1
|
Tusa I, Gagliardi S, Menconi A, Maresca L, Tubita A, Lulli M, Stecca B, Rovida E. The MEK5/ERK5 pathway promotes the activation of the Hedgehog/GLI signaling in melanoma cells. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01050-z. [PMID: 39998753 DOI: 10.1007/s13402-025-01050-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2025] [Indexed: 02/27/2025] Open
Abstract
PURPOSE Malignant melanoma is the deadliest skin cancer, with a poor prognosis in advanced stages. We reported that both Hedgehog-GLI (HH/GLI) and Mitogen-activated protein Kinase (MAPK) extracellular signal-regulated kinase 5 (ERK5) pathways promote melanoma growth, and that ERK5 activation is required for HH/GLI-dependent melanoma cell proliferation. Here, we explored whether ERK5 regulates HH/GLI signaling. METHODS Both genetic (using ERK5-specific shRNA) and pharmacologic (using the ERK5 inhibitors JWG-071 and AX15836, and the MAPK/ERK kinase 5, MEK5 inhibitors GW284543 and BIX02189) targeting approaches were used. Luciferase assay using the GLI-binding site luciferase reporter was performed to evaluate GLI transcriptional activity. A constitutively active form of MEK5 (MEK5DD) was used to induce ERK5 activation. 3D spheroid assays were performed in melanoma cells. RESULTS Genetic and pharmacologic ERK5 inhibition reduces GLI1 and GLI2 protein levels and transcriptional activity of endogenous HH/GLI pathway induced by the agonist SAG in NIH/3T3 cells. In these cells, MEK5DD overexpression potentiates transcriptional activity of endogenous HH/GLI pathway induced by SAG, whereas ERK5 silencing prevents this effect. Consistently, MEK5DD overexpression increases GLI1 and GLI2 protein levels. In melanoma cells, ERK5 silencing reduces GLI1 and GLI2 mRNA and protein levels and inhibits GLI transcriptional activity. MEK5DD further increases the transcriptional activity of the HH/GLI pathway and GLI1 protein levels. Combination of GLI and MEK5 inhibitors is more effective than single treatments in reducing melanoma spheroid growth. CONCLUSIONS MEK5-ERK5 is an activator of GLI transcription factors, and combined targeting of these pathways warrants further preclinical investigation as a potential innovative therapeutic approach for melanoma.
Collapse
Affiliation(s)
- Ignazia Tusa
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy.
| | - Sinforosa Gagliardi
- Core Research Laboratory - Institute for Cancer Research and Prevention (ISPRO), Florence, Italy
| | - Alessio Menconi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
| | - Luisa Maresca
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
- Core Research Laboratory - Institute for Cancer Research and Prevention (ISPRO), Florence, Italy
| | - Alessandro Tubita
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy
| | - Barbara Stecca
- Core Research Laboratory - Institute for Cancer Research and Prevention (ISPRO), Florence, Italy.
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134, Florence, Italy.
| |
Collapse
|
2
|
Wissemann J, Heidenreich A, Zimmermann H, Engelmann J, Jansen J, Suchanek D, Westermann D, Wolf D, Stachon P, Merz J. ADP as a novel stimulus for NLRP3-inflammasome activation in mice fails to translate to humans. Purinergic Signal 2024; 20:291-302. [PMID: 37410223 PMCID: PMC11189352 DOI: 10.1007/s11302-023-09953-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/21/2023] [Indexed: 07/07/2023] Open
Abstract
The NLRP3-inflammasome is a cytosolic multiprotein complex that triggers an inflammatory response to certain danger signals. Recently adenosine diphosphate (ADP) was found to activate the NLRP3-inflammasome in murine macrophages via the P2Y1 receptor. Blockade of this signaling pathway reduced disease severity in a murine colitis-model. However, the role of the ADP/P2Y1-axis has not yet been studied in humans. This present study confirmed ADP-dependent NLRP3-inflammasome activation in murine macrophages, but found no evidence for a role of ADP in inflammasome activation in humans. We investigated the THP1 cell line as well as primary monocytes and further looked at macrophages. Although all cells express the three human ADP-receptors P2Y1, P2Y12 and P2Y13, independent of priming, neither increased ASC-speck formation could be detected with flow cytometry nor additional IL-1β release be found in the culture supernatant of ADP stimulated cells. We now show for the first time that the responsiveness of monocytes and macrophages to ADP as well as the regulation of its purinergic receptors is very much dependent on the species. Therefore the signaling pathway found to contribute to colitis in mice is likely not applicable to humans.
Collapse
Affiliation(s)
- Julius Wissemann
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Adrian Heidenreich
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Helene Zimmermann
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Juliane Engelmann
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Jasper Jansen
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Dymphie Suchanek
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Dirk Westermann
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Dennis Wolf
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Peter Stachon
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Julian Merz
- Cardiology and Angiology, Medical Center, University Heart Center Freiburg-Bad Krozingen, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
3
|
Chen F, Wu P, Zhang H, Sun G. Signaling Pathways Triggering Therapeutic Hydrogels in Promoting Chronic Wound Healing. Macromol Biosci 2024; 24:e2300217. [PMID: 37831962 DOI: 10.1002/mabi.202300217] [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: 05/16/2023] [Revised: 10/08/2023] [Indexed: 10/15/2023]
Abstract
In recent years, there has been a significant increase in the prevalence of chronic wounds, such as pressure ulcers, diabetic foot ulcers, and venous ulcers of the lower extremities. The main contributors to chronic wound formation are bacterial infection, prolonged inflammation, and peripheral vascular disease. However, effectively treating these chronic wounds remains a global challenge. Hydrogels have extensively explored as wound healing dressing because of their excellent biocompatibility and structural similarity to extracellular matrix (ECM). Nonetheless, much is still unknown how the hydrogels promote wound repair and regeneration. Signaling pathways play critical roles in wound healing process by controlling and coordinating cells and biomolecules. Hydrogels, along with their therapeutic ingredients that impact signaling pathways, have the potential to significantly enhance the wound healing process and its ultimate outcomes. Understanding this interaction will undoubtedly provide new insights into developing advanced hydrogels for wound repair and regeneration. This paper reviews the latest studies on classical signaling pathways and potential targets influenced by hydrogel scaffolds in chronic wound healing. This work hopes that it will offer a different perspective in developing more efficient hydrogels for treating chronic wounds.
Collapse
Affiliation(s)
- Fang Chen
- Hebei Provincial Key Laboratory of Skeletal Metabolic Physiology of Chronic Kidney Disease, Affiliated Hospital of Hebei University, Baoding, 071000, China
- First Department of Bone Injury, Luzhou Municipal Hospital of Traditional Chinese Medicine, Luzhou, Sichuan, 646000, China
| | - Pingli Wu
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China
| | - Haisong Zhang
- Hebei Provincial Key Laboratory of Skeletal Metabolic Physiology of Chronic Kidney Disease, Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Guoming Sun
- Sunogel Biotechnologies Inc., Lutherville Timonium, 9 W Ridgely Road Ste 270, Maryland, 21093, USA
| |
Collapse
|
4
|
Zhang Y, Xue X, Meng L, Li D, Qiao W, Wang J, Xie D. Roles of autophagy-related genes in the therapeutic effects of Xuanfei Pingchuan capsules on chronic obstructive pulmonary disease based on transcriptome sequencing analysis. Front Pharmacol 2023; 14:1123882. [PMID: 37274101 PMCID: PMC10232735 DOI: 10.3389/fphar.2023.1123882] [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: 01/03/2023] [Accepted: 04/24/2023] [Indexed: 06/06/2023] Open
Abstract
Objective: Autophagy plays an important role in the occurrence and development of chronic obstructive pulmonary disease (COPD). We evaluated the effect of Xuanfei Pingchuan capsule (XFPC) on autophagy-related genes of COPD by a bioinformatics analysis and experimental verification. Methods: The best treatment duration was screened by CCK8 assays. HBE cells were divided into three groups: blank, CSE and XFPC. After intervened by XFPC, HBE cells were collected and sent to Shenzhen Huada Gene Company for transcriptome sequencing. Subsequently, differential expression analyses, target gene prediction, and function enrichment analyses were carried out. Expression changes were verified in HBE cells by real-time Quantitative PCR (RT-qPCR) and western blotting (WB). Results: The result of differential expression analysis displayed that 125 target genes of HBE cells were mainly related to mitogen-activated protein kinase (MKK) binding, interleukin 33 binding, 1-Pyrroline-5-carboxylate dehydrogenase activity, and the mitogen-activated protein kinase (MAPK) signal pathway. Among the target genes, the core genes related to autophagy obtained by maximum neighborhood component algorithm were CSF1, AREG, MAPK9, MAP3K7, and AKT3. RT-qPCR and WB methods were used to verify the result, it showed similar expression changes in CSF1, MAPK9, MAP3K7, and AKT3 in bronchial epithelial cells to those in the bioinformatics analysis. Conclusion: Through transcriptome sequencing and validation analysis, we predicted that CSF1, MAPK9, MAP3K7, and AKT3 may be the potential autophagy-related genes that play an important role in the pathogenesis of COPD. XFPC may regulate autophagy by down-regulating the expression of CSF1, MAPK9, MAP3K7, and AKT3, thus achieving the purpose of treating chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
| | - Xiaoming Xue
- Graduate School, Shanxi University of Chinese Medicine, Taiyuan, China
| | | | | | | | | | | |
Collapse
|
5
|
Tusa I, Menconi A, Tubita A, Rovida E. Pathophysiological Impact of the MEK5/ERK5 Pathway in Oxidative Stress. Cells 2023; 12:cells12081154. [PMID: 37190064 DOI: 10.3390/cells12081154] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/22/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Oxidative stress regulates many physiological and pathological processes. Indeed, a low increase in the basal level of reactive oxygen species (ROS) is essential for various cellular functions, including signal transduction, gene expression, cell survival or death, as well as antioxidant capacity. However, if the amount of generated ROS overcomes the antioxidant capacity, excessive ROS results in cellular dysfunctions as a consequence of damage to cellular components, including DNA, lipids and proteins, and may eventually lead to cell death or carcinogenesis. Both in vitro and in vivo investigations have shown that activation of the mitogen-activated protein kinase kinase 5/extracellular signal-regulated kinase 5 (MEK5/ERK5) pathway is frequently involved in oxidative stress-elicited effects. In particular, accumulating evidence identified a prominent role of this pathway in the anti-oxidative response. In this respect, activation of krüppel-like factor 2/4 and nuclear factor erythroid 2-related factor 2 emerged among the most frequent events in ERK5-mediated response to oxidative stress. This review summarizes what is known about the role of the MEK5/ERK5 pathway in the response to oxidative stress in pathophysiological contexts within the cardiovascular, respiratory, lymphohematopoietic, urinary and central nervous systems. The possible beneficial or detrimental effects exerted by the MEK5/ERK5 pathway in the above systems are also discussed.
Collapse
Affiliation(s)
- Ignazia Tusa
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Alessio Menconi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Alessandro Tubita
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| |
Collapse
|
6
|
Miller D, Harnor SJ, Martin MP, Noble RA, Wedge SR, Cano C. Modulation of ERK5 Activity as a Therapeutic Anti-Cancer Strategy. J Med Chem 2023; 66:4491-4502. [PMID: 37002872 PMCID: PMC10108346 DOI: 10.1021/acs.jmedchem.3c00072] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Indexed: 04/03/2023]
Abstract
The extracellular signal-regulated kinase 5 (ERK5) signaling pathway is one of four conventional mitogen-activated protein (MAP) kinase pathways. Genetic perturbation of ERK5 has suggested that modulation of ERK5 activity may have therapeutic potential in cancer chemotherapy. This Miniperspective examines the evidence for ERK5 as a drug target in cancer, the structure of ERK5, and the evolution of structurally distinct chemotypes of ERK5 kinase domain inhibitors. The emerging complexities of ERK5 pharmacology are discussed, including the confounding phenomenon of paradoxical ERK5 activation by small-molecule ERK5 inhibitors. The impact of the recent development and biological evaluation of potent and selective bifunctional degraders of ERK5 and future opportunities in ERK modulation are also explored.
Collapse
Affiliation(s)
- Duncan
C. Miller
- Cancer
Research Horizons Therapeutic Innovation, Newcastle Drug Discovery
Group, Newcastle University Centre for Cancer, School of Natural and
Environmental Sciences, Newcastle University, Bedson Building, Newcastle upon Tyne NE1 7RU, United Kingdom
| | - Suzannah J. Harnor
- Cancer
Research Horizons Therapeutic Innovation, Newcastle Drug Discovery
Group, Newcastle University Centre for Cancer, School of Natural and
Environmental Sciences, Newcastle University, Bedson Building, Newcastle upon Tyne NE1 7RU, United Kingdom
| | - Mathew P. Martin
- Cancer
Research Horizons Therapeutic Innovation, Newcastle Drug Discovery
Group, Translational and Clinical Research
Institute, Paul O’Gorman Building, Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Richard A. Noble
- Cancer
Research Horizons Therapeutic Innovation, Newcastle Drug Discovery
Group, Translational and Clinical Research
Institute, Paul O’Gorman Building, Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Stephen R. Wedge
- Cancer
Research Horizons Therapeutic Innovation, Newcastle Drug Discovery
Group, Translational and Clinical Research
Institute, Paul O’Gorman Building, Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Celine Cano
- Cancer
Research Horizons Therapeutic Innovation, Newcastle Drug Discovery
Group, Newcastle University Centre for Cancer, School of Natural and
Environmental Sciences, Newcastle University, Bedson Building, Newcastle upon Tyne NE1 7RU, United Kingdom
| |
Collapse
|
7
|
Filiberti S, Russo M, Lonardi S, Bugatti M, Vermi W, Tournier C, Giurisato E. Self-Renewal of Macrophages: Tumor-Released Factors and Signaling Pathways. Biomedicines 2022; 10:2709. [PMID: 36359228 PMCID: PMC9687165 DOI: 10.3390/biomedicines10112709] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/14/2022] [Accepted: 10/21/2022] [Indexed: 04/11/2024] Open
Abstract
Macrophages are the most abundant immune cells of the tumor microenvironment (TME) and have multiple important functions in cancer. During tumor growth, both tissue-resident macrophages and newly recruited monocyte-derived macrophages can give rise to tumor-associated macrophages (TAMs), which have been associated with poor prognosis in most cancers. Compelling evidence indicate that the high degree of plasticity of macrophages and their ability to self-renew majorly impact tumor progression and resistance to therapy. In addition, the microenvironmental factors largely affect the metabolism of macrophages and may have a major influence on TAMs proliferation and subsets functions. Thus, understanding the signaling pathways regulating TAMs self-renewal capacity may help to identify promising targets for the development of novel anticancer agents. In this review, we focus on the environmental factors that promote the capacity of macrophages to self-renew and the molecular mechanisms that govern TAMs proliferation. We also highlight the impact of tumor-derived factors on macrophages metabolism and how distinct metabolic pathways affect macrophage self-renewal.
Collapse
Affiliation(s)
- Serena Filiberti
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Mariapia Russo
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Silvia Lonardi
- Department of Molecular and Translational Medicine, University of Brescia, 25100 Brescia, Italy
| | - Mattia Bugatti
- Department of Molecular and Translational Medicine, University of Brescia, 25100 Brescia, Italy
| | - William Vermi
- Department of Molecular and Translational Medicine, University of Brescia, 25100 Brescia, Italy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Cathy Tournier
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
| | - Emanuele Giurisato
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
| |
Collapse
|
8
|
Tubita A, Lombardi Z, Tusa I, Lazzeretti A, Sgrignani G, Papini D, Menconi A, Gagliardi S, Lulli M, Dello Sbarba P, Esparís-Ogando A, Pandiella A, Stecca B, Rovida E. Inhibition of ERK5 Elicits Cellular Senescence in Melanoma via the Cyclin-Dependent Kinase Inhibitor p21. Cancer Res 2022; 82:447-457. [PMID: 34799355 PMCID: PMC9397638 DOI: 10.1158/0008-5472.can-21-0993] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 10/06/2021] [Accepted: 11/15/2021] [Indexed: 01/07/2023]
Abstract
Melanoma is the deadliest skin cancer with a very poor prognosis in advanced stages. Although targeted and immune therapies have improved survival, not all patients benefit from these treatments. The mitogen-activated protein kinase ERK5 supports the growth of melanoma cells in vitro and in vivo. However, ERK5 inhibition results in cell-cycle arrest rather than appreciable apoptosis. To clarify the role of ERK5 in melanoma growth, we performed transcriptomic analyses following ERK5 knockdown in melanoma cells expressing BRAFV600E and found that cellular senescence was among the most affected processes. In melanoma cells expressing either wild-type or mutant (V600E) BRAF, both genetic and pharmacologic inhibition of ERK5 elicited cellular senescence, as observed by a marked increase in senescence-associated β-galactosidase activity and p21 expression. In addition, depletion of ERK5 from melanoma cells resulted in increased levels of CXCL1, CXCL8, and CCL20, proteins typically involved in the senescence-associated secretory phenotype. Knockdown of p21 suppressed the induction of cellular senescence by ERK5 blockade, pointing to p21 as a key mediator of this process. In vivo, ERK5 knockdown or inhibition with XMD8-92 in melanoma xenografts promoted cellular senescence. Based on these results, small-molecule compounds targeting ERK5 constitute a rational series of prosenescence drugs that may be exploited for melanoma treatment. SIGNIFICANCE: This study shows that targeting ERK5 induces p21-mediated cellular senescence in melanoma, identifying a prosenescence effect of ERK5 inhibitors that may be exploited for melanoma treatment.
Collapse
Affiliation(s)
- Alessandro Tubita
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Zoe Lombardi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Ignazia Tusa
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Azzurra Lazzeretti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Giovanna Sgrignani
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Dimitri Papini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Alessio Menconi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Sinforosa Gagliardi
- Core Research Laboratory - Institute for Cancer Research and Prevention (ISPRO), Florence, Italy
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Persio Dello Sbarba
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Azucena Esparís-Ogando
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Instituto de Investigación Biomédica de Salamanca (IBSAL), CIBERONC, Salamanca, Spain
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Instituto de Investigación Biomédica de Salamanca (IBSAL), CIBERONC, Salamanca, Spain
- CSIC, Salamanca, Spain
| | - Barbara Stecca
- Core Research Laboratory - Institute for Cancer Research and Prevention (ISPRO), Florence, Italy
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy.
| |
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
|
Tusa I, Gagliardi S, Tubita A, Pandolfi S, Menconi A, Lulli M, Dello Sbarba P, Stecca B, Rovida E. The Hedgehog-GLI Pathway Regulates MEK5-ERK5 Expression and Activation in Melanoma Cells. Int J Mol Sci 2021; 22:11259. [PMID: 34681917 PMCID: PMC8538987 DOI: 10.3390/ijms222011259] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 12/28/2022] Open
Abstract
Malignant melanoma is the deadliest skin cancer, with a poor prognosis in advanced stages. We recently showed that the extracellular signal-regulated kinase 5 (ERK5), encoded by the MAPK7 gene, plays a pivotal role in melanoma by regulating cell functions necessary for tumour development, such as proliferation. Hedgehog-GLI signalling is constitutively active in melanoma and is required for proliferation. However, no data are available in literature about a possible interplay between Hedgehog-GLI and ERK5 pathways. Here, we show that hyperactivation of the Hedgehog-GLI pathway by genetic inhibition of the negative regulator Patched 1 increases the amount of ERK5 mRNA and protein. Chromatin immunoprecipitation showed that GLI1, the major downstream effector of Hedgehog-GLI signalling, binds to a functional non-canonical GLI consensus sequence at the MAPK7 promoter. Furthermore, we found that ERK5 is required for Hedgehog-GLI-dependent melanoma cell proliferation, and that the combination of GLI and ERK5 inhibitors is more effective than single treatments in reducing cell viability and colony formation ability in melanoma cells. Together, these findings led to the identification of a novel Hedgehog-GLI-ERK5 axis that regulates melanoma cell growth, and shed light on new functions of ERK5, paving the way for new therapeutic options in melanoma and other neoplasms with active Hedgehog-GLI and ERK5 pathways.
Collapse
Affiliation(s)
- Ignazia Tusa
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy; (I.T.); (A.T.); (A.M.); (M.L.); (P.D.S.)
| | - Sinforosa Gagliardi
- Core Research Laboratory-Institute for Cancer Research and Prevention (ISPRO), 50134 Florence, Italy; (S.G.); (S.P.)
| | - Alessandro Tubita
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy; (I.T.); (A.T.); (A.M.); (M.L.); (P.D.S.)
| | - Silvia Pandolfi
- Core Research Laboratory-Institute for Cancer Research and Prevention (ISPRO), 50134 Florence, Italy; (S.G.); (S.P.)
| | - Alessio Menconi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy; (I.T.); (A.T.); (A.M.); (M.L.); (P.D.S.)
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy; (I.T.); (A.T.); (A.M.); (M.L.); (P.D.S.)
| | - Persio Dello Sbarba
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy; (I.T.); (A.T.); (A.M.); (M.L.); (P.D.S.)
| | - Barbara Stecca
- Core Research Laboratory-Institute for Cancer Research and Prevention (ISPRO), 50134 Florence, Italy; (S.G.); (S.P.)
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy; (I.T.); (A.T.); (A.M.); (M.L.); (P.D.S.)
| |
Collapse
|
11
|
Gentilini A, Lori G, Caligiuri A, Raggi C, Di Maira G, Pastore M, Piombanti B, Lottini T, Arcangeli A, Madiai S, Navari N, Banales JM, Di Matteo S, Alvaro D, Duwe L, Andersen JB, Tubita A, Tusa I, Di Tommaso L, Campani C, Rovida E, Marra F. Extracellular Signal-Regulated Kinase 5 Regulates the Malignant Phenotype of Cholangiocarcinoma Cells. Hepatology 2021; 74:2007-2020. [PMID: 33959996 PMCID: PMC8518067 DOI: 10.1002/hep.31888] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 04/21/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Cholangiocarcinoma (CCA) is characterized by high resistance to chemotherapy and poor prognosis. Several oncogenic pathways converge on activation of extracellular signal-regulated kinase 5 (ERK5), whose role in CCA has not been explored. The aim of this study was to investigate the role of ERK5 in the biology of CCA. APPROACH AND RESULTS ERK5 expression was detected in two established (HuCCT-1 and CCLP-1) and two primary human intrahepatic CCA cell lines (iCCA58 and iCCA60). ERK5 phosphorylation was increased in CCA cells exposed to soluble mediators. In both HuCCT-1 and CCLP-1 cells, ERK5 was localized in the nucleus, and exposure to fetal bovine serum (FBS) further increased the amount of nuclear ERK5. In human CCA specimens, ERK5 mRNA expression was increased in tumor cells and positively correlated with portal invasion. ERK5 protein levels were significantly associated with tumor grade. Growth, migration, and invasion of CCA cells were decreased when ERK5 was silenced using specific short hairpin RNA (shRNA). The inhibitory effects on CCA cell proliferation, migration and invasion were recapitulated by treatment with small molecule inhibitors targeting ERK5. In addition, expression of the angiogenic factors VEGF and angiopoietin 1 was reduced after ERK5 silencing. Conditioned medium from ERK5-silenced cells had a lower ability to induce tube formation by human umbilical vein endothelial cells and to induce migration of myofibroblasts and monocytes/macrophages. In mice, subcutaneous injection of CCLP-1 cells silenced for ERK5 resulted in less frequent tumor development and smaller size of xenografts compared with cells transfected with nontargeting shRNA. CONCLUSIONS ERK5 is a key mediator of growth and migration of CCA cells and supports a protumorigenic crosstalk between the tumor and the microenvironment.
Collapse
Affiliation(s)
- Alessandra Gentilini
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| | - Giulia Lori
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| | - Alessandra Caligiuri
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| | - Chiara Raggi
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| | - Giovanni Di Maira
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| | - Mirella Pastore
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| | - Benedetta Piombanti
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| | - Tiziano Lottini
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| | - Annarosa Arcangeli
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| | - Stefania Madiai
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| | - Nadia Navari
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| | - Jesus M. Banales
- Department of Liver and Gastrointestinal DiseasesBiodonostia Health Research InstituteCIBERehdIkerbasqueSan SebastianSpain
| | - Sabina Di Matteo
- Department of ImmunologyBambino Gesù Children’s HospitalIRCCSRomeItaly
| | - Domenico Alvaro
- Department of Internal Medicine and Medical SpecialtiesSapienza University of RomeRomeItaly
| | - Lea Duwe
- Biotech Research and Innovation Centre (BRIC)Dept. of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Jesper B. Andersen
- Biotech Research and Innovation Centre (BRIC)Dept. of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Alessandro Tubita
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”University of FlorenceItaly
| | - Ignazia Tusa
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”University of FlorenceItaly
| | - Luca Di Tommaso
- Pathology UnitHumanitas Clinical and Research Center IRCCSRozzanoItaly
| | - Claudia Campani
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”University of FlorenceItaly
| | - Fabio Marra
- Department of Experimental and Clinical MedicineUniversity of FlorenceFlorenceItaly
| |
Collapse
|
12
|
Wang D, Deng L, Xu X, Ji Y, Jiao Z. Elevated SYNC Expression Is Associated with Gastric Tumorigenesis and Infiltration of M2-Polarized Macrophages in the Gastric Tumor Immune Microenvironment. Genet Test Mol Biomarkers 2021; 25:236-246. [PMID: 33734892 DOI: 10.1089/gtmb.2020.0131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aims: To assess the expression and epigenetic regulation of Syncoilin, intermediate filament protein (SYNC) in gastric cancer tissues, and to determine its associations with clinicopathological features; immune infiltration of macrophages in tumors; and patient survival. Materials and Methods: Clinicopathological features, expression profiles, and methylation data of the SYNC gene were obtained from multi-institutional real-world public datasets. A total of 1601 samples from patients with gastric cancer were examined. The associations between clinicopathological features and SYNC expression levels were assessed by the chi-square test; survival was assessed using the Kaplan-Meier analysis. The infiltration levels of M1, 2-polarized tumor-associated macrophages (TAMs) in a gastric tumor immune microenvironment were quantified using deconvolution, and the correlation between SYNC expression level and M1, 2-polarized macrophages' infiltration was examined using the Pearson correlation test. SYNC gene methylation data were analyzed to investigate epigenetic control of its expression. Results: SYNC expression was elevated in gastric cancer tissues (p < 0.01), and was associated with a poorer overall survival (p < 0.01) and poorer postprogression survival (p = 0.01). Higher SYNC levels were significantly associated with more aggressive clinicopathological features in gastric cancer patients (p < 0.05). SYNC was also associated with the infiltration of M2-polarized TAMs in the gastric tumor immune microenvironment (p < 0.001). Hypomethylation was shown to be associated with SYNC's upregulation (p < 0.05). Conclusion: SYNC is highly expressed in gastric cancer tissues and has the potential to be a therapeutic target and to serve as a prognostic marker.
Collapse
Affiliation(s)
- Dazhi Wang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pharmacy, Center for Precision Cancer Medicine, Clinical Oncology Pharmacist Training Bases (National Health Commission), Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lihua Deng
- Department of Oncology, Center for Precision Cancer Medicine, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xiaona Xu
- Department of Central Laboratories, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yinghui Ji
- Department of Pharmacy, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Zheng Jiao
- Department of Pharmacy, Center for Precision Cancer Medicine, Clinical Oncology Pharmacist Training Bases (National Health Commission), Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
13
|
Sreedurgalakshmi K, Srikar R, Rajkumari R. CRISPR-Cas deployment in non-small cell lung cancer for target screening, validations, and discoveries. Cancer Gene Ther 2020; 28:566-580. [PMID: 33191402 DOI: 10.1038/s41417-020-00256-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/14/2020] [Accepted: 10/29/2020] [Indexed: 12/24/2022]
Abstract
Continued advancements in CRISPR-Cas systems have accelerated genome research. Use of CRISPR-Cas in cancer research has been of great interest that is resulting in development of orthogonal methods for drug target validations and discovery of new therapeutic targets through genome-wide screens of cancer cells. CRISPR-based screens have also revealed several new cancer drivers through alterations in tumor suppressor genes (TSGs) and oncogenes inducing resistance to targeted therapies via activation of alternate signaling pathways. Given such dynamic status of cancer, we review the application of CRISPR-Cas in non-small cell lung cancer (NSCLC) for development of mutant models, drug screening, target validation, novel target discoveries, and other emerging potential applications. In addition, CRISPR-based approach for development of novel anticancer combination therapies is also discussed in this review.
Collapse
Affiliation(s)
- K Sreedurgalakshmi
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamilnadu, India.,Division of Biosimilars and Gene Therapy, R&D, Levim Biotech LLP, Chennai, Tamilnadu, India
| | - R Srikar
- Division of Biosimilars and Gene Therapy, R&D, Levim Biotech LLP, Chennai, Tamilnadu, India.
| | - Reena Rajkumari
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamilnadu, India.
| |
Collapse
|
14
|
Luiz JPM, Toller‐Kawahisa JE, Viacava PR, Nascimento DC, Pereira PT, Saraiva AL, Prado DS, Le Bert M, Giurisato E, Tournier C, Cunha TM, Cunha FQ, Quesniaux V, Ryffel B, Alves‐Filho JC. MEK5/ERK5 signaling mediates IL‐4‐induced M2 macrophage differentiation through regulation of c‐Myc expression. J Leukoc Biol 2020; 108:1215-1223. [DOI: 10.1002/jlb.1ma0520-016r] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/27/2020] [Accepted: 07/16/2020] [Indexed: 01/10/2023] Open
Affiliation(s)
- João Paulo M. Luiz
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| | - Juliana E. Toller‐Kawahisa
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| | - Paula R. Viacava
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| | - Daniele C. Nascimento
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| | - Priscilla T. Pereira
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| | - André L. Saraiva
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| | - Douglas S. Prado
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| | - Marc Le Bert
- CNRS, UMR7355 Orléans, France
- Experimental and Molecular Immunology and Neurogenetics University of Orléans Orléans France
| | - Emanuele Giurisato
- Division of Cancer Sciences School of Medical Sciences Faculty of Biology, Medicine and Health University of Manchester Manchester United Kingdom
| | - Cathy Tournier
- Division of Cancer Sciences School of Medical Sciences Faculty of Biology, Medicine and Health University of Manchester Manchester United Kingdom
| | - Thiago M. Cunha
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| | - Fernando Q. Cunha
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| | - Valerie Quesniaux
- CNRS, UMR7355 Orléans, France
- Experimental and Molecular Immunology and Neurogenetics University of Orléans Orléans France
| | - Bernhard Ryffel
- CNRS, UMR7355 Orléans, France
- Experimental and Molecular Immunology and Neurogenetics University of Orléans Orléans France
| | - José C. Alves‐Filho
- Center for Research in Inflammatory Diseases and Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto Brazil
| |
Collapse
|
15
|
Giurisato E, Lonardi S, Telfer B, Lussoso S, Risa-Ebrí B, Zhang J, Russo I, Wang J, Santucci A, Finegan KG, Gray NS, Vermi W, Tournier C. Extracellular-Regulated Protein Kinase 5-Mediated Control of p21 Expression Promotes Macrophage Proliferation Associated with Tumor Growth and Metastasis. Cancer Res 2020; 80:3319-3330. [PMID: 32561530 DOI: 10.1158/0008-5472.can-19-2416] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 04/07/2020] [Accepted: 06/12/2020] [Indexed: 12/14/2022]
Abstract
The presence of immunosuppressive macrophages that become activated in the tumor microenvironment constitutes a major factor responsible for tumor growth and malignancy. In line with this knowledge, we report here that macrophage proliferation is a significant feature of advanced stages of cancer. Moreover, we have found that a high proportion of proliferating macrophages in human tumors express ERK5. ERK5 was required for supporting the proliferation of macrophages in tumor grafts in mice. Furthermore, myeloid ERK5 deficiency negatively impacted the proliferation of both resident and infiltrated macrophages in metastatic lung nodules. ERK5 maintained the capacity of macrophages to proliferate by suppressing p21 expression to halt their differentiation program. Collectively, these data provide insight into the mechanism underpinning macrophage proliferation to support malignant tumor development, thereby strengthening the value of ERK5-targeted therapies to restore antitumor immunity through the blockade of protumorigenic macrophage activation. SIGNIFICANCE: These findings offer a new rationale for anti-ERK5 therapy to improve cancer patient outcomes by blocking the proliferative activity of tumor macrophages.
Collapse
Affiliation(s)
- Emanuele Giurisato
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Siena, Italy. .,Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Silvia Lonardi
- Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| | - Brian Telfer
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Sarah Lussoso
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Blanca Risa-Ebrí
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Jingwei Zhang
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Ilaria Russo
- School of Medicine, Keel University, Keel, United Kingdom.,Department of Medicine-Infectious Diseases, Washington University, Saint Louis, Missouri
| | - Jinhua Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - Annalisa Santucci
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Katherine G Finegan
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - William Vermi
- Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy.,Department of Pathology and Immunology, Washington University, Saint Louis, Missouri
| | - Cathy Tournier
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
16
|
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
|
17
|
Beyond Kinase Activity: ERK5 Nucleo-Cytoplasmic Shuttling as a Novel Target for Anticancer Therapy. Int J Mol Sci 2020; 21:ijms21030938. [PMID: 32023850 PMCID: PMC7038028 DOI: 10.3390/ijms21030938] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 01/18/2023] Open
Abstract
The importance of mitogen-activated protein kinases (MAPK) in human pathology is underlined by the relevance of abnormalities of MAPK-related signaling pathways to a number of different diseases, including inflammatory disorders and cancer. One of the key events in MAPK signaling, especially with respect to pro-proliferative effects that are crucial for the onset and progression of cancer, is MAPK nuclear translocation and its role in the regulation of gene expression. The extracellular signal-regulated kinase 5 (ERK5) is the most recently discovered classical MAPK and it is emerging as a possible target for cancer treatment. The bigger size of ERK5 when compared to other MAPK enables multiple levels of regulation of its expression and activity. In particular, the phosphorylation of kinase domain and C-terminus, as well as post-translational modifications and chaperone binding, are involved in ERK5 regulation. Likewise, different mechanisms control ERK5 nucleo-cytoplasmic shuttling, underscoring the key role of ERK5 in the nuclear compartment. In this review, we will focus on the mechanisms involved in ERK5 trafficking between cytoplasm and nucleus, and discuss how these processes might be exploited to design new strategies for cancer treatment.
Collapse
|
18
|
Impact of ERK5 on the Hallmarks of Cancer. Int J Mol Sci 2019; 20:ijms20061426. [PMID: 30901834 PMCID: PMC6471124 DOI: 10.3390/ijms20061426] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/15/2019] [Accepted: 03/18/2019] [Indexed: 12/15/2022] Open
Abstract
Extracellular signal-regulated kinase 5 (ERK5) belongs to the mitogen-activated protein kinase (MAPK) family that consists of highly conserved enzymes expressed in all eukaryotic cells and elicits several biological responses, including cell survival, proliferation, migration, and differentiation. In recent years, accumulating lines of evidence point to a relevant role of ERK5 in the onset and progression of several types of cancer. In particular, it has been reported that ERK5 is a key signaling molecule involved in almost all the biological features of cancer cells so that its targeting is emerging as a promising strategy to suppress tumor growth and spreading. Based on that, in this review, we pinpoint the hallmark-specific role of ERK5 in cancer in order to identify biological features that will potentially benefit from ERK5 targeting.
Collapse
|
19
|
Tusa I, Cheloni G, Poteti M, Gozzini A, DeSouza NH, Shan Y, Deng X, Gray NS, Li S, Rovida E, Dello Sbarba P. Targeting the Extracellular Signal-Regulated Kinase 5 Pathway to Suppress Human Chronic Myeloid Leukemia Stem Cells. Stem Cell Reports 2018; 11:929-943. [PMID: 30245209 PMCID: PMC6178886 DOI: 10.1016/j.stemcr.2018.08.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 08/22/2018] [Accepted: 08/22/2018] [Indexed: 12/20/2022] Open
Abstract
Tyrosine kinase inhibitors (TKi) are effective against chronic myeloid leukemia (CML), but their inefficacy on leukemia stem cells (LSCs) may lead to relapse. To identify new druggable targets alternative to BCR/ABL, we investigated the role of the MEK5/ERK5 pathway in LSC maintenance in low oxygen, a feature of bone marrow stem cell niches. We found that MEK5/ERK5 pathway inhibition reduced the growth of CML patient-derived cells and cell lines in vitro and the number of leukemic cells in vivo. Treatment in vitro of primary CML cells with MEK5/ERK5 inhibitors, but not TKi, strikingly reduced culture repopulation ability (CRA), serial colony formation ability, long-term culture-initiating cells (LTC-ICs), and CD26-expressing cells. Importantly, MEK5/ERK5 inhibition was effective on CML cells regardless of the presence or absence of imatinib, and did not reduce CRA or LTC-ICs of normal CD34+ cells. Thus, targeting MEK/ERK5 may represent an innovative therapeutic approach to suppress CML progenitor/stem cells. ERK5 is constitutively active in chronic myeloid leukemia (CML) cells ERK5 pathway inhibition reduces the growth of CML cells in vitro and in vivo ERK5 pathway inhibition strikingly reduces CML progenitor/stem cell maintenance The combination of ERK5i with imatinib reduces the expression of stem cell proteins
Collapse
Affiliation(s)
- Ignazia Tusa
- Department of Experimental and Clinical Biomedical Sciences, Università degli Studi di Firenze, viale G.B. Morgagni, 50, Firenze 50134, Italy; Istituto Toscano Tumori (ITT), Firenze 50134, Italy
| | - Giulia Cheloni
- Department of Experimental and Clinical Biomedical Sciences, Università degli Studi di Firenze, viale G.B. Morgagni, 50, Firenze 50134, Italy; Istituto Toscano Tumori (ITT), Firenze 50134, Italy
| | - Martina Poteti
- Department of Experimental and Clinical Biomedical Sciences, Università degli Studi di Firenze, viale G.B. Morgagni, 50, Firenze 50134, Italy
| | - Antonella Gozzini
- Hematology Unit, Careggi University Hospital (AOUC), Firenze 50134, Italy
| | - Ngoc Ho DeSouza
- Department of Medicine, University of Massachusetts, Worcester, MA 01605, USA
| | - Yi Shan
- Department of Medicine, University of Massachusetts, Worcester, MA 01605, USA
| | - Xianming Deng
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Nathanael S Gray
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Shaoguang Li
- Department of Medicine, University of Massachusetts, Worcester, MA 01605, USA
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Sciences, Università degli Studi di Firenze, viale G.B. Morgagni, 50, Firenze 50134, Italy; Istituto Toscano Tumori (ITT), Firenze 50134, Italy.
| | - Persio Dello Sbarba
- Department of Experimental and Clinical Biomedical Sciences, Università degli Studi di Firenze, viale G.B. Morgagni, 50, Firenze 50134, Italy; Istituto Toscano Tumori (ITT), Firenze 50134, Italy.
| |
Collapse
|
20
|
Tusa I, Gagliardi S, Tubita A, Pandolfi S, Urso C, Borgognoni L, Wang J, Deng X, Gray NS, Stecca B, Rovida E. ERK5 is activated by oncogenic BRAF and promotes melanoma growth. Oncogene 2018; 37:2601-2614. [PMID: 29483645 PMCID: PMC5945581 DOI: 10.1038/s41388-018-0164-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/21/2017] [Accepted: 01/17/2018] [Indexed: 02/07/2023]
Abstract
Malignant melanoma is among the most aggressive cancers and its incidence is increasing worldwide. Targeted therapies and immunotherapy have improved the survival of patients with metastatic melanoma in the last few years; however, available treatments are still unsatisfactory. While the role of the BRAF-MEK1/2-ERK1/2 pathway in melanoma is well established, the involvement of mitogen-activated protein kinases MEK5-ERK5 remains poorly explored. Here we investigated the function of ERK5 signaling in melanoma. We show that ERK5 is consistently expressed in human melanoma tissues and is active in melanoma cells. Genetic silencing and pharmacological inhibition of ERK5 pathway drastically reduce the growth of melanoma cells and xenografts harboring wild-type (wt) or mutated BRAF (V600E). We also found that oncogenic BRAF positively regulates expression, phosphorylation, and nuclear localization of ERK5. Importantly, ERK5 kinase and transcriptional transactivator activities are enhanced by BRAF. Nevertheless, combined pharmacological inhibition of BRAFV600E and MEK5 is required to decrease nuclear ERK5, that is critical for the regulation of cell proliferation. Accordingly, combination of MEK5 or ERK5 inhibitors with BRAFV600E inhibitor vemurafenib is more effective than single treatments in reducing colony formation and growth of BRAFV600E melanoma cells and xenografts. Overall, these data support a key role of the ERK5 pathway for melanoma growth in vitro and in vivo and suggest that targeting ERK5, alone or in combination with BRAF-MEK1/2 inhibitors, might represent a novel approach for melanoma treatment.
Collapse
Affiliation(s)
- Ignazia Tusa
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
| | | | - Alessandro Tubita
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
| | - Silvia Pandolfi
- Core Research Laboratory - Istituto Toscano Tumori, Florence, Italy
- VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
| | - Carmelo Urso
- Anatomic Pathology Unit, Dermatopathology Section, S.M. Annunziata Hospital, Florence, Italy
| | - Lorenzo Borgognoni
- Plastic Surgery Unit, Regional Melanoma Referral Center, S.M. Annunziata Hospital, Florence, Italy
| | - Jinhua Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Xianming Deng
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Barbara Stecca
- Core Research Laboratory - Istituto Toscano Tumori, Florence, Italy.
| | - Elisabetta Rovida
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy.
| |
Collapse
|
21
|
Myeloid ERK5 deficiency suppresses tumor growth by blocking protumor macrophage polarization via STAT3 inhibition. Proc Natl Acad Sci U S A 2018; 115:E2801-E2810. [PMID: 29507229 PMCID: PMC5866536 DOI: 10.1073/pnas.1707929115] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Macrophages can be functionally reprogrammed by the tumor microenvironment to further tumor growth and malignancy. In this study, we have discovered that this pathological process is dependent on the ERK5 MAPK. Accordingly, we demonstrated that inactivation of ERK5 in macrophages blocked the phosphorylation of STAT3, a transcription factor crucial for determining macrophage polarity, and impaired the growth of melanoma and carcinoma grafts. These results raise the possibility that targeting protumor macrophages via anti-ERK5 therapy constitutes a very attractive strategy for cancer treatment. This is important given that the detection of large numbers of macrophages in human tumors often correlates with poor prognosis, but also with a poor response of the tumor to anticancer agents. Owing to the prevalence of tumor-associated macrophages (TAMs) in cancer and their unique influence upon disease progression and malignancy, macrophage-targeted interventions have attracted notable attention in cancer immunotherapy. However, tractable targets to reduce TAM activities remain very few and far between because the signaling mechanisms underpinning protumor macrophage phenotypes are largely unknown. Here, we have investigated the role of the extracellular-regulated protein kinase 5 (ERK5) as a determinant of macrophage polarity. We report that the growth of carcinoma grafts was halted in myeloid ERK5-deficient mice. Coincidentally, targeting ERK5 in macrophages induced a transcriptional switch in favor of proinflammatory mediators. Further molecular analyses demonstrated that activation of the signal transducer and activator of transcription 3 (STAT3) via Tyr705 phosphorylation was impaired in erk5-deleted TAMs. Our study thus suggests that blocking ERK5 constitutes a treatment strategy to reprogram macrophages toward an antitumor state by inhibiting STAT3-induced gene expression.
Collapse
|
22
|
Loveridge CJ, van 't Hof RJ, Charlesworth G, King A, Tan EH, Rose L, Daroszewska A, Prior A, Ahmad I, Welsh M, Mui EJ, Ford C, Salji M, Sansom O, Blyth K, Leung HY. Analysis of Nkx3.1:Cre-driven Erk5 deletion reveals a profound spinal deformity which is linked to increased osteoclast activity. Sci Rep 2017; 7:13241. [PMID: 29038439 PMCID: PMC5643304 DOI: 10.1038/s41598-017-13346-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 09/21/2017] [Indexed: 12/15/2022] Open
Abstract
Extracellular signal-regulated protein kinase 5 (ERK5) has been implicated during development and carcinogenesis. Nkx3.1-mediated Cre expression is a useful strategy to genetically manipulate the mouse prostate. While grossly normal at birth, we observed an unexpected phenotype of spinal protrusion in Nkx3.1:Cre;Erk5 fl/fl (Erk5 fl/fl) mice by ~6-8 weeks of age. X-ray, histological and micro CT (µCT) analyses showed that 100% of male and female Erk5 fl/fl mice had a severely deformed curved thoracic spine, with an associated loss of trabecular bone volume. Although sex-specific differences were observed, histomorphometry measurements revealed that both bone resorption and bone formation parameters were increased in male Erk5 fl/fl mice compared to wild type (WT) littermates. Osteopenia occurs where the rate of bone resorption exceeds that of bone formation, so we investigated the role of the osteoclast compartment. We found that treatment of RANKL-stimulated primary bone marrow-derived macrophage (BMDM) cultures with small molecule ERK5 pathway inhibitors increased osteoclast numbers. Furthermore, osteoclast numbers and expression of osteoclast marker genes were increased in parallel with reduced Erk5 expression in cultures generated from Erk5 fl/fl mice compared to WT mice. Collectively, these results reveal a novel role for Erk5 during bone maturation and homeostasis in vivo.
Collapse
Affiliation(s)
- Carolyn J Loveridge
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden, Glasgow, G61 1BD, UK
- Beatson Institute for Cancer Research, Bearsden, Glasgow, G61 1BD, UK
| | - Rob J van 't Hof
- Institute of Ageing and Chronic Disease, University of Liverpool, WH Duncan Building, West Derby Street, Liverpool, L7 8TX, UK.
| | - Gemma Charlesworth
- Institute of Ageing and Chronic Disease, University of Liverpool, WH Duncan Building, West Derby Street, Liverpool, L7 8TX, UK
| | - Ayala King
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden, Glasgow, G61 1BD, UK
- Beatson Institute for Cancer Research, Bearsden, Glasgow, G61 1BD, UK
| | - Ee Hong Tan
- Beatson Institute for Cancer Research, Bearsden, Glasgow, G61 1BD, UK
| | - Lorraine Rose
- Centre for Molecular Medicine, MRC IGMM, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Anna Daroszewska
- Institute of Ageing and Chronic Disease, University of Liverpool, WH Duncan Building, West Derby Street, Liverpool, L7 8TX, UK
| | - Amanda Prior
- Institute of Ageing and Chronic Disease, University of Liverpool, WH Duncan Building, West Derby Street, Liverpool, L7 8TX, UK
| | - Imran Ahmad
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden, Glasgow, G61 1BD, UK
- Beatson Institute for Cancer Research, Bearsden, Glasgow, G61 1BD, UK
| | - Michelle Welsh
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Ernest J Mui
- Beatson Institute for Cancer Research, Bearsden, Glasgow, G61 1BD, UK
| | - Catriona Ford
- Beatson Institute for Cancer Research, Bearsden, Glasgow, G61 1BD, UK
| | - Mark Salji
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden, Glasgow, G61 1BD, UK
- Beatson Institute for Cancer Research, Bearsden, Glasgow, G61 1BD, UK
| | - Owen Sansom
- Beatson Institute for Cancer Research, Bearsden, Glasgow, G61 1BD, UK
| | - Karen Blyth
- Beatson Institute for Cancer Research, Bearsden, Glasgow, G61 1BD, UK
| | - Hing Y Leung
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden, Glasgow, G61 1BD, UK.
- Beatson Institute for Cancer Research, Bearsden, Glasgow, G61 1BD, UK.
| |
Collapse
|
23
|
Wang H, Dai YY, Zhang WQ, Hsu PC, Yang YL, Wang YC, Chan G, Au A, Xu ZD, Jiang SJ, Wang W, Jablons DM, You L. DCLK1 is correlated with MET and ERK5 expression, and associated with prognosis in malignant pleural mesothelioma. Int J Oncol 2017; 51:91-103. [PMID: 28560410 PMCID: PMC5467791 DOI: 10.3892/ijo.2017.4021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 03/20/2017] [Indexed: 01/07/2023] Open
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive cancer for which more effective treatments are needed. In this study, strong to moderate staining of MET and ERK5 was detected in 67.1 and 48% of the analyzed 73 human mesothelioma tumors, and significant correlation of MET and ERK5 expression was identified (P<0.05). We evaluated the doublecortin-like kinase 1 (DCLK1) expression in human mesothelioma tumors. Our results showed that 50.7% of the immunohistochemistry analyzed human mesothelioma tumors have strong to moderate staining of DCLK1, and its expression is significantly correlated with MET or ERK5 expression (P<0.05). Also, the upregulation of DCLK1 is correlated with poor prognosis in MPM patients (P=0.0235). To investigate whether DCLK1 is downstream of MET/ERK5 signaling in human mesothelioma, the effect of DCLK1 expression was analyzed after treatments with either the MET inhibitor XL184 or the ERK5 inhibitor XMD8-92 in human mesothelioma cell lines. Our results showed that the MET inhibitor XL184 reduced the expression of phospho‑ERK5 and DCLK1 expression in human mesothelioma cell lines. In addition, the ERK5 inhibitor XMD8-92 reduced the expression of phospho-ERK5 and DCLK1 expression in human mesothelioma cell lines. Furthermore, XML184 and XMD8-92 treatment impaired invasion and tumor sphere formation ability of H290 mesothelioma cells. These results suggest that DCLK1 is regulated by MET/ERK5 signaling in human mesothelioma, and the MET/ERK5/DCLK1 signaling cascade could be further developed into a promising therapeutic target against mesothelioma.
Collapse
Affiliation(s)
- Hui Wang
- Department of Pulmonary Medicine, Affiliated Shandong Provincial Hospital, Shandong university, Shandong, P.R. China
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA
- Department of Respiratory Medicine, The Second Hospital of Shandong University, Shandong
| | - Yu-Yuan Dai
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA
| | - Wen-Qian Zhang
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA
- Department of Thoracic Surgery, Beijing Chao-Yang Hospital, Affiliated with Capital university of Medical Science, Beijing, P.R. China
| | - Ping-Chih Hsu
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan, R.O.C
| | - Yi-Lin Yang
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA
| | - Yu-Cheng Wang
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA
| | - Geraldine Chan
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA
| | - Alfred Au
- Division of Diagnostic Pathology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA
| | - Zhi-Dong Xu
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA
| | - Shu-Juan Jiang
- Department of Pulmonary Medicine, Affiliated Shandong Provincial Hospital, Shandong university, Shandong, P.R. China
| | - Wei Wang
- Department of Respiratory Medicine, The Second Hospital of Shandong University, Shandong
| | - David M. Jablons
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA
| | - Liang You
- Thoracic Oncology Laboratory, Department of Surgery, Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
24
|
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
|
25
|
Digiacomo G, Tusa I, Bacci M, Cipolleschi MG, Dello Sbarba P, Rovida E. Fibronectin induces macrophage migration through a SFK-FAK/CSF-1R pathway. Cell Adh Migr 2016; 11:327-337. [PMID: 27588738 DOI: 10.1080/19336918.2016.1221566] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Integrins, following binding to proteins of the extracellular matrix (ECM) including collagen, laminin and fibronectin (FN), are able to transduce molecular signals inside the cells and to regulate several biological functions such as migration, proliferation and differentiation. Besides activation of adaptor molecules and kinases, integrins transactivate Receptor Tyrosine Kinases (RTK). In particular, adhesion to the ECM may promote RTK activation in the absence of growth factors. The Colony-Stimulating Factor-1 Receptor (CSF-1R) is a RTK that supports the survival, proliferation, and motility of monocytes/macrophages, which are essential components of innate immunity and cancer development. Macrophage interaction with FN is recognized as an important aspect of host defense and wound repair. The aim of the present study was to investigate on a possible cross-talk between FN-elicited signals and CSF-1R in macrophages. FN induced migration in BAC1.2F5 and J774 murine macrophage cell lines and in human primary macrophages. Adhesion to FN determined phosphorylation of the Focal Adhesion Kinase (FAK) and Src Family Kinases (SFK) and activation of the SFK/FAK complex, as witnessed by paxillin phosphorylation. SFK activity was necessary for FAK activation and macrophage migration. Moreover, FN-induced migration was dependent on FAK in either murine macrophage cell lines or human primary macrophages. FN also induced FAK-dependent/ligand-independent CSF-1R phosphorylation, as well as the interaction between CSF-1R and β1. CSF-1R activity was necessary for FN-induced macrophage migration. Indeed, genetic or pharmacological inhibition of CSF-1R prevented FN-induced macrophage migration. Our results identified a new SFK-FAK/CSF-1R signaling pathway that mediates FN-induced migration of macrophages.
Collapse
Affiliation(s)
- Graziana Digiacomo
- a Department of Experimental and Clinical Biomedical Sciences , Università degli Studi di Firenze and Istituto Toscano Tumori , Florence , Italy
| | - Ignazia Tusa
- a Department of Experimental and Clinical Biomedical Sciences , Università degli Studi di Firenze and Istituto Toscano Tumori , Florence , Italy
| | - Marina Bacci
- a Department of Experimental and Clinical Biomedical Sciences , Università degli Studi di Firenze and Istituto Toscano Tumori , Florence , Italy
| | - Maria Grazia Cipolleschi
- a Department of Experimental and Clinical Biomedical Sciences , Università degli Studi di Firenze and Istituto Toscano Tumori , Florence , Italy
| | - Persio Dello Sbarba
- a Department of Experimental and Clinical Biomedical Sciences , Università degli Studi di Firenze and Istituto Toscano Tumori , Florence , Italy
| | - Elisabetta Rovida
- a Department of Experimental and Clinical Biomedical Sciences , Università degli Studi di Firenze and Istituto Toscano Tumori , Florence , Italy
| |
Collapse
|
26
|
Chen H, Tucker J, Wang X, Gavine PR, Phillips C, Augustin MA, Schreiner P, Steinbacher S, Preston M, Ogg D. Discovery of a novel allosteric inhibitor-binding site in ERK5: comparison with the canonical kinase hinge ATP-binding site. Acta Crystallogr D Struct Biol 2016; 72:682-93. [PMID: 27139631 PMCID: PMC4854315 DOI: 10.1107/s2059798316004502] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 03/16/2016] [Indexed: 01/20/2023] Open
Abstract
MAP kinases act as an integration point for multiple biochemical signals and are involved in a wide variety of cellular processes such as proliferation, differentiation, regulation of transcription and development. As a member of the MAP kinase family, ERK5 (MAPK7) is involved in the downstream signalling pathways of various cell-surface receptors, including receptor tyrosine kinases and G protein-coupled receptors. In the current study, five structures of the ERK5 kinase domain co-crystallized with ERK5 inhibitors are reported. Interestingly, three of the compounds bind at a novel allosteric binding site in ERK5, while the other two bind at the typical ATP-binding site. Binding of inhibitors at the allosteric site is accompanied by displacement of the P-loop into the ATP-binding site and is shown to be ATP-competitive in an enzymatic assay of ERK5 kinase activity. Kinase selectivity data show that the most potent allosteric inhibitor exhibits superior kinase selectivity compared with the two inhibitors that bind at the canonical ATP-binding site. An analysis of these structures and comparison with both a previously published ERK5-inhibitor complex structure (PDB entry 4b99) and the structures of three other kinases (CDK2, ITK and MEK) in complex with allosteric inhibitors are presented.
Collapse
Affiliation(s)
- Hongming Chen
- Chemistry Innovation Centre, Discovery Sciences, AstraZeneca R&D Mölndal, 431 83 Mölndal, Sweden
| | - Julie Tucker
- Structure and Biophysics, Discovery Sciences, AstraZeneca R&D Alderley Park, Macclesfield SK10 4TG, England
| | - Xiaotao Wang
- Innovation Centre China, AstraZeneca Asia and Emerging Markets iMed, Shanghai 201203, People’s Republic of China
| | - Paul R. Gavine
- Innovation Centre China, AstraZeneca Asia and Emerging Markets iMed, Shanghai 201203, People’s Republic of China
| | - Chris Phillips
- Structure and Biophysics, Discovery Sciences, AstraZeneca R&D Alderley Park, Macclesfield SK10 4TG, England
| | - Martin A. Augustin
- Proteros biostructures GmbH, Bunsenstrasse 7a, 82152 Martinsried, Germany
| | - Patrick Schreiner
- Proteros biostructures GmbH, Bunsenstrasse 7a, 82152 Martinsried, Germany
| | - Stefan Steinbacher
- Proteros biostructures GmbH, Bunsenstrasse 7a, 82152 Martinsried, Germany
| | - Marian Preston
- Screening Sciences, Discovery Sciences, AstraZeneca R&D Alderley Park, Macclesfield SK10 4TG, England
| | - Derek Ogg
- Structure and Biophysics, Discovery Sciences, AstraZeneca R&D Alderley Park, Macclesfield SK10 4TG, England
| |
Collapse
|
27
|
Obara Y, Nagasawa R, Nemoto W, Pellegrino MJ, Takahashi M, Habecker BA, Stork PJS, Ichiyanagi O, Ito H, Tomita Y, Ishii K, Nakahata N. ERK5 induces ankrd1 for catecholamine biosynthesis and homeostasis in adrenal medullary cells. Cell Signal 2015; 28:177-189. [PMID: 26739108 DOI: 10.1016/j.cellsig.2015.12.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 12/24/2015] [Indexed: 01/04/2023]
Abstract
Extracellular signal-regulated kinases (ERKs) play important roles in proliferation, differentiation and gene expression. In our previous study, we demonstrated that both ERK5 and ERK1/2 were responsible for neurite outgrowth and tyrosine hydroxylase (TH) expression in rat pheochromocytoma cells (PC12) (J Biol Chem 284, 23,564-23,573, 2009). However, the functional differences between ERK5 and ERK1/2 signaling in neural differentiation remain unclear. In the present study, we show that ERK5, but not ERK1/2 regulates TH levels in rat sympathetic neurons. Furthermore, microarray analysis performed in PC12 cells using ERK5 and ERK1/2-specific inhibitors, identified ankyrin repeat domain 1 (ankrd1) as an ERK5-dependent and ERK1/2-independent gene. Here, we report a novel role of the ERK5/ankrd1 signaling in regulating TH levels and catecholamine biosynthesis. Ankrd1 mRNA was induced by nerve growth factor in time- and concentration-dependent manners. TH levels were reduced by ankrd1 knockdown with no changes in the mRNA levels, suggesting that ankrd1 was involved in stabilization of TH protein. Interestingly, ubiquitination of TH was enhanced and catecholamine biosynthesis was reduced by ankrd1 knockdown. Finally, we examined the relationship of ERK5 to TH levels in human adrenal pheochromocytomas. Whereas TH levels were correlated with ERK5 levels in normal adrenal medullas, ERK5 was down-regulated and TH was up-regulated in pheochromocytomas, indicating that TH levels are regulated by alternative mechanisms in tumors. Taken together, ERK5 signaling is required for catecholamine biosynthesis during neural differentiation, in part to induce ankrd1, and to maintain appropriate TH levels. This pathway is disrupted in pathological conditions.
Collapse
Affiliation(s)
- Yutaro Obara
- Department of Cellular Signaling, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba 6-3, Aramaki, Aoba-ku, Sendai 980-8578, Japan; Department of Pharmacology, Yamagata University School of Medicine, Iida-Nishi 2-2-2, Yamagata 990-9585, Japan.
| | - Ryusuke Nagasawa
- Department of Cellular Signaling, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba 6-3, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Wataru Nemoto
- Department of Cellular Signaling, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba 6-3, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Michael J Pellegrino
- Department of Physiology and Pharmacology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239-3098, USA
| | - Maho Takahashi
- The Vollum Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239-3098, USA
| | - Beth A Habecker
- Department of Physiology and Pharmacology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239-3098, USA
| | - Philip J S Stork
- The Vollum Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239-3098, USA
| | - Osamu Ichiyanagi
- Department of Urology, Yamagata University School of Medicine, Iida-Nishi 2-2-2, Yamagata 990-9585, Japan
| | - Hiromi Ito
- Department of Urology, Yamagata University School of Medicine, Iida-Nishi 2-2-2, Yamagata 990-9585, Japan
| | - Yoshihiko Tomita
- Department of Urology, Yamagata University School of Medicine, Iida-Nishi 2-2-2, Yamagata 990-9585, Japan
| | - Kuniaki Ishii
- Department of Pharmacology, Yamagata University School of Medicine, Iida-Nishi 2-2-2, Yamagata 990-9585, Japan
| | - Norimichi Nakahata
- Department of Cellular Signaling, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba 6-3, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Gavine PR, Wang M, Yu D, Hu E, Huang C, Xia J, Su X, Fan J, Zhang T, Ye Q, Zheng L, Zhu G, Qian Z, Luo Q, Hou YY, Ji Q. Identification and validation of dysregulated MAPK7 (ERK5) as a novel oncogenic target in squamous cell lung and esophageal carcinoma. BMC Cancer 2015; 15:454. [PMID: 26040563 PMCID: PMC4453990 DOI: 10.1186/s12885-015-1455-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 05/20/2015] [Indexed: 01/23/2023] Open
Abstract
Background MAPK7/ERK5 (extracellular-signal-regulated kinase 5) functions within a canonical three-tiered MAPK (mitogen activated protein kinase) signaling cascade comprising MEK (MAPK/ERK kinase) 5, MEKK(MEK kinase) 2/3 and ERK5 itself. Despite being the least well studied of the MAPK-modules, evidence supports a role for MAPK7-signaling in the pathology of several cancer types. Methods and results Fluorescence in situ hybridization (FISH) analysis identified MAPK7 gene amplification in 4 % (3/74) of non-small cell lung cancers (NSCLC) (enriched to 6 % (3/49) in squamous cell carcinoma) and 2 % (2/95) of squamous esophageal cancers (sqEC). Immunohistochemical (IHC) analysis revealed a good correlation between MAPK7 gene amplification and protein expression. MAPK7 was validated as a proliferative oncogenic driver by performing in vitro siRNA knockdown of MAPK7 in tumor cell lines. Finally, a novel MEK5/MAPK7 co-transfected HEK293 cell line was developed and used for routine cell-based pharmacodynamic screening. Phosphorylation antibody microarray analysis also identified novel downstream pharmacodynamic (PD) biomarkers of MAPK7 kinase inhibition in tumor cells (pMEF2A and pMEF2D). Conclusions Together, these data highlight a broader role for dysregulated MAPK7 in driving tumorigenesis within niche populations of highly prevalent tumor types, and describe current efforts in establishing a robust drug discovery screening cascade. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1455-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Paul R Gavine
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, 201203, People's Republic of China.
| | - Mei Wang
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, 201203, People's Republic of China.
| | - Dehua Yu
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, 201203, People's Republic of China.
| | - Eva Hu
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, 201203, People's Republic of China.
| | - Chunlei Huang
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, 201203, People's Republic of China.
| | - Jenny Xia
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, 201203, People's Republic of China.
| | - Xinying Su
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, 201203, People's Republic of China.
| | - Joan Fan
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, 201203, People's Republic of China.
| | - Tianwei Zhang
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, 201203, People's Republic of China.
| | - Qingqing Ye
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, 201203, People's Republic of China.
| | - Li Zheng
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, 201203, People's Republic of China.
| | - Guanshan Zhu
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, 201203, People's Republic of China.
| | - Ziliang Qian
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, 201203, People's Republic of China.
| | - Qingquan Luo
- Shanghai Chest Hospital, Shanghai, People's Republic of China.
| | - Ying Yong Hou
- Shanghai Zhongshan Hospital, Shanghai, People's Republic of China.
| | - Qunsheng Ji
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, 201203, People's Republic of China.
| |
Collapse
|
31
|
Digiacomo G, Ziche M, Dello Sbarba P, Donnini S, Rovida E. Prostaglandin E2 transactivates the colony-stimulating factor-1 receptor and synergizes with colony-stimulating factor-1 in the induction of macrophage migration via the mitogen-activated protein kinase ERK1/2. FASEB J 2015; 29:2545-2554. [PMID: 25757564 DOI: 10.1096/fj.14-258939] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 02/17/2015] [Indexed: 01/02/2025]
Abstract
Prostaglandin E2 (PGE2), a key mediator of immunity, inflammation, and cancer, acts through 4 G-protein-coupled E-prostanoid receptors (EPs 1-4). Crosstalk between EPs and receptor tyrosine kinases also occurs. Colony-stimulating factor-1 receptor (CSF-1R) is an RTK that sustains the survival, proliferation, and motility of monocytes/macrophages, which are an essential component of innate immunity and cancer development. The aim of this study was to investigate on a possible crosstalk between EP and CSF-1R. In BAC1.2F5 and RAW264.7 murine macrophages, CSF-1 (EC₅₀ = 18.1 and 10.2 ng/ml, respectively) and PGE2 (EC₅₀ = 1.5 and 5.5 nM, respectively) promoted migration. PGE2 induced rapid CSF-1R phosphorylation that was dependent on Src family kinases (SFKs). CSF-1R inhibition reduced PGE2-elicited ERK1/2 phosphorylation and macrophage migration, indicating that CSF-1R plays a role in PGE2-mediated immunoregulation. EP4 appeared responsible for functional PGE2/CSF-1R crosstalk. Furthermore, PGE2 synergized with CSF-1 in inducing ERK1/2 phosphorylation and macrophage migration. ERK1/2 inhibition completely blocked migration induced by the combination CSF-1/PGE2. CSF-1/PGE2 functional interaction with respect to migration also occurred in bone marrow-derived murine macrophages (EC₅₀ CSF-1, 6.7 ng/ml; EC₅₀ PGE2, 16.7 nM). These results indicated that PGE2 transactivates CSF-1R and synergizes with its signaling at ERK1/2 level in promoting macrophage migration.
Collapse
Affiliation(s)
- Graziana Digiacomo
- *Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Florence, Italy; Dipartimento di Scienze della Vita, Università degli Studi di Siena, Siena, Italy; and Istituto Toscano Tumori, Florence, Italy
| | - Marina Ziche
- *Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Florence, Italy; Dipartimento di Scienze della Vita, Università degli Studi di Siena, Siena, Italy; and Istituto Toscano Tumori, Florence, Italy
| | - Persio Dello Sbarba
- *Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Florence, Italy; Dipartimento di Scienze della Vita, Università degli Studi di Siena, Siena, Italy; and Istituto Toscano Tumori, Florence, Italy
| | - Sandra Donnini
- *Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Florence, Italy; Dipartimento di Scienze della Vita, Università degli Studi di Siena, Siena, Italy; and Istituto Toscano Tumori, Florence, Italy
| | - Elisabetta Rovida
- *Dipartimento di Scienze Biomediche Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Florence, Italy; Dipartimento di Scienze della Vita, Università degli Studi di Siena, Siena, Italy; and Istituto Toscano Tumori, Florence, Italy
| |
Collapse
|
32
|
Honda T, Obara Y, Yamauchi A, Couvillon AD, Mason JJ, Ishii K, Nakahata N. Phosphorylation of ERK5 on Thr732 is associated with ERK5 nuclear localization and ERK5-dependent transcription. PLoS One 2015; 10:e0117914. [PMID: 25689862 PMCID: PMC4331489 DOI: 10.1371/journal.pone.0117914] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 01/05/2015] [Indexed: 12/20/2022] Open
Abstract
Extracellular signal-regulated kinases (ERKs) play critical roles in numerous cellular processes, including proliferation and differentiation. ERK5 contains a kinase domain at the N-terminal, and the unique extended C-terminal includes multiple autophosphorylation sites that enhance ERK5-dependent transcription. However, the impact of phosphorylation at the various sites remain unclear. In this study, we examined the role of phosphorylation at the ERK5 C-terminal. We found that a constitutively active MEK5 mutant phosphorylated ERK5 at the TEY motif, resulting in the sequential autophosphorylation of multiple C-terminal residues, including Thr732 and Ser769/773/775. However, when ERK1/2 was selectively activated by an oncogenic RAS mutant, ERK5 phosphorylation at Thr732 was induced without affecting the phosphorylation status at TEY or Ser769/773/775. The Thr732 phosphorylation was U0126-sensitive and was observed in a kinase-dead mutant of ERK5 as well, suggesting that ERK1/2 can phosphorylate ERK5 at Thr732. This phosphorylation was also promoted by epidermal growth factor and nerve growth factor in HEK293 and PC12 cells, respectively. The ERK5–T732A mutant was localized in the cytosol under basal conditions. In contrast, ERK5 phosphorylated at Thr732 via the RAS-ERK1/2 pathway and ERK5–T732E, which mimics the phosphorylated form, were localized in both the nucleus and cytosol. Finally, ER–32A and U0126 blocked ERK5-dependent MEF2C transcriptional activity. Based on these findings, we propose a novel cross-talk mechanism in which ERK1/2, following activation by growth factor stimulation, phosphorylates ERK5 at Thr732. This phosphorylation event is responsible for ERK5 nuclear localization and ERK5-dependent transcription.
Collapse
Affiliation(s)
- Takuto Honda
- Department of Cellular Signaling, Graduate School of Pharmaceutical Sciences, Tohoku University, 6–3 Aoba, Aramaki, Aoba-ku, Sendai 980–8578, Japan
| | - Yutaro Obara
- Department of Cellular Signaling, Graduate School of Pharmaceutical Sciences, Tohoku University, 6–3 Aoba, Aramaki, Aoba-ku, Sendai 980–8578, Japan
- Department of Pharmacology, Yamagata University School of Medicine, 2–2–2 Iida-Nishi, Yamagata, 990–9585, Japan
- * E-mail:
| | - Arata Yamauchi
- Department of Cellular Signaling, Graduate School of Pharmaceutical Sciences, Tohoku University, 6–3 Aoba, Aramaki, Aoba-ku, Sendai 980–8578, Japan
| | - Anthony D. Couvillon
- Cell Signaling Technology, 3 Trask Lane, Danvers, MA 01923, United States of America
| | - Justin J. Mason
- Cell Signaling Technology, 3 Trask Lane, Danvers, MA 01923, United States of America
| | - Kuniaki Ishii
- Department of Pharmacology, Yamagata University School of Medicine, 2–2–2 Iida-Nishi, Yamagata, 990–9585, Japan
| | - Norimichi Nakahata
- Department of Cellular Signaling, Graduate School of Pharmaceutical Sciences, Tohoku University, 6–3 Aoba, Aramaki, Aoba-ku, Sendai 980–8578, Japan
| |
Collapse
|
33
|
Wang X, Pesakhov S, Harrison JS, Kafka M, Danilenko M, Studzinski GP. The MAPK ERK5, but not ERK1/2, inhibits the progression of monocytic phenotype to the functioning macrophage. Exp Cell Res 2014; 330:199-211. [PMID: 25447310 DOI: 10.1016/j.yexcr.2014.10.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 09/04/2014] [Accepted: 10/03/2014] [Indexed: 02/06/2023]
Abstract
Intracellular signaling pathways present targets for pharmacological agents with potential for treatment of neoplastic diseases, with some disease remissions already recorded. However, cellular compensatory mechanisms usually negate the initial success. For instance, attempts to interrupt aberrant signaling downstream of the frequently mutated ras by inhibiting ERK1/2 has shown only limited usefulness for cancer therapy. Here, we examined how ERK5, that overlaps the functions of ERK1/2 in cell proliferation and survival, functions in a manner distinct from ERK1/2 in human AML cells induced to differentiate by 1,25D-dihydroxyvitamin D3 (1,25D). Using inhibitors of ERK1/2 and of MEK5/ERK5 at concentrations specific for each kinase in HL60 and U937 cells, we observed that selective inhibition of the kinase activity of ERK5, but not of ERK1/2, in the presence of 1,25D resulted in macrophage-like cell morphology and enhancement of phagocytic activity. Importantly, this was associated with increased expression of the macrophage colony stimulating factor receptor (M-CSFR), but was not seen when M-CSFR expression was knocked down. Interestingly, inhibition of ERK1/2 led to activation of ERK5 in these cells. Our results support the hypothesis that ERK5 negatively regulates the expression of M-CSFR, and thus has a restraining function on macrophage differentiation. The addition of pharmacological inhibitors of ERK5 may influence trials of differentiation therapy of AML.
Collapse
Affiliation(s)
- Xuening Wang
- Department of Pathology & Laboratory Medicine, Rutgers, NJ Medical School, 185 South Orange Ave, Newark, NJ 07103, USA
| | - Stella Pesakhov
- Department of Clinical Biochemistry & Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, PO Box 653, 84105 Beer-Sheva, Israel
| | - Jonathan S Harrison
- Department of Medicine, Rutgers, Robert Wood Johnson Medical School, New Brunswick, NJ 08903, USA
| | - Michael Kafka
- Department of Clinical Biochemistry & Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, PO Box 653, 84105 Beer-Sheva, Israel
| | - Michael Danilenko
- Department of Clinical Biochemistry & Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, PO Box 653, 84105 Beer-Sheva, Israel
| | - George P Studzinski
- Department of Pathology & Laboratory Medicine, Rutgers, NJ Medical School, 185 South Orange Ave, Newark, NJ 07103, USA.
| |
Collapse
|
34
|
Abstract
The CSF-1 receptor (CSF-1R) is activated by the homodimeric growth factors colony-stimulating factor-1 (CSF-1) and interleukin-34 (IL-34). It plays important roles in development and in innate immunity by regulating the development of most tissue macrophages and osteoclasts, of Langerhans cells of the skin, of Paneth cells of the small intestine, and of brain microglia. It also regulates the differentiation of neural progenitor cells and controls functions of oocytes and trophoblastic cells in the female reproductive tract. Owing to this broad tissue expression pattern, it plays a central role in neoplastic, inflammatory, and neurological diseases. In this review we summarize the evolution, structure, and regulation of expression of the CSF-1R gene. We discuss the structures of CSF-1, IL-34, and the CSF-1R and the mechanism of ligand binding to and activation of the receptor. We further describe the pathways regulating macrophage survival, proliferation, differentiation, and chemotaxis downstream from the CSF-1R.
Collapse
Affiliation(s)
- E Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
35
|
Heo KS, Cushman HJ, Akaike M, Woo CH, Wang X, Qiu X, Fujiwara K, Abe JI. ERK5 activation in macrophages promotes efferocytosis and inhibits atherosclerosis. Circulation 2014; 130:180-91. [PMID: 25001623 DOI: 10.1161/circulationaha.113.005991] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Efferocytosis is a process by which dead and dying cells are removed by phagocytic cells. Efferocytosis by macrophages is thought to curb the progression of atherosclerosis, but the mechanistic insight of this process is lacking. METHODS AND RESULTS When macrophages were fed apoptotic cells or treated with pitavastatin in vitro, efferocytosis-related signaling and phagocytic capacity were upregulated in an ERK5 activity-dependent manner. Macrophages isolated from macrophage-specific ERK5-null mice exhibited reduced efferocytosis and levels of gene and protein expression of efferocytosis-related molecules. When these mice were crossed with low-density lipoprotein receptor(-/-) mice and fed a high-cholesterol diet, atherosclerotic plaque formation was accelerated, and the plaques had more advanced and vulnerable morphology. CONCLUSIONS Our results demonstrate that ERK5, which is robustly activated by statins, is a hub molecule that upregulates macrophage efferocytosis, thereby suppressing atherosclerotic plaque formation. Molecules that upregulate ERK5 and its signaling in macrophages may be good drug targets for suppressing cardiovascular diseases.
Collapse
Affiliation(s)
- Kyung-Sun Heo
- From the Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY (K.H., H.J.C., C.W., K.F., J.A.); Department of Medical Education, Institute of Health Biosciences, University of Tokushima Graduate School, Kuramoto-cho, Tokushima, Japan (M.A.); Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom (X.W.); and Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY (X.Q.).
| | - Hannah J Cushman
- From the Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY (K.H., H.J.C., C.W., K.F., J.A.); Department of Medical Education, Institute of Health Biosciences, University of Tokushima Graduate School, Kuramoto-cho, Tokushima, Japan (M.A.); Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom (X.W.); and Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY (X.Q.)
| | - Masashi Akaike
- From the Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY (K.H., H.J.C., C.W., K.F., J.A.); Department of Medical Education, Institute of Health Biosciences, University of Tokushima Graduate School, Kuramoto-cho, Tokushima, Japan (M.A.); Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom (X.W.); and Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY (X.Q.)
| | - Chang-Hoon Woo
- From the Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY (K.H., H.J.C., C.W., K.F., J.A.); Department of Medical Education, Institute of Health Biosciences, University of Tokushima Graduate School, Kuramoto-cho, Tokushima, Japan (M.A.); Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom (X.W.); and Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY (X.Q.)
| | - Xin Wang
- From the Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY (K.H., H.J.C., C.W., K.F., J.A.); Department of Medical Education, Institute of Health Biosciences, University of Tokushima Graduate School, Kuramoto-cho, Tokushima, Japan (M.A.); Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom (X.W.); and Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY (X.Q.)
| | - Xing Qiu
- From the Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY (K.H., H.J.C., C.W., K.F., J.A.); Department of Medical Education, Institute of Health Biosciences, University of Tokushima Graduate School, Kuramoto-cho, Tokushima, Japan (M.A.); Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom (X.W.); and Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY (X.Q.)
| | - Keigi Fujiwara
- From the Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY (K.H., H.J.C., C.W., K.F., J.A.); Department of Medical Education, Institute of Health Biosciences, University of Tokushima Graduate School, Kuramoto-cho, Tokushima, Japan (M.A.); Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom (X.W.); and Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY (X.Q.)
| | - Jun-ichi Abe
- From the Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY (K.H., H.J.C., C.W., K.F., J.A.); Department of Medical Education, Institute of Health Biosciences, University of Tokushima Graduate School, Kuramoto-cho, Tokushima, Japan (M.A.); Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom (X.W.); and Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY (X.Q.).
| |
Collapse
|
36
|
Liu J, Krautzberger AM, Sui SH, Hofmann OM, Chen Y, Baetscher M, Grgic I, Kumar S, Humphreys BD, Hide WA, McMahon AP. Cell-specific translational profiling in acute kidney injury. J Clin Invest 2014; 124:1242-54. [PMID: 24569379 DOI: 10.1172/jci72126] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 12/05/2013] [Indexed: 02/01/2023] Open
Abstract
Acute kidney injury (AKI) promotes an abrupt loss of kidney function that results in substantial morbidity and mortality. Considerable effort has gone toward identification of diagnostic biomarkers and analysis of AKI-associated molecular events; however, most studies have adopted organ-wide approaches and have not elucidated the interplay among different cell types involved in AKI pathophysiology. To better characterize AKI-associated molecular and cellular events, we developed a mouse line that enables the identification of translational profiles in specific cell types. This strategy relies on CRE recombinase-dependent activation of an EGFP-tagged L10a ribosomal protein subunit, which allows translating ribosome affinity purification (TRAP) of mRNA populations in CRE-expressing cells. Combining this mouse line with cell type-specific CRE-driver lines, we identified distinct cellular responses in an ischemia reperfusion injury (IRI) model of AKI. Twenty-four hours following IRI, distinct translational signatures were identified in the nephron, kidney interstitial cell populations, vascular endothelium, and macrophages/monocytes. Furthermore, TRAP captured known IRI-associated markers, validating this approach. Biological function annotation, canonical pathway analysis, and in situ analysis of identified response genes provided insight into cell-specific injury signatures. Our study provides a deep, cell-based view of early injury-associated molecular events in AKI and documents a versatile, genetic tool to monitor cell-specific and temporal-specific biological processes in disease modeling.
Collapse
|
37
|
Chromatin-associated CSF-1R binds to the promoter of proliferation-related genes in breast cancer cells. Oncogene 2013; 33:4359-64. [PMID: 24362524 PMCID: PMC4141303 DOI: 10.1038/onc.2013.542] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 10/04/2013] [Accepted: 11/04/2013] [Indexed: 12/18/2022]
Abstract
The colony-stimulating factor-1 (CSF-1) and its receptor CSF-1R physiologically regulate the monocyte/macrophage system, trophoblast implantation and breast development. An abnormal CSF-1R expression has been documented in several human epithelial tumors, including breast carcinomas. We recently demonstrated that CSF-1/CSF-1R signaling drives proliferation of breast cancer cells via ‘classical' receptor tyrosine kinase signaling, including activation of the extracellular signal-regulated kinase 1/2. In this paper, we show that CSF-1R can also localize within the nucleus of breast cancer cells, either cell lines or tissue specimens, irrespectively of their intrinsic molecular subtype. We found that the majority of nuclear CSF-1R is located in the chromatin-bound subcellular compartment. Chromatin immunoprecipitation revealed that CSF-1R, once in the nucleus, binds to the promoters of the proliferation-related genes CCND1, c-JUN and c-MYC. CSF-1R also binds the promoter of its ligand CSF-1 and positively regulates CSF-1 expression. The existence of such a receptor/ligand regulatory loop is a novel aspect of CSF-1R signaling. Moreover, our results provided the first evidence of a novel localization site of CSF-1R in breast cancer cells, suggesting that CSF-1R could act as a transcriptional regulator on proliferation-related genes.
Collapse
|
38
|
Obara Y. [Roles of ERK5 in neuronal cells]. Nihon Yakurigaku Zasshi 2013; 141:251-5. [PMID: 23665555 DOI: 10.1254/fpj.141.251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
39
|
Shukla A, Miller JM, Cason C, Sayan M, MacPherson MB, Beuschel SL, Hillegass J, Vacek PM, Pass HI, Mossman BT. Extracellular signal-regulated kinase 5: a potential therapeutic target for malignant mesotheliomas. Clin Cancer Res 2013; 19:2071-83. [PMID: 23446998 DOI: 10.1158/1078-0432.ccr-12-3202] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE Malignant mesothelioma is a devastating disease with a need for new treatment strategies. In the present study, we showed the importance of extracellular signal-regulated kinase 5 (ERK5) in malignant mesothelioma tumor growth and treatment. EXPERIMENTAL DESIGN ERK5 as a target for malignant mesothelioma therapy was verified using mesothelial and mesothelioma cell lines as well as by xenograft severe combined immunodeficient (SCID) mouse models. RESULTS We first showed that crocidolite asbestos activated ERK5 in LP9 cells and mesothelioma cell lines exhibit constitutive activation of ERK5. Addition of doxorubicin resulted in further activation of ERK5 in malignant mesothelioma cells. ERK5 silencing increased doxorubicin-induced cell death and doxorubicin retention in malignant mesothelioma cells. In addition, shERK5 malignant mesothelioma lines exhibited both attenuated colony formation on soft agar and invasion of malignant mesothelioma cells in vitro that could be related to modulation of gene expression linked to cell proliferation, apoptosis, migration/invasion, and drug resistance as shown by microarray analysis. Most importantly, injection of shERK5 malignant mesothelioma cell lines into SCID mice showed significant reduction in tumor growth using both subcutaneous and intraperitoneal models. Assessment of selected human cytokine profiles in peritoneal lavage fluid from intraperitoneal shERK5 and control tumor-bearing mice showed that ERK5 was critical in regulation of various proinflammatory (RANTES/CCL5, MCP-1) and angiogenesis-related (interleukin-8, VEGF) cytokines. Finally, use of doxorubicin and cisplatin in combination with ERK5 inhibition showed further reduction in tumor weight and volume in the intraperitoneal model of tumor growth. CONCLUSION ERK5 inhibition in combination with chemotherapeutic drugs is a beneficial strategy for combination therapy in patients with malignant mesothelioma.
Collapse
Affiliation(s)
- Arti Shukla
- Department of Pathology, University of Vermont College of Medicine, Burlington, Vermont 05405, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Perez-Madrigal D, Finegan KG, Paramo B, Tournier C. The extracellular-regulated protein kinase 5 (ERK5) promotes cell proliferation through the down-regulation of inhibitors of cyclin dependent protein kinases (CDKs). Cell Signal 2012; 24:2360-8. [PMID: 22917534 DOI: 10.1016/j.cellsig.2012.08.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 07/27/2012] [Accepted: 08/07/2012] [Indexed: 01/11/2023]
Abstract
Activation of the extracellular-regulated protein kinase 5 (ERK5) has been associated with mitogenic signal transduction. However, conflicting findings have challenged the idea that ERK5 is a critical regulator of cell proliferation. We have addressed this issue by testing the effect of the conditional loss of ERK5 in primary fibroblasts. We have discovered that ERK5 suppressed the expression of the cyclin dependent protein kinase (CDKs) inhibitors, p21 and p27, by decreasing mRNA and protein stability, respectively. As a result, low level CDK2 activity detected in ERK5-deficient cells correlated with a defect in G1 to S phase transition of the cell cycle. Similarly, we found that the malignant MDA-MB-231 human breast cancer cell line was dependent on ERK5 to proliferate. We propose that ERK5 blocks p21 expression in MDA-MB-231 cells via a mechanism that implicates c-Myc-dependent transcriptional regulation of the miR-17-92 cluster. Together with evidence that cancer patients with poor prognosis display a high level of expression of components of the ERK5 signaling pathway, these findings support the hypothesis that ERK5 can be a potential target for cancer therapy.
Collapse
|
41
|
Wilhelmsen K, Mesa KR, Lucero J, Xu F, Hellman J. ERK5 protein promotes, whereas MEK1 protein differentially regulates, the Toll-like receptor 2 protein-dependent activation of human endothelial cells and monocytes. J Biol Chem 2012; 287:26478-94. [PMID: 22707717 DOI: 10.1074/jbc.m112.359489] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Endothelial cell (EC) Toll-like receptor 2 (TLR2) activation up-regulates the expression of inflammatory mediators and of TLR2 itself and modulates important endothelial functions, including coagulation and permeability. We defined TLR2 signaling pathways in EC and tested the hypothesis that TLR2 signaling differs in EC and monocytes. We found that ERK5, heretofore unrecognized as mediating TLR2 activation in any cell type, is a central mediator of TLR2-dependent inflammatory signaling in human umbilical vein endothelial cells, primary human lung microvascular EC, and human monocytes. Additionally, we observed that, although MEK1 negatively regulates TLR2 signaling in EC, MEK1 promotes TLR2 signaling in monocytes. We also noted that activation of TLR2 led to the up-regulation of intracellularly expressed TLR2 and inflammatory mediators via NF-κB, JNK, and p38-MAPK. Finally, we found that p38-MAPK, JNK, ERK5, and NF-κB promote the attachment of human neutrophils to lung microvascular EC that were pretreated with TLR2 agonists. This study newly identifies ERK5 as a key regulator of TLR2 signaling in EC and monocytes and indicates that there are fundamental differences in TLR signaling pathways between EC and monocytes.
Collapse
Affiliation(s)
- Kevin Wilhelmsen
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, California 94143,USA.
| | | | | | | | | |
Collapse
|
42
|
Abstract
The MEK5 [MAPK (mitogen-activated protein kinase)/ERK (extracellular-signal-regulated kinase) kinase 5]/ERK5 pathway is the least well studied MAPK signalling module. It has been proposed to play a role in the pathology of cancer. In the present paper, we review the role of the MEK5/ERK5 pathway using the 'hallmarks of cancer' as a framework and consider how this pathway is deregulated. As well as playing a key role in endothelial cell survival and tubular morphogenesis during tumour neovascularization, ERK5 is also emerging as a regulator of tumour cell invasion and migration. Several oncogenes can stimulate ERK5 activity, and protein levels are increased by a novel amplification at chromosome locus 17p11 and by down-regulation of the microRNAs miR-143 and miR-145. Together, these finding underscore the case for further investigation into understanding the role of ERK5 in cancer.
Collapse
|
43
|
Shannon LA, McBurney TM, Wells MA, Roth ME, Calloway PA, Bill CA, Islam S, Vines CM. CCR7/CCL19 controls expression of EDG-1 in T cells. J Biol Chem 2012; 287:11656-64. [PMID: 22334704 DOI: 10.1074/jbc.m111.310045] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
T lymphocytes circulate between the blood, tissues, and lymph. These T cells carry out immune functions, using the C-C chemokine receptor 7 (CCR7) and its cognate ligands, CCL19 and CCL21, to enter and travel through the lymph nodes. Distinct roles for each ligand in regulating T lymphocyte trafficking have remained elusive. We report that in the human T cell line HuT78 and in primary murine T lymphocytes, signaling from CCR7/CCL19 leads to increased expression and phosphorylation of extracellular signal-regulated kinase 5 (ERK5) within eight hours of stimulation. Within 48-72 h we observed peak levels of endothelial differentiation gene 1 (EDG-1), which mediates the egress of T lymphocytes from lymph nodes. The increased expression of EDG-1 was preceded by up-regulation of its transcription factor, Krüppel-like factor 2 (KLF-2). To determine the cellular effect of disrupting ERK5 signaling from CCR7, we examined the migration of ERK5(flox/flox)/Lck-Cre murine T cells to EDG-1 ligands. While CCL19-stimulated ERK5(flox/flox) naïve T cells showed increased migration to EDG-1 ligands at 48 h, the migration of ERK5(flox/flox)/Lck-Cre T cells remained at a basal level. Accordingly, we define a novel signaling pathway that controls EDG-1 up-regulation following stimulation of T cells by CCR7/CCL19. This is the first report to link the two signaling events that control migration through the lymph nodes: CCR7 mediates entry into the lymph nodes and EDG-1 signaling controls their subsequent exit.
Collapse
Affiliation(s)
- Laura A Shannon
- Department of Microbiology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Wang Y, Piper MG, Marsh CB. The role of Src family kinases in mediating M-CSF receptor signaling and monocytic cell survival. ACTA ACUST UNITED AC 2012. [DOI: 10.4236/abb.2012.35077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
45
|
Tesser-Gamba F, Petrilli AS, de Seixas Alves MT, Filho RJG, Juliano Y, Toledo SRC. MAPK7 and MAP2K4 as prognostic markers in osteosarcoma. Hum Pathol 2011; 43:994-1002. [PMID: 22154052 DOI: 10.1016/j.humpath.2011.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 08/10/2011] [Accepted: 08/12/2011] [Indexed: 10/14/2022]
Abstract
Osteosarcoma is a class of cancer originating from the bone, affecting mainly children and young adults. Cytogenetic studies showed the presence of rearrangements and recurrent gains in specific chromosomal regions, indicating the possible involvement of genes located in these regions during the pathogenesis of osteosarcoma. These studies investigated expression of 10 genes located in the chromosomal region involved in abnormalities in osteosarcoma, 1p36, 17p, and chromosome 19. The purpose of this study was to investigate the expression profile of genes located in regions involved in chromosomal rearrangements in osteosarcoma. We used quantitative real-time polymerase chain reaction to investigate the expression of 10 genes located in 1p36.3 (MTHFR, ERRFI1, FGR, E2F2), 17p (MAPK7, MAP2K4), and chromosome 19 (BBC3, FOSB, JUND, and RRAS), in 70 samples taken from 30 patients (30 prechemotherapy, 30 postchemotherapy, and 10 metastases specimens) and 10 healthy bones as a control sample. The most interesting results showed a strong association between the expression levels of MAPK7 and MAP2K4 genes and clinical parameters of osteosarcoma. Overexpression of these genes was significantly associated to a poor response to treatment (P = .0001 and P = .0049, respectively), tumor progression, and worse overall survival (P = .0052 and P = .0085, respectively), suggesting that MAPK7 and MAP2K4 could play an important role in osteosarcoma tumorigenesis. Thus, these genes could be good markers in assessing response to treatment and development of osteosarcoma.
Collapse
Affiliation(s)
- Francine Tesser-Gamba
- Department of Pediatrics, Pediatric Oncology Institute (Grupo de Apoio ao Adolescente e à Criança com Câncer), Federal University of São Paulo, São Paulo, SP 04023-062, Brazil.
| | | | | | | | | | | |
Collapse
|
46
|
Morandi A, Barbetti V, Riverso M, Dello Sbarba P, Rovida E. The colony-stimulating factor-1 (CSF-1) receptor sustains ERK1/2 activation and proliferation in breast cancer cell lines. PLoS One 2011; 6:e27450. [PMID: 22096574 PMCID: PMC3212567 DOI: 10.1371/journal.pone.0027450] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 10/17/2011] [Indexed: 12/20/2022] Open
Abstract
Breast cancer is the second leading cause of cancer-related deaths in western countries. Colony-Stimulating Factor-1 (CSF-1) and its receptor (CSF-1R) regulate macrophage and osteoclast production, trophoblast implantation and mammary gland development. The expression of CSF-1R and/or CSF-1 strongly correlates with poor prognosis in several human epithelial tumors, including breast carcinomas. We demonstrate that CSF-1 and CSF-1R are expressed, although at different levels, in 16/17 breast cancer cell lines tested with no differences among molecular subtypes. The role of CSF-1/CSF-1R in the proliferation of breast cancer cells was then studied in MDAMB468 and SKBR3 cells belonging to different subtypes. CSF-1 administration induced ERK1/2 phosphorylation and enhanced cell proliferation in both cell lines. Furthermore, the inhibition of CSF-1/CSF-1R signaling, by CSF-1R siRNA or imatinib treatment, impaired CSF-1 induced ERK1/2 activation and cell proliferation. We also demonstrate that c-Jun, cyclin D1 and c-Myc, known for their involvement in cell proliferation, are downstream CSF-1R in breast cancer cells. The presence of a proliferative CSF-1/CSF-1R autocrine loop involving ERK1/2 was also found. The wide expression of the CSF-1/CSF-1R pair across breast cancer cell subtypes supports CSF-1/CSF-1R targeting in breast cancer therapy.
Collapse
Affiliation(s)
- Andrea Morandi
- Dipartimento di Patologia e Oncologia Sperimentali, Università degli Studi di Firenze, and Istituto Toscano Tumori, Firenze, Italy
| | - Valentina Barbetti
- Dipartimento di Patologia e Oncologia Sperimentali, Università degli Studi di Firenze, and Istituto Toscano Tumori, Firenze, Italy
| | - Maria Riverso
- Dipartimento di Patologia e Oncologia Sperimentali, Università degli Studi di Firenze, and Istituto Toscano Tumori, Firenze, Italy
| | - Persio Dello Sbarba
- Dipartimento di Patologia e Oncologia Sperimentali, Università degli Studi di Firenze, and Istituto Toscano Tumori, Firenze, Italy
- * E-mail: (PDS); (ER)
| | - Elisabetta Rovida
- Dipartimento di Patologia e Oncologia Sperimentali, Università degli Studi di Firenze, and Istituto Toscano Tumori, Firenze, Italy
- * E-mail: (PDS); (ER)
| |
Collapse
|
47
|
Zhao J, Kyotani Y, Itoh S, Nakayama H, Isosaki M, Yoshizumi M. Big mitogen-activated protein kinase 1 protects cultured rat aortic smooth muscle cells from oxidative damage. J Pharmacol Sci 2011; 116:173-80. [PMID: 21597237 DOI: 10.1254/jphs.11015fp] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Oxidative stress is considered a major mediator of arteriosclerosis. In vascular smooth muscle cells, oxidative stress-induced cell death (including apoptosis) is probably related to arterial calcification in arteriosclerosis. Big mitogen-activated protein kinase-1 / extracellular signal-regulated kinase 5 (BMK1/ERK5) is a newly identified member of the mitogen-activated protein kinases family. Like Src tyrosine kinase, BMK1/ERK5 is known to be sensitive to oxidative stress; however, its pathophysiological significance is poorly understood. In this study, we investigated the involvement of BMK1 and Src in H(2)O(2)-induced cell death using cultured rat aortic smooth muscle cells (RASMCs). Cell apoptosis was evaluated by using the TdT-mediated dUTP nick end labeling (TUNEL) method, and BMK1 and Src activities were determined by Western blotting. The main results are as follows: 1) BMK1 and Src were activated by H(2)O(2) in a time- and concentration-dependent manner in RASMCs; 2) BMK1 activation by H(2)O(2) was attenuated both in Src-knockdown RASMCs and in RASMCs pretreated with 4-amino-5-(4-chloro-phenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP2), a Src family kinases inhibitor; and 3) H(2)O(2)-induced cell death was increased in BMK1- and Src-knockdown RASMCs as well as in PP2-treated RASMCs. These findings suggested that Src and BMK1 may play defensive and resistive roles against oxidative stress-induced death in RASMCs.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Pharmacology, Nara Medical University School of Medicine, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Barra V, Kuhn AM, von Knethen A, Weigert A, Brüne B. Apoptotic cell-derived factors induce arginase II expression in murine macrophages by activating ERK5/CREB. Cell Mol Life Sci 2011; 68:1815-27. [PMID: 20949368 PMCID: PMC11115119 DOI: 10.1007/s00018-010-0537-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2010] [Revised: 08/27/2010] [Accepted: 09/23/2010] [Indexed: 02/07/2023]
Abstract
Apoptotic cell (AC)-derived factors alter the physiology of macrophages (MΦs) towards a regulatory phenotype, characterized by reduced nitric oxide (NO) production. Impaired NO formation in response to AC-conditioned medium (CM) was facilitated by arginase II (ARG II) expression, which competes with inducible NO synthase for L-arginine. Here we explored signaling pathways allowing CM to upregulate ARG II in RAW264.7 MΦs. Sphingosine-1-phosphate (S1P) was required and acted synergistically with a so far unidentified factor to elicit high ARG II expression. S1P activated S1P(2), since S1P(2) knockdown prevented ARG II upregulation. Furthermore, ERK5 knockdown attenuated CM-mediated ARG II protein induction. CREB was implicated as shown by EMSA analysis and decoy-oligonucleotides scavenging CREB in RAW264.7 MΦs, which blocked ARG II expression. We conclude that AC-derived S1P binds to S1P(2) and acts synergistically with other factors to activate ERK5 and concomitantly CREB. This signaling cascade shapes an anti-inflammatory MΦ phenotype by ARG II induction.
Collapse
Affiliation(s)
- Vera Barra
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Anne-Marie Kuhn
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Andreas von Knethen
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Andreas Weigert
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Bernhard Brüne
- Faculty of Medicine, Institute of Biochemistry I, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| |
Collapse
|
49
|
Abstract
The big mitogen activated protein kinase 1 (BMK1) pathway is the most recently discovered and least-studied mammalian mitogen-activated protein (MAP) kinase cascade, ubiquitously expressed in all types of cancer cells tested so far. Mitogens and oncogenic signals strongly activate this cellular MAP kinase pathway, thereby passing down proliferative, survival, chemoresistance, invasive, and angiogenic signals in tumor cells. Recently, several pharmacologic small molecule inhibitors of this pathway have been developed. Among them, the BMK1 inhibitor XMD8-92 blocks cellular BMK1 activation and significantly suppresses tumor growth in lung and cervical tumor models and is well tolerated in animals. On the other hand, MEK5 inhibitors, BIX02188, BIX02189, and compound 6, suppress cellular MEK5 activity, but no data exist to date on their effectiveness in animals.
Collapse
Affiliation(s)
- Qingkai Yang
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, USA
| | | |
Collapse
|
50
|
Wang X, Gocek E, Novik V, Harrison JS, Danilenko M, Studzinski GP. Inhibition of Cot1/Tlp2 oncogene in AML cells reduces ERK5 activation and up-regulates p27Kip1 concomitant with enhancement of differentiation and cell cycle arrest induced by silibinin and 1,25-dihydroxyvitamin D(3). Cell Cycle 2010; 9:4542-51. [PMID: 21084834 DOI: 10.4161/cc.9.22.13790] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Acute myelogenous leukemia (AML) is a disease characterized by dysregulated cell proliferation associated with impaired cell differentiation, and current treatment regimens rarely save the patient. Thus, new mechanism-based approaches are needed to improve prognosis of this disease. We have investigated in preclinical studies the potential anti-leukemia use of the plant-derived polyphenol Silibinin (SIL) in combination with 1,25-dihydroxyvitamin D3 (1,25D). Although most of the leukemic blasts ex vivo responded by differentiation to treatment with this combination, the reasons for the absence of SIL-1,25D synergy in some cases were unclear. Here we report that failure of SIL to enhance the action of 1,25D is likely due to the SIL-induced increase in the activity of differentiation-antagonizing cell components, such as ERK5. This kinase is under the control of Cot1/Tlp2, and inhibition of Cot1 activity by a specific pharmacological inhibitor 4-(3-chloro-4-fluorophenylamino)-6-(pyridin-3-yl-methylamino-3-cyano-[1-7]-naphthyridine, or by Cot1 siRNA, increases the differentiation by SIL/1,25D combinations. Conversely, over-expression of a Cot1 construct increases the cellular levels of P-ERK5, and SIL/1,25D-induced differentiation and cell cycle arrest are diminished. It appears that reduction in ERK5 activity by inhibition of Cot1 allows SIL to augment the expression of 1,25D-induced differentiation promoting factors and cell cycle regulators such as p27 (Kip1) , which leads to cell cycle arrest. This study shows that in some cell contexts SIL/1,25D can promote expression of both differentiation-promoting and differentiation-inhibiting genes, and that the latter can be neutralized by a highly specific pharmacological inhibitor, suggesting a potential for supplementing treatment of AML with this combination of agents.
Collapse
Affiliation(s)
- Xuening Wang
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, University of Medicine and Dentistry New Jersey, Newark, USA
| | | | | | | | | | | |
Collapse
|