newcastle Drug Discovery, Northern Institute for Cancer Research, School of Chemistry, Bedson Building, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| | - Mercedes Arasta
- Newcastle Drug Discovery, Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Ruth H Bawn
- Newcastle Drug Discovery, Northern Institute for Cancer Research, School of Chemistry, Bedson Building, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| | - Timothy J Blackburn
- Newcastle Drug Discovery, Northern Institute for Cancer Research, School of Chemistry, Bedson Building, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| | - Simon J Cook
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Noel Edwards
- Newcastle Drug Discovery, Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Jane A Endicott
- Newcastle Drug Discovery, Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Bernard T Golding
- Newcastle Drug Discovery, Northern Institute for Cancer Research, School of Chemistry, Bedson Building, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| | - Roger J Griffin
- Newcastle Drug Discovery, Northern Institute for Cancer Research, School of Chemistry, Bedson Building, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| | - Tim Hammonds
- Cancer Research UK Therapeutic Discovery Laboratories, London Bioscience Innovation Centre, 2 Royal College Street, London, NW1 0NH, UK
| | - Ian R Hardcastle
- Newcastle Drug Discovery, Northern Institute for Cancer Research, School of Chemistry, Bedson Building, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| | - Suzannah J Harnor
- Newcastle Drug Discovery, Northern Institute for Cancer Research, School of Chemistry, Bedson Building, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| | - Amy B Heptinstall
- Newcastle Drug Discovery, Northern Institute for Cancer Research, School of Chemistry, Bedson Building, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| | - Pamela A Lochhead
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Mathew P Martin
- Newcastle Drug Discovery, Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Nick C Martin
- Newcastle Drug Discovery, Northern Institute for Cancer Research, School of Chemistry, Bedson Building, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| | - David R Newell
- Newcastle Drug Discovery, Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Paul J Owen
- Cancer Research UK Therapeutic Discovery Laboratories, London Bioscience Innovation Centre, 2 Royal College Street, London, NW1 0NH, UK
| | - Leon C Pang
- Cancer Research UK Therapeutic Discovery Laboratories, London Bioscience Innovation Centre, 2 Royal College Street, London, NW1 0NH, UK
| | - Tristan Reuillon
- Newcastle Drug Discovery, Northern Institute for Cancer Research, School of Chemistry, Bedson Building, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
| | - Laurent J M Rigoreau
- Cancer Research UK Therapeutic Discovery Laboratories, Jonas Webb Building, Babraham Campus, Babraham, Cambridgeshire, CB22 3AT, UK
| | - Huw D Thomas
- Newcastle Drug Discovery, Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Julie A Tucker
- Newcastle Drug Discovery, Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Lan-Zhen Wang
- Newcastle Drug Discovery, Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Ai-Ching Wong
- Cancer Research UK Therapeutic Discovery Laboratories, London Bioscience Innovation Centre, 2 Royal College Street, London, NW1 0NH, UK
| | - Martin E M Noble
- Newcastle Drug Discovery, Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
| | - Stephen R Wedge
- Newcastle Drug Discovery, Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
| | - Celine Cano
- Newcastle Drug Discovery, Northern Institute for Cancer Research, School of Chemistry, Bedson Building, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK.
| |
Collapse
|
18
|
Sánchez-Fdez A, Ortiz-Ruiz MJ, Re-Louhau MF, Ramos I, Blanco-Múñez Ó, Ludeña D, Abad M, Sánchez-Martín M, Pandiella A, Esparís-Ogando A. MEK5 promotes lung adenocarcinoma. Eur Respir J 2019; 53:13993003.01327-2018. [PMID: 30442718 PMCID: PMC6393765 DOI: 10.1183/13993003.01327-2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 11/05/2018] [Indexed: 11/05/2022]
Abstract
Lung cancer represents the leading cause of cancer death worldwide [1]. Because of that, intense efforts are being devoted to the development of novel therapeutic strategies to fight the disease [2]. In this respect, identification of new oncogenic drivers offers therapeutic opportunities in tumours in which those molecules or other cooperating elements play a pathophysiological role. Here, we show that the MEK5 mitogen-activated protein kinase kinase has a pivotal role in lung cancer. MEK5 acts as an oncogenic driver in mice lung cancer and is pivotal for human lung adenocarcinomahttp://ow.ly/M9e830mZb8N
Collapse
Affiliation(s)
- Adrián Sánchez-Fdez
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, Salamanca, Spain.,IBSAL, Salamanca, Spain.,CIBERONC, Salamanca, Spain
| | - María Jesús Ortiz-Ruiz
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, Salamanca, Spain.,IBSAL, Salamanca, Spain
| | - María Florencia Re-Louhau
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, Salamanca, Spain.,IBSAL, Salamanca, Spain
| | - Isabel Ramos
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, Salamanca, Spain
| | - Óscar Blanco-Múñez
- IBSAL, Salamanca, Spain.,Pathology Service, University Hospital, Salamanca, Spain
| | - Dolores Ludeña
- IBSAL, Salamanca, Spain.,Pathology Service, University Hospital, Salamanca, Spain
| | - Mar Abad
- IBSAL, Salamanca, Spain.,Pathology Service, University Hospital, Salamanca, Spain
| | - Manuel Sánchez-Martín
- CIBERONC, Salamanca, Spain.,Transgenic Facility of the Nucleus Platform, University of Salamanca, Salamanca, Spain
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, Salamanca, Spain.,IBSAL, Salamanca, Spain.,CIBERONC, Salamanca, Spain
| | - Azucena Esparís-Ogando
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, Salamanca, Spain.,IBSAL, Salamanca, Spain.,CIBERONC, Salamanca, Spain
| |
Collapse
|
19
|
Structure activity relationships of anthranilic acid-based compounds on cellular and in vivo mitogen activated protein kinase-5 signaling pathways. Bioorg Med Chem Lett 2018; 28:2294-2301. [DOI: 10.1016/j.bmcl.2018.05.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 05/11/2018] [Accepted: 05/12/2018] [Indexed: 11/15/2022]
|
20
|
Absence of ERK5/MAPK7 delays tumorigenesis in Atm-/- mice. Oncotarget 2018; 7:74435-74447. [PMID: 27793024 PMCID: PMC5342677 DOI: 10.18632/oncotarget.12908] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 10/13/2016] [Indexed: 01/08/2023] Open
Abstract
Ataxia-telangiectasia mutated (ATM) is a cell cycle checkpoint kinase that upon activation by DNA damage leads to cell cycle arrest and DNA repair or apoptosis. The absence of Atm or the occurrence of loss-of-function mutations in Atm predisposes to tumorigenesis. MAPK7 has been implicated in numerous types of cancer with pro-survival and pro-growth roles in tumor cells, but its functional relation with tumor suppressors is not clear. In this study, we show that absence of MAPK7 delays death due to spontaneous tumor development in Atm−/− mice. Compared with Atm−/− thymocytes, Mapk7−/−Atm−/− thymocytes exhibited an improved response to DNA damage (increased phosphorylation of H2AX) and a restored apoptotic response after treatment of mice with ionizing radiation. These findings define an antagonistic function of ATM and MAPK7 in the thymocyte response to DNA damage, and suggest that the lack of MAPK7 inhibits thymic lymphoma growth in Atm−/− mice by partially restoring the DNA damage response in thymocytes.
Collapse
|
21
|
Pereira DM, Simões AES, Gomes SE, Castro RE, Carvalho T, Rodrigues CMP, Borralho PM. MEK5/ERK5 signaling inhibition increases colon cancer cell sensitivity to 5-fluorouracil through a p53-dependent mechanism. Oncotarget 2018; 7:34322-40. [PMID: 27144434 PMCID: PMC5085159 DOI: 10.18632/oncotarget.9107] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 03/31/2016] [Indexed: 12/22/2022] Open
Abstract
The MEK5/ERK5 signaling pathway is emerging as an important contributor to colon cancer onset, progression and metastasis; however, its relevance to chemotherapy resistance remains unknown. Here, we evaluated the impact of the MEK5/ERK5 cascade in colon cancer cell sensitivity to 5-fluorouracil (5-FU). Increased ERK5 expression was correlated with poor overall survival in colon cancer patients. In colon cancer cells, 5-FU exposure impaired endogenous KRAS/MEK5/ERK5 expression and/or activation. In turn, MEK5 constitutive activation reduced 5-FU-induced cytotoxicity. Using genetic and pharmacological approaches, we showed that ERK5 inhibition increased caspase-3/7 activity and apoptosis following 5-FU exposure. Mechanistically, this was further associated with increased p53 transcriptional activation of p21 and PUMA. In addition, ERK5 inhibition increased the response of HCT116 p53+/+ cells to 5-FU, but failed to sensitize HCT116 p53−/− cells to the cytotoxic effects of this chemotherapeutic agent, suggesting a p53-dependent axis mediating 5-FU sensitization. Finally, ERK5 inhibition using XMD8-92 was shown to increase the antitumor effects of 5-FU in a murine subcutaneous xenograft model, enhancing apoptosis while markedly reducing tumor growth. Collectively, our results suggest that ERK5-targeted in hibition provides a promising therapeutic approach to overcome resistance to 5-FU-based chemotherapy and improve colon cancer treatment.
Collapse
Affiliation(s)
- Diane M Pereira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - André E S Simões
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Sofia E Gomes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Rui E Castro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Tânia Carvalho
- Histology and Comparative Pathology Laboratory, Instituto de Medicina Molecular, Lisbon, Portugal
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Pedro M Borralho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
22
|
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
|
23
|
Jo M, Chung AY, Yachie N, Seo M, Jeon H, Nam Y, Seo Y, Kim E, Zhong Q, Vidal M, Park HC, Roth FP, Suk K. Yeast genetic interaction screen of human genes associated with amyotrophic lateral sclerosis: identification of MAP2K5 kinase as a potential drug target. Genome Res 2017; 27:1487-1500. [PMID: 28596290 PMCID: PMC5580709 DOI: 10.1101/gr.211649.116] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 06/06/2017] [Indexed: 12/13/2022]
Abstract
To understand disease mechanisms, a large-scale analysis of human–yeast genetic interactions was performed. Of 1305 human disease genes assayed, 20 genes exhibited strong toxicity in yeast. Human–yeast genetic interactions were identified by en masse transformation of the human disease genes into a pool of 4653 homozygous diploid yeast deletion mutants with unique barcode sequences, followed by multiplexed barcode sequencing to identify yeast toxicity modifiers. Subsequent network analyses focusing on amyotrophic lateral sclerosis (ALS)-associated genes, such as optineurin (OPTN) and angiogenin (ANG), showed that the human orthologs of the yeast toxicity modifiers of these ALS genes are enriched for several biological processes, such as cell death, lipid metabolism, and molecular transport. When yeast genetic interaction partners held in common between human OPTN and ANG were validated in mammalian cells and zebrafish, MAP2K5 kinase emerged as a potential drug target for ALS therapy. The toxicity modifiers identified in this study may deepen our understanding of the pathogenic mechanisms of ALS and other devastating diseases.
Collapse
Affiliation(s)
- Myungjin Jo
- Department of Pharmacology, Brain Science and Engineering Institute, and Department of Biomedical Sciences, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, 41944, Korea
| | - Ah Young Chung
- Department of Biomedical Sciences, Korea University Ansan Hospital, Ansan-si, Gyeonggi-do, 425-707, Korea
| | - Nozomu Yachie
- Donnelly Centre and Departments of Molecular Genetics and Computer Science, University of Toronto and Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario M5G 1X5, Canada
| | - Minchul Seo
- Department of Pharmacology, Brain Science and Engineering Institute, and Department of Biomedical Sciences, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, 41944, Korea
| | - Hyejin Jeon
- Department of Pharmacology, Brain Science and Engineering Institute, and Department of Biomedical Sciences, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, 41944, Korea
| | - Youngpyo Nam
- Department of Pharmacology, Brain Science and Engineering Institute, and Department of Biomedical Sciences, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, 41944, Korea
| | - Yeojin Seo
- Department of Pharmacology, Brain Science and Engineering Institute, and Department of Biomedical Sciences, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, 41944, Korea
| | - Eunmi Kim
- Department of Biomedical Sciences, Korea University Ansan Hospital, Ansan-si, Gyeonggi-do, 425-707, Korea
| | - Quan Zhong
- Department of Biological Sciences, Wright State University, Dayton, Ohio 45435, USA
| | - Marc Vidal
- Department of Biological Sciences, Wright State University, Dayton, Ohio 45435, USA
| | - Hae Chul Park
- Department of Biomedical Sciences, Korea University Ansan Hospital, Ansan-si, Gyeonggi-do, 425-707, Korea
| | - Frederick P Roth
- Donnelly Centre and Departments of Molecular Genetics and Computer Science, University of Toronto and Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario M5G 1X5, Canada.,Canadian Institute for Advanced Research, Toronto, Ontario M5G 1Z8, Canada
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science and Engineering Institute, and Department of Biomedical Sciences, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, 41944, Korea
| |
Collapse
|
24
|
Jiang Y, Dai J, Yao Z, Shelley G, Keller ET. Abituzumab Targeting of αV-Class Integrins Inhibits Prostate Cancer Progression. Mol Cancer Res 2017; 15:875-883. [PMID: 28314844 DOI: 10.1158/1541-7786.mcr-16-0447] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/17/2017] [Accepted: 03/10/2017] [Indexed: 12/22/2022]
Abstract
Integrins that contain an integrin αV subunit contribute to multiple functions that promote cancer progression. The goal of this study was to determine whether abituzumab (DI17E6, EMD 525797), a humanized monoclonal antibody (mAb) against integrin αV impacts, prostate cancer progression. To evaluate this, prostate cancer cells were treated with DI17E6 and its effects on proliferation, apoptosis, cell-cycle, adhesion, detachment, migration, invasion and phosphorylation of downstream targets, including FAK, Akt, and ERK, were determined. DI17E6 promoted detachment and inhibited adhesion of prostate cancer cells to several extracellular matrix (ECM) proteins and cells found in the bone microenvironment but had no impact on cell viability, cell-cycle, and caspase-3/7 activity. DI17E6 inhibited migration and invasion of prostate cancer cells. In addition, DI7E6 decreased phosphorylation of FAK, Akt, and ERK. These results indicate that inhibition of integrin αV with DI17E6 inhibits several prometastatic phenotypes of prostate cancer cells and therefore provide a rationale for further evaluation of DI17E6 for diminishing prostate cancer progression.Implications: This work identifies that therapeutic targeting of integrins containing an αV integrin unit inhibits cancer progression and thus may be of clinical benefit. Mol Cancer Res; 15(7); 875-83. ©2017 AACR.
Collapse
Affiliation(s)
- Yuan Jiang
- Department of Urology, University of Michigan, Ann Arbor, Michigan
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Jinlu Dai
- Department of Urology, University of Michigan, Ann Arbor, Michigan
| | - Zhi Yao
- Department of Immunology, Tianjin Medical University, Tianjin, China
| | - Greg Shelley
- Department of Urology, University of Michigan, Ann Arbor, Michigan
| | - Evan T Keller
- Department of Urology, University of Michigan, Ann Arbor, Michigan.
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
25
|
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
|
26
|
Yamamoto H, Ryu J, Min E, Oi N, Bai R, Zykova TA, Yu DH, Moriyama K, Bode AM, Dong Z. TRAF1 Is Critical for DMBA/Solar UVR-Induced Skin Carcinogenesis. J Invest Dermatol 2017; 137:1322-1332. [PMID: 28131816 DOI: 10.1016/j.jid.2016.12.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 12/27/2016] [Accepted: 12/30/2016] [Indexed: 12/15/2022]
Abstract
TRAF1 is a member of the TRAF protein family, which regulates the canonical and noncanonical NF-κB signaling cascades. Although aberrant TRAF1 expression in tumors has been reported, the role of TRAF1 remains elusive. Here, we report that TRAF1 is required for solar UV-induced skin carcinogenesis. Immunohistochemical analysis showed that TRAF1 expression is up-regulated in human actinic keratosis and squamous cell carcinoma. In vivo studies indicated that TRAF1 expression levels in mouse skin are induced by short-term solar UV irradiation, and a long-term skin carcinogenesis study showed that deletion of TRAF1 in mice results in a significant inhibition of skin tumor formation. Moreover, we show that TRAF1 is required for solar UV-induced extracellular signal-regulated kinase-5 (ERK5) phosphorylation and the expression of AP-1 family members (c-Fos/c-Jun). Mechanistic studies showed that TRAF1 expression enhances the ubiquitination of ERK5 on lysine 184, which is necessary for its kinase activity and AP-1 activation. Overall, our results suggest that TRAF1 mediates ERK5 activity by regulating the upstream effectors of ERK5 and also by modulating its ubiquitination status. Targeting TRAF1 function might lead to strategies for preventing and treating skin cancer.
Collapse
Affiliation(s)
- Hiroyuki Yamamoto
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Joohyun Ryu
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Eli Min
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Naomi Oi
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Ruihua Bai
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Tatyana A Zykova
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Dong Hoon Yu
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Kenji Moriyama
- Department of Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya, Hyogo, Japan
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA.
| |
Collapse
|
27
|
Liu F, Zhang H, Song H. Upregulation of MEK5 by Stat3 promotes breast cancer cell invasion and metastasis. Oncol Rep 2017; 37:83-90. [PMID: 27878304 DOI: 10.3892/or.2016.5256] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 10/03/2016] [Indexed: 02/05/2023] Open
Abstract
Mitogen extracellular-signal-regulated kinase kinase 5 (MEK5) plays an important role in promoting cell proliferation and tumorigenesis. The aberrant expression of MEK5 has been reported in various malignant diseases including cancers of breast, prostate, lung, colorectal and brain. However, the function and regulation of MEK5 signaling pathway are ambiguous and remain elusive with respect to its oncogenic roles in various cancers, especially in the regulation of the initiation and progression of cancer invasion and metastasis. Ectopic expression of MEK5 or knockdown of MEK5 by shRNA with in vitro cell based models demonstrated the role of MEK5 in regulation of epithelial mesenchymal transition (EMT) and breast cancer invasion and metastasis. Here, we show that MEK5 upregulated by Stat3 promotes breast cancer cell invasion through EMT. Further study demonstrated that Stat3 could bind to promoter region of MEK5 and enhanced MEK5 transcription and expression. In addition, the phosphorylation of MEK5 significantly increased in breast cancer cells corresponding to metastatic capability of breast cancer cells. The depletion of MEK5 by shRNA significantly decreased breast cancer invasion. Ectopic expression of MEK5 could confer non-invasive breast cancer cells to become invasion capable cells. Moreover, the phosphorylation of Erk5, a MEK5-regulated downstream kinase, was also upregulated consistent with the increased level of active MEK5. Our studies provide insights into a molecular mechanism by which MEK5 transcriptionally upregulated by Stat3 augments breast cancer cell EMT, which subsequently enhances cancer cell invasion and metastasis. This finding may suggest that Stat3 and MEK5/Erk5 pathways could be an effective therapeutic target for inhibition of breast cancer invasion and metastasis.
Collapse
Affiliation(s)
- Fang Liu
- Xi'an Jiaotong University Suzhou Academy, Suzhou, Jiangsu 215123, P.R. China
| | - Hao Zhang
- Shantou University Medical College Cancer Research Center, Shantou, Guangdong 515041, P.R. China
| | - Hui Song
- Xi'an Jiaotong University Suzhou Academy, Suzhou, Jiangsu 215123, P.R. China
| |
Collapse
|
28
|
Cimas FJ, Callejas-Valera JL, Pascual-Serra R, García-Cano J, Garcia-Gil E, De la Cruz-Morcillo MA, Ortega-Muelas M, Serrano-Oviedo L, Gutkind JS, Sánchez-Prieto R. MKP1 mediates chemosensitizer effects of E1a in response to cisplatin in non-small cell lung carcinoma cells. Oncotarget 2016; 6:44095-107. [PMID: 26689986 PMCID: PMC4792544 DOI: 10.18632/oncotarget.6574] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/25/2015] [Indexed: 12/19/2022] Open
Abstract
The adenoviral gene E1a is known to enhance the antitumor effect of cisplatin, one of the cornerstones of the current cancer chemotherapy. Here we study the molecular basis of E1a mediated sensitivity to cisplatin in an experimental model of Non-small cell lung cancer. Our data show how E1a blocks the induction of autophagy triggered by cisplatin and promotes the apoptotic response in resistant cells. Interestingly, at the molecular level, we present evidences showing how the phosphatase MKP1 is a major determinant of cisplatin sensitivity and its upregulation is strictly required for the induction of chemosensitivity mediated by E1a. Indeed, E1a is almost unable to promote sensitivity in H460, in which the high expression of MKP1 remains unaffected by E1a. However, in resistant cell as H1299, H23 or H661, which display low levels of MKP1, E1a expression promotes a dramatic increase in the amount of MKP1 correlating with cisplatin sensitivity. Furthermore, effective knock down of MKP1 in H1299 E1a expressing cells restores resistance to a similar extent than parental cells. In summary, the present work reinforce the critical role of MKP1 in the cellular response to cisplatin highlighting the importance of this phosphatase in future gene therapy approach based on E1a gene.
Collapse
Affiliation(s)
- Francisco J Cimas
- Unidad de Medicina Molecular, Laboratorio de Oncología, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain.,Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | | | - Raquel Pascual-Serra
- Unidad de Medicina Molecular, Laboratorio de Oncología, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain.,Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - Jesus García-Cano
- Unidad de Medicina Molecular, Laboratorio de Oncología, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain.,Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - Elena Garcia-Gil
- Unidad de Medicina Molecular, Laboratorio de Oncología, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain.,Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - Miguel A De la Cruz-Morcillo
- Unidad de Medicina Molecular, Laboratorio de Oncología, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain.,Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - Marta Ortega-Muelas
- Unidad de Medicina Molecular, Laboratorio de Oncología, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain.,Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | - Leticia Serrano-Oviedo
- Unidad de Medicina Molecular, Laboratorio de Oncología, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain.,Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| | | | - Ricardo Sánchez-Prieto
- Unidad de Medicina Molecular, Laboratorio de Oncología, Centro Regional de Investigaciones Biomédicas, Universidad de Castilla-La Mancha, Albacete, Spain.,Unidad de Biomedicina UCLM-CSIC, Albacete, Spain
| |
Collapse
|
29
|
Chen JH, Yang R, Zhang W, Wang YP. Functions of microRNA-143 in the apoptosis, invasion and migration of nasopharyngeal carcinoma. Exp Ther Med 2016; 12:3749-3755. [PMID: 28101165 DOI: 10.3892/etm.2016.3847] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 09/13/2016] [Indexed: 01/02/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a common cancer characterized by poor prognosis in areas of Southern China where it is endemic. microRNAs (miRNAs) are a class of naturally occurring small noncoding RNAs, some of which contribute to the initiation and development of cancer. The current study was designed to examine the expression level of miR-143 in NPC tissues. The potential functional targets of miR-143 involved in tumor apoptosis, invasion and migration were also investigated. Reverse transcription-quantitative polymerase chain reaction was used to evaluate the expression levels of miR-143 in clinical NPC specimens. Western blotting was used to explore the expression levels of extracellular signal regulated kinase (ERK)-5, Kirsten rat sarcoma viral oncogene homolog (KRAS), caspase 3 and B-cell lymphoma 2 (Bcl-2) in CNE-2Z cells following transfection with miR-143. Significantly decreased expression levels of miR-143 were observed in NPC tissues in comparison with matched normal nasopharyngeal tissues. Moreover, negative associations of miR-143 with tumor invasion depth, as well as lymph node metastasis were found. The enforced expression of miR-143 induced NPC cell apoptosis in addition to the suppression of growth, migration and invasion. The functions of miR-143 in NPC are mediated, at least in part, by the inhibition of ERK-5 activity and promotion of caspase 3 and KRAS expression. These findings suggest that miR-143 may function as a tumor suppressor in the development and progression of NPC.
Collapse
Affiliation(s)
- Jin-Hui Chen
- Department of Otorhinolaryngology, Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Rui Yang
- Department of Otorhinolaryngology, Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei Zhang
- Department of Otorhinolaryngology, Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yong-Ping Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
30
|
Zhuang K, Zhang J, Xiong M, Wang X, Luo X, Han L, Meng Y, Zhang Y, Liao W, Liu S. CDK5 functions as a tumor promoter in human colorectal cancer via modulating the ERK5-AP-1 axis. Cell Death Dis 2016; 7:e2415. [PMID: 27735944 PMCID: PMC5133995 DOI: 10.1038/cddis.2016.333] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/19/2016] [Accepted: 09/22/2016] [Indexed: 01/26/2023]
Abstract
Abnormal expression of cyclin-dependent kinase 5 (CDK5) has been found in several human cancers, whereas the role of CDK5 in the malignant development of colorectal cancer (CRC) has not been well characterized. Here we investigated the role of CDK5 in CRC and found that its expression was much higher in CRC tissues than that in normal tissues with a higher expression level of CDK5 closely correlating to advanced American Joint Committee on Cancer (AJCC) stage, poor differentiation, increased tumor size and poor prognosis of CRC. Biological function experiments showed that CDK5 regulated CRC cell proliferation and metastasis ability. Whole-genome microarray analysis, co-immunoprecipitation, in vitro kinase assay, western blotting, luciferase reporter assays and electrophoretic mobility shift assay (EMSA) showed that CDK5 could directly phosphorylate ERK5 at threonine (Thr) 732 and finally modulate the oncogenic ERK5–AP-1 axis. Further researches showed that CDK5–ERK5–AP-1 axis could promote progression of CRC carcinogenesis and had a significant correlation in human CRC samples. In summary, this study revealed the functional and mechanistic links between CDK5 and the oncogenic ERK5–AP-1 signaling pathway in the pathogenesis of CRC. These findings suggest that CDK5 has an important role in CRC development and may serve as a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Kangmin Zhuang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Juchang Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Man Xiong
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xianfei Wang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xiaobei Luo
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lu Han
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yan Meng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yali Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenting Liao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
31
|
ERK5 kinase activity is dispensable for cellular immune response and proliferation. Proc Natl Acad Sci U S A 2016; 113:11865-11870. [PMID: 27679845 DOI: 10.1073/pnas.1609019113] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Unlike other members of the MAPK family, ERK5 contains a large C-terminal domain with transcriptional activation capability in addition to an N-terminal canonical kinase domain. Genetic deletion of ERK5 is embryonic lethal, and tissue-restricted deletions have profound effects on erythroid development, cardiac function, and neurogenesis. In addition, depletion of ERK5 is antiinflammatory and antitumorigenic. Small molecule inhibition of ERK5 has been shown to have promising activity in cell and animal models of inflammation and oncology. Here we report the synthesis and biological characterization of potent, selective ERK5 inhibitors. In contrast to both genetic depletion/deletion of ERK5 and inhibition with previously reported compounds, inhibition of the kinase with the most selective of the new inhibitors had no antiinflammatory or antiproliferative activity. The source of efficacy in previously reported ERK5 inhibitors is shown to be off-target activity on bromodomains, conserved protein modules involved in recognition of acetyl-lysine residues during transcriptional processes. It is likely that phenotypes reported from genetic deletion or depletion of ERK5 arise from removal of a noncatalytic function of ERK5. The newly reported inhibitors should be useful in determining which of the many reported phenotypes are due to kinase activity and delineate which can be pharmacologically targeted.
Collapse
|
32
|
Gomez N, Erazo T, Lizcano JM. ERK5 and Cell Proliferation: Nuclear Localization Is What Matters. Front Cell Dev Biol 2016; 4:105. [PMID: 27713878 PMCID: PMC5031611 DOI: 10.3389/fcell.2016.00105] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 09/06/2016] [Indexed: 12/31/2022] Open
Abstract
ERK5, the last MAP kinase family member discovered, is activated by the upstream kinase MEK5 in response to growth factors and stress stimulation. MEK5-ERK5 pathway has been associated to different cellular processes, playing a crucial role in cell proliferation in normal and cancer cells by mechanisms that are both dependent and independent of its kinase activity. Thus, nuclear ERK5 activates transcription factors by either direct phosphorylation or acting as co-activator thanks to a unique transcriptional activation TAD domain located at its C-terminal tail. Consequently, ERK5 has been proposed as an interesting target to tackle different cancers, and either inhibitors of ERK5 activity or silencing the protein have shown antiproliferative activity in cancer cells and to block tumor growth in animal models. Here, we review the different mechanisms involved in ERK5 nuclear translocation and their consequences. Inactive ERK5 resides in the cytosol, forming a complex with Hsp90-Cdc37 superchaperone. In a canonical mechanism, MEK5-dependent activation results in ERK5 C-terminal autophosphorylation, Hsp90 dissociation, and nuclear translocation. This mechanism integrates signals such as growth factors and stresses that activate the MEK5-ERK5 pathway. Importantly, two other mechanisms, MEK5-independent, have been recently described. These mechanisms allow nuclear shuttling of kinase-inactive forms of ERK5. Although lacking kinase activity, these forms activate transcription by interacting with transcription factors through the TAD domain. Both mechanisms also require Hsp90 dissociation previous to nuclear translocation. One mechanism involves phosphorylation of the C-terminal tail of ERK5 by kinases that are activated during mitosis, such as Cyclin-dependent kinase-1. The second mechanism involves overexpression of chaperone Cdc37, an oncogene that is overexpressed in cancers such as prostate adenocarcinoma, where it collaborates with ERK5 to promote cell proliferation. Although some ERK5 kinase inhibitors have shown antiproliferative activity it is likely that those tumors expressing kinase-inactive nuclear ERK5 will not respond to these inhibitors.
Collapse
Affiliation(s)
| | | | - Jose M. Lizcano
- Protein Kinases and Signal Transduction Laboratory, Institut de Neurociencies and Departament de Bioquimica i Biologia Molecular, Facultat de Medicina, Universitat Autonoma de BarcelonaBarcelona, Spain
| |
Collapse
|
33
|
Zheng F, Zhang J, Luo S, Yi J, Wang P, Zheng Q, Wen Y. miR-143 is associated with proliferation and apoptosis involving ERK5 in HeLa cells. Oncol Lett 2016; 12:3021-3027. [PMID: 27698893 DOI: 10.3892/ol.2016.5016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 07/28/2016] [Indexed: 12/20/2022] Open
Abstract
Inappropriate expression of microRNA (miR) is strongly associated with carcinogenesis. miR-143 was reported to be one of the most prominent miRs implicated in the genesis and progression of human cancer. However, its correlation with cell proliferation and apoptosis in cervical cancer remains to be fully elucidated. In the present study, it was demonstrated that miR-143 is able to suppress the proliferation of cervical cancer HeLa cells and induce cell apoptosis in a time- and dose-dependent manner. The present study also investigated the potential targets of miR-143, extracellular-signal-regulated kinase 5 (ERK5) and its downstream substrate oncoprotein c-Fos, both of which are involved in cell proliferation and apoptosis. Upon increasing the miR-143 level, the ERK5 and c-Fos protein expression was significantly decreased without the effect of ERK5 transcription. Therefore, miR-143 is able to suppress cell proliferation and induce apoptosis in HeLa cells, potentially through negative regulation of ERK5 at its post-transcriptional stage.
Collapse
Affiliation(s)
- Fang Zheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jiahe Zhang
- Department of Public Health, Public Health Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Siyu Luo
- Department of Public Health, Public Health Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jing Yi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Ping Wang
- Department of Obstetrics and Gynecology, Shaanxi Tumor Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Quanqing Zheng
- Department of Public Health, Public Health Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yurong Wen
- Center for Translational Medicine, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P.R. China
| |
Collapse
|
34
|
Park SJ, Choi YS, Lee S, Lee YJ, Hong S, Han S, Kim BC. BIX02189 inhibits TGF-β1-induced lung cancer cell metastasis by directly targeting TGF-β type I receptor. Cancer Lett 2016; 381:314-22. [PMID: 27543359 DOI: 10.1016/j.canlet.2016.08.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/10/2016] [Accepted: 08/11/2016] [Indexed: 12/26/2022]
Abstract
Transforming growth factor-β1 (TGF-β1) promotes tumor metastasis by inducing an epithelial-to-mesenchymal transition (EMT) in cancer cells. In this study, we investigated the effects of BIX02189 and XMD8-92, pharmacologic inhibitors of the MEK5 [mitogen-activated protein kinase/extracellular-signal-regulated kinase (ERK)5] signaling pathway, on the EMT and migration of cancer cells induced by TGF-β1. In human A549 lung cancer cells, TGF-β1-induced EMT, cell motility, and expression of matrix metalloproteinase-2 were completely inhibited by BIX02189, but not by XMD8-92 or small interference RNAs specific to MEK5 and ERK5. Interestingly, BIX02189 strongly blocked the activation of TGF-β1 signaling components, and this inhibitory effect was not reproduced by MEK5 inhibition. Molecular docking simulation and kinase assays revealed that BIX02189 binds directly to the ATP-binding site of the TGF-β receptor type I (TβRI) and suppresses its kinase activity. Finally, the anti-metastatic effect of BIX02189 was validated in a TβRI-derived A549 xenograft mouse model. Collectively, these findings newly characterize BIX02189 as a potent inhibitor of TβRI that can block the tumor metastatic activity of TGF-β1.
Collapse
Affiliation(s)
- Seong Ji Park
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon 200-701, Republic of Korea
| | - Yu Sun Choi
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon 200-701, Republic of Korea
| | - Seungkoo Lee
- Department of Anatomic Pathology, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon 200-701, Republic of Korea
| | - Young Jae Lee
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Suntaek Hong
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Sanghwa Han
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon 200-701, Republic of Korea
| | - Byung-Chul Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon 200-701, Republic of Korea.
| |
Collapse
|
35
|
Myers SM, Bawn RH, Bisset LC, Blackburn TJ, Cottyn B, Molyneux L, Wong AC, Cano C, Clegg W, Harrington RW, Leung H, Rigoreau L, Vidot S, Golding BT, Griffin RJ, Hammonds T, Newell DR, Hardcastle IR. High-Throughput Screening and Hit Validation of Extracellular-Related Kinase 5 (ERK5) Inhibitors. ACS COMBINATORIAL SCIENCE 2016; 18:444-55. [PMID: 27400250 DOI: 10.1021/acscombsci.5b00155] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The extracellular-related kinase 5 (ERK5) is a promising target for cancer therapy. A high-throughput screen was developed for ERK5, based on the IMAP FP progressive binding system, and used to identify hits from a library of 57 617 compounds. Four distinct chemical series were evident within the screening hits. Resynthesis and reassay of the hits demonstrated that one series did not return active compounds, whereas three series returned active hits. Structure-activity studies demonstrated that the 4-benzoylpyrrole-2-carboxamide pharmacophore had excellent potential for further development. The minimum kinase binding pharmacophore was identified, and key examples demonstrated good selectivity for ERK5 over p38α kinase.
Collapse
Affiliation(s)
- Stephanie M Myers
- Newcastle Cancer Centre, Northern Institute for Cancer Research and School of Chemistry, Bedson Building, Newcastle University , Newcastle upon Tyne, NE1 7RU, U.K
| | - Ruth H Bawn
- Newcastle Cancer Centre, Northern Institute for Cancer Research and School of Chemistry, Bedson Building, Newcastle University , Newcastle upon Tyne, NE1 7RU, U.K
| | - Louise C Bisset
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Medical School, Framlington Place, Newcastle University , Paul O'Gorman Building, Newcastle upon Tyne, NE2 4HH, U.K
| | - Timothy J Blackburn
- Newcastle Cancer Centre, Northern Institute for Cancer Research and School of Chemistry, Bedson Building, Newcastle University , Newcastle upon Tyne, NE1 7RU, U.K
| | - Betty Cottyn
- Newcastle Cancer Centre, Northern Institute for Cancer Research and School of Chemistry, Bedson Building, Newcastle University , Newcastle upon Tyne, NE1 7RU, U.K
| | - Lauren Molyneux
- Newcastle Cancer Centre, Northern Institute for Cancer Research and School of Chemistry, Bedson Building, Newcastle University , Newcastle upon Tyne, NE1 7RU, U.K
| | - Ai-Ching Wong
- Cancer Research Technology, Ltd., Discovery Laboratories, Wolfson Institute for Biomedical Research, The Cruciform Building, Gower Street, London, WC1E 6BT, U.K
| | - Celine Cano
- Newcastle Cancer Centre, Northern Institute for Cancer Research and School of Chemistry, Bedson Building, Newcastle University , Newcastle upon Tyne, NE1 7RU, U.K
| | - William Clegg
- School of Chemistry, Newcastle University , Bedson Building, Newcastle upon Tyne, NE1 7RU, U.K
| | - Ross W Harrington
- School of Chemistry, Newcastle University , Bedson Building, Newcastle upon Tyne, NE1 7RU, U.K
| | - Hing Leung
- The Beatson Institute for Cancer Research , Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, U.K
| | - Laurent Rigoreau
- Cancer Research Technology, Ltd., Discovery Laboratories, Wolfson Institute for Biomedical Research, The Cruciform Building, Gower Street, London, WC1E 6BT, U.K
| | - Sandrine Vidot
- Newcastle Cancer Centre, Northern Institute for Cancer Research and School of Chemistry, Bedson Building, Newcastle University , Newcastle upon Tyne, NE1 7RU, U.K
| | - Bernard T Golding
- Newcastle Cancer Centre, Northern Institute for Cancer Research and School of Chemistry, Bedson Building, Newcastle University , Newcastle upon Tyne, NE1 7RU, U.K
| | - Roger J Griffin
- Newcastle Cancer Centre, Northern Institute for Cancer Research and School of Chemistry, Bedson Building, Newcastle University , Newcastle upon Tyne, NE1 7RU, U.K
| | - Tim Hammonds
- Cancer Research Technology, Ltd., Discovery Laboratories, Wolfson Institute for Biomedical Research, The Cruciform Building, Gower Street, London, WC1E 6BT, U.K
| | - David R Newell
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Medical School, Framlington Place, Newcastle University , Paul O'Gorman Building, Newcastle upon Tyne, NE2 4HH, U.K
| | - Ian R Hardcastle
- Newcastle Cancer Centre, Northern Institute for Cancer Research and School of Chemistry, Bedson Building, Newcastle University , Newcastle upon Tyne, NE1 7RU, U.K
| |
Collapse
|
36
|
Tsioumpekou M, Papadopoulos N, Burovic F, Heldin CH, Lennartsson J. Platelet-derived growth factor (PDGF)-induced activation of Erk5 MAP-kinase is dependent on Mekk2, Mek1/2, PKC and PI3-kinase, and affects BMP signaling. Cell Signal 2016; 28:1422-1431. [PMID: 27339033 DOI: 10.1016/j.cellsig.2016.06.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 06/16/2016] [Accepted: 06/16/2016] [Indexed: 02/09/2023]
Abstract
Platelet-derived growth factor-BB (PDGF-BB) binds to its tyrosine kinase receptors (PDGFRs) and stimulates mitogenicity and survival of cells of mesenchymal origin. Activation of PDGFRs initiates a number of downstream signaling pathways, including phosphatidyl 3'-inositol kinase (PI3-kinase), phospholipase Cγ and MAP kinase pathways. In this report, we show that Erk5 MAP kinase is activated in response to PDGF-BB in the smooth muscle cell line MOVAS in a manner dependent on Mekk2, Mek1/2, Mek5, PI3-kinase and protein kinase C (PKC). The co-operation of Mek1/2 and Mekk2 in the activation of Erk5, suggests a close co-regulation between the Erk1/2 and Erk5 MAP kinase pathways. Furthermore, we found that classical PKCs are important for Erk5 activation. In addition, we found that PKCζ interacts with Erk5 and may exert a negative feed-back effect. We observed no nuclear accumulation of Erk5 in response to PDGF-BB stimulation, however, we identified a mechanism by which cytoplasmic Erk5 influences gene expression; Erk5 was essential for PDGF-BB-mediated Smad1/5/8 signaling by stimulating release and/or activation of bone morphogenetic protein(s) (BMPs). Thus, PDGF-BB-induced Erk5 activation involves parallel stimulatory and inhibitory pathways and promotes Smad1/5/8 signaling.
Collapse
Affiliation(s)
- Maria Tsioumpekou
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE-751 24 Uppsala, Sweden
| | - Natalia Papadopoulos
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE-751 24 Uppsala, Sweden
| | - Fatima Burovic
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE-751 24 Uppsala, Sweden
| | - Carl-Henrik Heldin
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE-751 24 Uppsala, Sweden
| | - Johan Lennartsson
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE-751 24 Uppsala, Sweden.
| |
Collapse
|
37
|
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
|
38
|
Garbuzenko DV, Arefyev NO, Belov DV. Mechanisms of adaptation of the hepatic vasculature to the deteriorating conditions of blood circulation in liver cirrhosis. World J Hepatol 2016; 8:665-672. [PMID: 27326313 PMCID: PMC4909428 DOI: 10.4254/wjh.v8.i16.665] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 04/25/2016] [Accepted: 05/17/2016] [Indexed: 02/06/2023] Open
Abstract
PubMed, EMBASE, Orphanet, MIDLINE, Google Scholar and Cochrane Library were searched for articles published between 1983 and 2015. Relevant articles were selected by using the following terms: "Liver cirrhosis", "Endothelial dysfunction", "Sinusoidal remodeling", "Intrahepatic angiogenesis" and "Pathogenesis of portal hypertension". Then the reference lists of identified articles were searched for other relevant publications as well. Besides gross hepatic structural disorders related to diffuse fibrosis and formation of regenerative nodules, the complex morphofunctional rearrangement of the hepatic microvascular bed and intrahepatic angiogenesis also play important roles in hemodynamic disturbances in liver cirrhosis. It is characterized by endothelial dysfunction and impaired paracrine interaction between activated stellate hepatocytes and sinusoidal endotheliocytes, sinusoidal remodeling and capillarization, as well as development of the collateral microcirculation. In spite of the fact that complex morphofunctional rearrangement of the hepatic microvascular bed and intrahepatic angiogenesis in liver cirrhosis are the compensatory-adaptive reaction to the deteriorating conditions of blood circulation, they contribute to progression of disease and development of serious complications, in particular, related to portal hypertension.
Collapse
Affiliation(s)
- Dmitry Victorovich Garbuzenko
- Dmitry Victorovich Garbuzenko, Nikolay Olegovich Arefyev, Department of Faculty Surgery, South Ural State Medical University, 454092 Chelyabinsk, Russia
| | - Nikolay Olegovich Arefyev
- Dmitry Victorovich Garbuzenko, Nikolay Olegovich Arefyev, Department of Faculty Surgery, South Ural State Medical University, 454092 Chelyabinsk, Russia
| | - Dmitry Vladimirovich Belov
- Dmitry Victorovich Garbuzenko, Nikolay Olegovich Arefyev, Department of Faculty Surgery, South Ural State Medical University, 454092 Chelyabinsk, Russia
| |
Collapse
|
39
|
The role of MAPK signaling pathway in the Her-2-positive meningiomas. Oncol Rep 2016; 36:685-95. [PMID: 27279438 PMCID: PMC4933551 DOI: 10.3892/or.2016.4849] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Accepted: 03/18/2016] [Indexed: 01/16/2023] Open
Abstract
Meningiomas are common types of adult nerve system tumors. Although most cases are considered benign, due to its high rate of recurrence and easy malignant progression to anaplastic meningioma they present a puzzle for the current treatment. The HER-2 oncogene has important value for meningioma cells development and progression. So far, little is known about the effect on the exact underlying signal pathway and molecular mechanisms of HER-2-positive meningioma cells. The goal of the present study was to determine the effects of HER-2 gene and possible involvement of MAPK signal pathway in human malignant meningioma. We applied q-PCR analysis, immunofluorescence (IF) staining, western blot analysis, animal model, MAPK inhibition, MTT assay and cell invasion analysis for the investigation. The results demonstrated that the downregulation of the expression of HER-2 significantly inhibited cell motility and proliferation of human meningioma cells in vivo. Accordingly, in the HER-2-overexpression meningioma cells with the inhibition of ERK1/2, ERK5, JNK, in the cells with the ERK1/2, ERK5 inhibition, protein expression was markedly suppressed as well as the cell proliferation resistance. No difference was observed in the HER-2-overexpression meningioma cells with the inhibition of JNK. These findings suggest that HER-2 gene can affect the proliferation ability of human meningioma cells in vivo and MAPK signal pathway may contribute to the carcinogenesis and development of human meningiomas combinating with HER-2.
Collapse
|
40
|
Wu L, Chen J, Qin Y, Mo X, Huang M, Ru H, Yang Y, Liu J, Lin Y. SATB2 suppresses gastric cancer cell proliferation and migration. Tumour Biol 2016; 37:4597-4602. [PMID: 26508023 DOI: 10.1007/s13277-015-4282-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 10/19/2015] [Indexed: 02/01/2023] Open
Abstract
Gastric cancer is one of the death-related malignant tumors worldwide. It remains a challenge for the diagnosis and treatment of gastric cancer. Special AT-rich sequence-binding protein 2 (SATB2) is a new tumor suppressive gene and plays important roles in many cancers. However, the role of SATB2 in gastric cancer is still unknown. In the present study, we demonstrated that downregulation of SATB2 was associated with shortened survival in patients with gastric cancer. Ectopic expression of SATB2 inhibited gastric cancer cell proliferation, colony formation, and migration. Overexpression of SATB2 repressed the expression of extracellular signal-regulated kinase 5 (ERK5), and activation of ERK5 restored the SATB2-induced inhibition of proliferation and migration in gastric cancer. This study provided evidence that SATB2 acted as a tumor suppressive gene gastric cancer, serving as a potential therapeutic target.
Collapse
Affiliation(s)
- Liucheng Wu
- Gastrointestinal Surgery Department, Guangxi Medical University, Tumor Hospital, Nanning, Guangxi, 530021, People's Republic of China.
| | - Jiansi Chen
- Gastrointestinal Surgery Department, Guangxi Medical University, Tumor Hospital, Nanning, Guangxi, 530021, People's Republic of China
| | - Yuzhou Qin
- Gastrointestinal Surgery Department, Guangxi Medical University, Tumor Hospital, Nanning, Guangxi, 530021, People's Republic of China
| | - Xianwei Mo
- Gastrointestinal Surgery Department, Guangxi Medical University, Tumor Hospital, Nanning, Guangxi, 530021, People's Republic of China
| | - Minwei Huang
- Gastrointestinal Surgery Department, Guangxi Medical University, Tumor Hospital, Nanning, Guangxi, 530021, People's Republic of China
| | - Haiming Ru
- Gastrointestinal Surgery Department, Guangxi Medical University, Tumor Hospital, Nanning, Guangxi, 530021, People's Republic of China
| | - Yang Yang
- Gastrointestinal Surgery Department, Guangxi Medical University, Tumor Hospital, Nanning, Guangxi, 530021, People's Republic of China
| | - Jungang Liu
- Gastrointestinal Surgery Department, Guangxi Medical University, Tumor Hospital, Nanning, Guangxi, 530021, People's Republic of China
| | - Yuan Lin
- Gastrointestinal Surgery Department, Guangxi Medical University, Tumor Hospital, Nanning, Guangxi, 530021, People's Republic of China
| |
Collapse
|
41
|
Shen H, McHale CM, Haider SI, Jung C, Zhang S, Smith MT, Zhang L. Identification of Genes That Modulate Susceptibility to Formaldehyde and Imatinib by Functional Genomic Screening in Human Haploid KBM7 Cells. Toxicol Sci 2016; 151:10-22. [PMID: 27008852 DOI: 10.1093/toxsci/kfw032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Though current functional genomic screening systems are useful for investigating human susceptibility to chemical toxicity, they have limitations. Well-established, high-throughput yeast mutant screens identify only evolutionarily conserved processes. RNA interference can be applied in human cells but is limited by incomplete gene knockout and off-target effects. Human haploid cell screening is advantageous as it requires knockdown of only a single copy of each gene. A human haploid cell mutant library (KBM7-Mu), derived from a chronic myeloid leukemia (CML) patient, was recently developed and has been used to identify genes that modulate sensitivity to infectious agents and pharmaceutical drugs. Here, we sought to improve the KBM7-Mu screening process to enable efficient screening of environmental chemicals. We developed a semi-solid medium based screening approach that cultures individual mutant colonies from chemically resistant cells, faster (by 2-3 weeks) and with less labor than the original liquid medium-based approach. As proof of principle, we identified genetic mutants that confer resistance to the carcinogen formaldehyde (FA, 12 genes, 18 hits) and the CML chemotherapeutic agent imatinib (6 genes, 13 hits). Validation experiments conducted on KBM7 mutants lacking each of the 18 genes confirmed resistance of 6 FA mutants (CTC1, FCRLA, GOT1, LPR5, M1AP, and MAP2K5) and 1 imatinib-resistant mutant (LYRM9). Despite the improvements to the method, it remains technically challenging to limit false positive findings. Nonetheless, our findings demonstrate the broad applicability of this optimized haploid approach to screen toxic chemicals to identify novel susceptibility genes and gain insight into potential mechanisms of toxicity.
Collapse
Affiliation(s)
- Hua Shen
- Superfund Research Program, School of Public Health, University of California, Berkeley, California 94720
| | - Cliona M McHale
- Superfund Research Program, School of Public Health, University of California, Berkeley, California 94720
| | - Syed I Haider
- Superfund Research Program, School of Public Health, University of California, Berkeley, California 94720
| | - Cham Jung
- Superfund Research Program, School of Public Health, University of California, Berkeley, California 94720
| | - Susie Zhang
- Superfund Research Program, School of Public Health, University of California, Berkeley, California 94720
| | - Martyn T Smith
- Superfund Research Program, School of Public Health, University of California, Berkeley, California 94720
| | - Luoping Zhang
- Superfund Research Program, School of Public Health, University of California, Berkeley, California 94720
| |
Collapse
|
42
|
Giurisato E, Tournier C. Can tumor cells proliferate without ERK5? Cell Cycle 2016; 15:619-20. [PMID: 26822978 PMCID: PMC4845921 DOI: 10.1080/15384101.2016.1143272] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 01/13/2016] [Indexed: 10/22/2022] Open
Affiliation(s)
- Emanuele Giurisato
- Faculty of Life Sciences, University of Manchester, Manchester, UK
- Department of Developmental and Molecular Medicine, University of Siena, Siena, Italy
| | - Cathy Tournier
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
43
|
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
|
44
|
Regad T. Targeting RTK Signaling Pathways in Cancer. Cancers (Basel) 2015; 7:1758-84. [PMID: 26404379 PMCID: PMC4586793 DOI: 10.3390/cancers7030860] [Citation(s) in RCA: 286] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 08/24/2015] [Accepted: 08/26/2015] [Indexed: 12/21/2022] Open
Abstract
The RAS/MAP kinase and the RAS/PI3K/AKT pathways play a key role in the regulation of proliferation, differentiation and survival. The induction of these pathways depends on Receptor Tyrosine Kinases (RTKs) that are activated upon ligand binding. In cancer, constitutive and aberrant activations of components of those pathways result in increased proliferation, survival and metastasis. For instance, mutations affecting RTKs, Ras, B-Raf, PI3K and AKT are common in perpetuating the malignancy of several types of cancers and from different tissue origins. Therefore, these signaling pathways became prime targets for cancer therapy. This review aims to provide an overview about the most frequently encountered mutations, the pathogenesis that results from such mutations and the known therapeutic strategies developed to counteract their aberrant functions.
Collapse
Affiliation(s)
- Tarik Regad
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Clifton Lane, NG11 8NS Nottingham, UK.
| |
Collapse
|
45
|
Rovida E, Di Maira G, Tusa I, Cannito S, Paternostro C, Navari N, Vivoli E, Deng X, Gray NS, Esparís-Ogando A, David E, Pandiella A, Dello Sbarba P, Parola M, Marra F. The mitogen-activated protein kinase ERK5 regulates the development and growth of hepatocellular carcinoma. Gut 2015; 64:1454-65. [PMID: 25183205 DOI: 10.1136/gutjnl-2014-306761] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 08/04/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The extracellular signal-regulated kinase 5 (ERK5 or BMK1) is involved in tumour development. The ERK5 gene may be amplified in hepatocellular carcinoma (HCC), but its biological role has not been clarified. In this study, we explored the role of ERK5 expression and activity in HCC in vitro and in vivo. DESIGN ERK5 expression was evaluated in human liver tissue. Cultured HepG2 and Huh-7 were studied after ERK5 knockdown by siRNA or in the presence of the specific pharmacological inhibitor, XMD8-92. The role of ERK5 in vivo was assessed using mouse Huh-7 xenografts. RESULTS In tissue specimens from patients with HCC, a higher percentage of cells with nuclear ERK5 expression was found both in HCC and in the surrounding cirrhotic tissue compared with normal liver tissue. Inhibition of ERK5 decreased HCC cell proliferation and increased the proportion of cells in G0/G1 phase. These effects were associated with increased expression of p27 and p15 and decreased CCND1. Treatment with XMD8-92 or ERK5 silencing prevented cell migration induced by epidermal growth factor or hypoxia and caused cytoskeletal remodelling. In mouse xenografts, the rate of tumour appearance and the size of tumours were significantly lower when Huh-7 was silenced for ERK5. Moreover, systemic treatment with XMD8-92 of mice with established HCC xenografts markedly reduced tumour growth and decreased the expression of the proto-oncogene c-Rel. CONCLUSIONS ERK5 regulates the biology of HCC cells and modulates tumour development and growth in vivo. This pathway should be investigated as a possible therapeutic target in HCC.
Collapse
Affiliation(s)
- Elisabetta Rovida
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche, Università di Firenze, Italy
| | - Giovanni Di Maira
- Dipartimento di Medicina Sperimentale e Clinica Università di Firenze, Italy
| | - Ignazia Tusa
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche, Università di Firenze, Italy
| | - Stefania Cannito
- Dipartimento di Medicina e Oncologia Sperimentali, Università di Torino, Italy
| | - Claudia Paternostro
- Dipartimento di Medicina e Oncologia Sperimentali, Università di Torino, Italy
| | - Nadia Navari
- Dipartimento di Medicina Sperimentale e Clinica Università di Firenze, Italy
| | - Elisa Vivoli
- Dipartimento di Medicina Sperimentale e Clinica Università di Firenze, Italy
| | - Xianming Deng
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Nathanael S Gray
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - Azucena Esparís-Ogando
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Spain
| | - Ezio David
- Pathology Unit, Ospedale S. Giovanni Battista, Torino, Italy
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Spain
| | - Persio Dello Sbarba
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche, Università di Firenze, Italy
| | - Maurizio Parola
- Dipartimento di Medicina e Oncologia Sperimentali, Università di Torino, Italy
| | - Fabio Marra
- Dipartimento di Medicina Sperimentale e Clinica Università di Firenze, Italy
| |
Collapse
|
46
|
Rovida E, Stecca B. Mitogen-activated protein kinases and Hedgehog-GLI signaling in cancer: A crosstalk providing therapeutic opportunities? Semin Cancer Biol 2015; 35:154-67. [PMID: 26292171 DOI: 10.1016/j.semcancer.2015.08.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/07/2015] [Accepted: 08/10/2015] [Indexed: 01/07/2023]
Abstract
The Hedgehog-GLI (HH-GLI) signaling is of critical importance during embryonic development, where it regulates a number of cellular processes, including patterning, proliferation and differentiation. Its aberrant activation has been linked to several types of cancer. HH-GLI signaling is triggered by binding of ligands to the transmembrane receptor patched and is subsequently mediated by transcriptional effectors belonging to the GLI family, whose function is fine tuned by a series of molecular interactions and modifications. Several HH-GLI inhibitors have been developed and are in clinical trials. Similarly, the mitogen-activated protein kinases (MAPK) are involved in a number of biological processes and play an important role in many diseases including cancer. Inhibiting molecules targeting MAPK signaling, especially those elicited by the MEK1/2-ERK1/2 pathway, have been developed and are moving into clinical trials. ERK1/2 may be activated as a consequence of aberrant activation of upstream signaling molecules or during development of drug resistance following treatment with kinase inhibitors such as those for PI3K or BRAF. Evidence of a crosstalk between HH-GLI and other oncogenic signaling pathways has been reported in many tumor types, as shown by recent reviews. Here we will focus on the interaction between HH-GLI and the final MAPK effectors ERK1/2, p38 and JNK in cancer in view of its possible implications for cancer therapy. Several reports highlight the existence of a consistent crosstalk between HH signaling and MAPK, especially with the MEK1/2-ERK1/2 pathway, and this fact should be taken into consideration for designing optimal treatment and prevent tumor relapse.
Collapse
Affiliation(s)
- Elisabetta Rovida
- Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Sezione di Patologia, Università degli Studi di Firenze, Firenze, Italy
| | - Barbara Stecca
- Laboratory of Tumor Cell Biology, Core Research Laboratory-Istituto Toscano Tumori (CRL-ITT), Florence, Italy; Department of Oncology, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy.
| |
Collapse
|
47
|
Utility of microRNAs and siRNAs in cervical carcinogenesis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:374924. [PMID: 25874209 PMCID: PMC4385600 DOI: 10.1155/2015/374924] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 10/13/2014] [Accepted: 10/14/2014] [Indexed: 01/08/2023]
Abstract
MicroRNAs and siRNAs belong to a family of small noncoding RNAs which bind through partial sequence complementarity to 3'-UTR regions of mRNA from target genes, resulting in the regulation of gene expression. MicroRNAs have become an attractive target for genetic and pharmacological modulation due to the critical function of their target proteins in several signaling pathways, and their expression profiles have been found to be altered in various cancers. A promising technology platform for selective silencing of cell and/or viral gene expression using siRNAs is currently in development. Cervical cancer is the most common cancer in women in the developing world and sexually transmitted infection with HPV is the cause of this malignancy. Therefore, a cascade of abnormal events is induced during cervical carcinogenesis, including the induction of genomic instability, reprogramming of cellular metabolic pathways, deregulation of cell proliferation, inhibition of apoptotic mechanisms, disruption of cell cycle control mechanisms, and alteration of gene expression. Thus, in the present review article, we highlight new research on microRNA expression profiles which may be utilized as biomarkers for cervical cancer. Furthermore, we discuss selective silencing of HPV E6 and E7 with siRNAs which represents a potential gene therapy strategy against cervical cancer.
Collapse
|
48
|
Mansour MA, Hyodo T, Ito S, Kurita K, Kokuryo T, Uehara K, Nagino M, Takahashi M, Hamaguchi M, Senga T. SATB2 suppresses the progression of colorectal cancer cells via inactivation of MEK5/ERK5 signaling. FEBS J 2015; 282:1394-405. [DOI: 10.1111/febs.13227] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/15/2015] [Accepted: 02/04/2015] [Indexed: 12/16/2022]
Affiliation(s)
- Mohammed A. Mansour
- Division of Cancer Biology; Nagoya University Graduate School of Medicine; Nagoya Japan
- Biochemistry Section; Department of Chemistry; Faculty of Science; Tanta University; Egypt
| | - Toshinori Hyodo
- Division of Cancer Biology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Satoko Ito
- Division of Cancer Biology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Kenji Kurita
- Department of Surgical Oncology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Toshio Kokuryo
- Department of Surgical Oncology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Keisuke Uehara
- Department of Surgical Oncology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Masato Nagino
- Department of Surgical Oncology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Masahide Takahashi
- Department of Pathology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Michinari Hamaguchi
- Division of Cancer Biology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Takeshi Senga
- Division of Cancer Biology; Nagoya University Graduate School of Medicine; Nagoya Japan
| |
Collapse
|
49
|
Finegan KG, Perez-Madrigal D, Hitchin JR, Davies CC, Jordan AM, Tournier C. ERK5 is a critical mediator of inflammation-driven cancer. Cancer Res 2015; 75:742-53. [PMID: 25649771 PMCID: PMC4333217 DOI: 10.1158/0008-5472.can-13-3043] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Chronic inflammation is a hallmark of many cancers, yet the pathogenic mechanisms that distinguish cancer-associated inflammation from benign persistent inflammation are still mainly unclear. Here, we report that the protein kinase ERK5 controls the expression of a specific subset of inflammatory mediators in the mouse epidermis, which triggers the recruitment of inflammatory cells needed to support skin carcinogenesis. Accordingly, inactivation of ERK5 in keratinocytes prevents inflammation-driven tumorigenesis in this model. In addition, we found that anti-ERK5 therapy cooperates synergistically with existing antimitotic regimens, enabling efficacy of subtherapeutic doses. Collectively, our findings identified ERK5 as a mediator of cancer-associated inflammation in the setting of epidermal carcinogenesis. Considering that ERK5 is expressed in almost all tumor types, our findings suggest that targeting tumor-associated inflammation via anti-ERK5 therapy may have broad implications for the treatment of human tumors.
Collapse
Affiliation(s)
- Katherine G Finegan
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom.
| | | | - James R Hitchin
- Drug Discovery Unit Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Clare C Davies
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Allan M Jordan
- Drug Discovery Unit Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Cathy Tournier
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
50
|
Poulsen A, William AD, Dymock BW. Designed Macrocyclic Kinase Inhibitors. MACROCYCLES IN DRUG DISCOVERY 2014. [DOI: 10.1039/9781782623113-00141] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Cancer continues to present as an increasing and serious global unmet medical need in today's aging population.1 Macrocyclic kinase inhibitors have reached advanced clinical testing and are making an impact in oncologic conditions including myelofibrosis, lymphomas and leukemias. Rheumatoid arthritis (RA) is also beginning to be impacted with the first macrocycle having entered Phase I clinical evaluation in healthy volunteers. Increasing reports of innovative macrocycles in preclinical research are appearing in the literature. Desirable, selective, multi-kinase inhibitory profiles against specific kinases known to be abrogated in cancer, RA, and other diseases have been achieved in a first generation series of clinical stage compact small molecule macrocyclic kinase inhibitors. Herein we discuss their design, synthesis, structure activity relationships and assessment of the latest clinical data in a range of oncologic conditions. Macrocyclic kinase inhibitors have the potential to offer new hope to patients and their families.
Collapse
Affiliation(s)
- Anders Poulsen
- Experimental Therapeutics Centre, A*STAR 11 Biopolis Way, #03-10/11 The Helios 138667 Singapore
| | - Anthony D. William
- Institute of Chemical and Engineering Sciences, A*STAR 11 Biopolis Way, The Helios #03-08 138667 Singapore
| | - Brian W. Dymock
- Department of Pharmacy, National University of Singapore 18 Science Drive 4 117543 Singapore
| |
Collapse
|