1
|
Kubatka P, Bojkova B, Nosalova N, Huniadi M, Samuel SM, Sreenesh B, Hrklova G, Kajo K, Hornak S, Cizkova D, Bubnov R, Smokovski I, Büsselberg D, Golubnitschaja O. Targeting the MAPK signaling pathway: implications and prospects of flavonoids in 3P medicine as modulators of cancer cell plasticity and therapeutic resistance in breast cancer patients. EPMA J 2025. [DOI: 10.1007/s13167-025-00407-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 03/24/2025] [Indexed: 04/21/2025]
Abstract
Abstract
Cancer drug resistance poses a significant challenge in oncology, primarily driven by cancer cell plasticity, which promotes tumor initiation, progression, metastasis, and therapeutic evasion in many different cancers. Breast cancers (BCs) are a prominent example of that, with an estimated 2.3 million new cases and 670,000 BC-related deaths registered worldwide annually. Triple-negative BC is especially challenging for treatments demonstrating particularly aggressive disease course, an early manifestation of metastatic disease, frequent drug-resistant cancer types, and poor individual outcomes. Although chemosensitizing agents have been developed, their clinical utility in oncology remains unproven. The mitogen-activated protein kinase (MAPK) pathway is considered a critical regulator of intracellular and extracellular signaling highly relevant for both — genetic and epigenetic modifications. Dysregulation of the MAPK signaling pathways plays a significant role in conferring chemoresistance in BC. Contextually, targeting the MAPK pathway represents a promising strategy for overcoming drug resistance and enhancing the therapeutic efficacy of anticancer agents in BC treatment. On the other hand, flavonoids, a prominent class of phytochemicals, are key modulators of MAPK signaling. Flavonoids interact with the ERK, JNK, p38, and ERK5 pathways of the MAPK signaling cascade and present a promising avenue for developing novel anti-cancer therapies and re-sensitizing agents for the treatment of BC. Compounds such as quercetin, kaempferol, genistein, luteolin, myricetin, EGCG, baicalein, baicalin, nobiletin, morin, delphinidin, acacetin, isorhamnetin, apigenin, silymarin, among others, have been identified as specific modulators of MAPK signaling, exerting complex downstream effects in BC cells increasing therewith drug efficacy and suppressing tumor growth and aggressivity. These properties reflect mechanisms of great clinical relevance to overcome therapeutic resistance in overall BC management. This article highlights corresponding mechanisms and provides clinically relevant illustrations in the framework of 3P medicine for primary (protection of individuals at high risk against health-to-disease transition) and secondary care (protection against metastatic BC progression). 3PM novelty makes good use of patient phenotyping and stratification, predictive multi-level diagnostics, and application of Artificial Intelligence (AI) tools to the individualized interpretation of big data — all proposed for cost-effective treatments tailored to individualized patient profiles with clear benefits to patients and advanced BC management.
Collapse
|
2
|
Romero-Pérez I, Díaz-Rodríguez E, Sánchez-Díaz L, Montero JC, Pandiella A. Peptidylarginine deiminase 3 modulates response to neratinib in HER2 positive breast cancer. Oncogenesis 2024; 13:30. [PMID: 39097594 PMCID: PMC11297914 DOI: 10.1038/s41389-024-00531-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/22/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024] Open
Abstract
Neratinib is a tyrosine kinase inhibitor that is used for the therapy of patients with HER2+ breast tumors. However, despite its clinical benefit, resistance to the drug may arise. Here we have created cellular models of neratinib resistance to investigate the mechanisms underlying such resistance. Chronic neratinib exposure of BT474 human HER2+ breast cancer cells resulted in the selection of several clones resistant to the antiproliferative action of the drug. The clones were characterized biochemically and biologically using a variety of techniques. These clones retained HER2 levels similar to parental cells. Knockdown experiments showed that the neratinib-resistant clones retained oncogenic dependence on HER2. Moreover, the tyrosine phosphorylation status of BT474 and the resistant clones was equally sensitive to neratinib. Transcriptomic and Western analyses showed that peptidylarginine deiminase 3 was overexpressed in the three neratinib-resistant clones studied but was undetectable in BT474 cells. Experiments performed in the neratinib-resistant clones showed that reduction of PADI3 or inhibition of its function restored sensitivity to the antiproliferative action of neratinib. Moreover, overexpression of FLAG-tagged PADI3 in BT474 cells provoked resistance to the antiproliferative action of neratinib. Together, these results uncover a role of PADI3 in the regulation of sensitivity to neratinib in breast cancer cells overexpressing HER2 and open the possibility of using PADI3 inhibitors to fight resistance to neratinib.
Collapse
Affiliation(s)
- Inés Romero-Pérez
- Instituto de Biología Molecular y Celular del Cáncer. CSIC-Universidad de Salamanca, Salamanca, Spain
- CIBERONC, Madrid, Spain
| | - Elena Díaz-Rodríguez
- Instituto de Biología Molecular y Celular del Cáncer. CSIC-Universidad de Salamanca, Salamanca, Spain
- CIBERONC, Madrid, Spain
- Department of Biochemistry, Universidad de Salamanca, Salamanca, Spain
| | - Laura Sánchez-Díaz
- Instituto de Biología Molecular y Celular del Cáncer. CSIC-Universidad de Salamanca, Salamanca, Spain
- CIBERONC, Madrid, Spain
| | - Juan Carlos Montero
- Instituto de Biología Molecular y Celular del Cáncer. CSIC-Universidad de Salamanca, Salamanca, Spain
- CIBERONC, Madrid, Spain
- Department of Pathologic Anatomy and IBSAL, Salamanca, Spain
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer. CSIC-Universidad de Salamanca, Salamanca, Spain.
- CIBERONC, Madrid, Spain.
| |
Collapse
|
3
|
Montero JC, Del Carmen S, Abad M, Sayagués JM, Barbáchano A, Fernández-Barral A, Muñoz A, Pandiella A. An amino acid transporter subunit as an antibody-drug conjugate target in colorectal cancer. J Exp Clin Cancer Res 2023; 42:200. [PMID: 37559159 PMCID: PMC10410906 DOI: 10.1186/s13046-023-02784-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/28/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Advanced colorectal cancer (CRC) is difficult to treat. For that reason, the development of novel therapeutics is necessary. Here we describe a potentially actionable plasma membrane target, the amino acid transporter protein subunit CD98hc. METHODS Western blot and immunohistochemical analyses of CD98hc protein expression were carried out on paired normal and tumoral tissues from patients with CRC. Immunofluorescence and western studies were used to characterize the action of a DM1-based CD98hc-directed antibody-drug conjugate (ADC). MTT and Annexin V studies were performed to evaluate the effect of the anti-CD98hc-ADC on cell proliferation and apoptosis. CRISPR/Cas9 and shRNA were used to explore the specificity of the ADC. In vitro analyses of the antitumoral activity of the anti-CD98hc-ADC on 3D patient-derived normal as well as tumoral organoids were also carried out. Xenografted CRC cells and a PDX were used to analyze the antitumoral properties of the anti-CD98hc-ADC. RESULTS Genomic as well proteomic analyses of paired normal and tumoral samples showed that CD98hc expression was significantly higher in tumoral tissues as compared to levels of CD98hc present in the normal colonic tissue. In human CRC cell lines, an ADC that recognized the CD98hc ectodomain, reached the lysosomes and exerted potent antitumoral activity. The specificity of the CD98hc-directed ADC was demonstrated using CRC cells in which CD98hc was decreased by shRNA or deleted using CRISPR/Cas9. Studies in patient-derived organoids verified the antitumoral action of the anti-CD98hc-ADC, which largely spared normal tissue-derived colon organoids. In vivo studies using xenografted CRC cells or patient-derived xenografts confirmed the antitumoral activity of the anti-CD98hc-ADC. CONCLUSIONS The studies herewith reported indicate that CD98hc may represent a novel ADC target that, upon well-designed clinical trials, could be used to increase the therapeutic armamentarium against CRC.
Collapse
Affiliation(s)
- Juan Carlos Montero
- Institute of Biomedical Research of Salamanca (IBSAL), Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca), Salamanca, Spain.
- Department of Pathology and IBSAL, University Hospital of Salamanca, Salamanca, Spain.
- CIBERONC, Madrid, Spain.
| | - Sofía Del Carmen
- Department of Pathology and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| | - Mar Abad
- Department of Pathology and IBSAL, University Hospital of Salamanca, Salamanca, Spain
| | - José M Sayagués
- Department of Pathology and IBSAL, University Hospital of Salamanca, Salamanca, Spain
- CIBERONC, Madrid, Spain
| | - Antonio Barbáchano
- CIBERONC, Madrid, Spain
- Instituto de Investigaciones Biomédicas 'Alberto Sols', CSIC-Autonomous University of Madrid, and Instituto de Investigación Sanitaria del Hospital Universitario La Paz, Madrid, Spain
| | - Asunción Fernández-Barral
- CIBERONC, Madrid, Spain
- Instituto de Investigaciones Biomédicas 'Alberto Sols', CSIC-Autonomous University of Madrid, and Instituto de Investigación Sanitaria del Hospital Universitario La Paz, Madrid, Spain
| | - Alberto Muñoz
- CIBERONC, Madrid, Spain
- Instituto de Investigaciones Biomédicas 'Alberto Sols', CSIC-Autonomous University of Madrid, and Instituto de Investigación Sanitaria del Hospital Universitario La Paz, Madrid, Spain
| | - Atanasio Pandiella
- Institute of Biomedical Research of Salamanca (IBSAL), Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca), Salamanca, Spain.
- CIBERONC, Madrid, Spain.
| |
Collapse
|
4
|
Sánchez-Fdez A, Matilla-Almazán S, Del Carmen S, Abad M, Arconada-Luque E, Jiménez-Suárez J, Chinchilla-Tábora LM, Ruíz-Hidalgo MJ, Sánchez-Prieto R, Pandiella A, Esparís-Ogando A. Etiopathogenic role of ERK5 signaling in sarcoma: prognostic and therapeutic implications. Exp Mol Med 2023; 55:1247-1257. [PMID: 37332046 PMCID: PMC10317974 DOI: 10.1038/s12276-023-01008-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 06/20/2023] Open
Abstract
Sarcomas constitute a heterogeneous group of rare and difficult-to-treat tumors that can affect people of all ages, representing one of the most common forms of cancer in childhood and adolescence. Little is known about the molecular entities involved in sarcomagenesis. Therefore, the identification of processes that lead to the development of the disease may uncover novel therapeutic opportunities. Here, we show that the MEK5/ERK5 signaling pathway plays a critical role in the pathogenesis of sarcomas. By developing a mouse model engineered to express a constitutively active form of MEK5, we demonstrate that the exclusive activation of the MEK5/ERK5 pathway can promote sarcomagenesis. Histopathological analyses identified these tumors as undifferentiated pleomorphic sarcomas. Bioinformatic studies revealed that sarcomas are the tumors in which ERK5 is most frequently amplified and overexpressed. Moreover, analysis of the impact of ERK5 protein expression on overall survival in patients diagnosed with different sarcoma types in our local hospital showed a 5-fold decrease in median survival in patients with elevated ERK5 expression compared with those with low expression. Pharmacological and genetic studies revealed that targeting the MEK5/ERK5 pathway drastically affects the proliferation of human sarcoma cells and tumor growth. Interestingly, sarcoma cells with knockout of ERK5 or MEK5 were unable to form tumors when engrafted into mice. Taken together, our results reveal a role of the MEK5/ERK5 pathway in sarcomagenesis and open a new scenario to be considered in the treatment of patients with sarcoma in which the ERK5 pathway is pathophysiologically involved.
Collapse
Affiliation(s)
- Adrián Sánchez-Fdez
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC)-CSIC, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-Universidad de Salamanca, Salamanca, Spain
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Sofía Matilla-Almazán
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC)-CSIC, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-Universidad de Salamanca, Salamanca, Spain
| | - Sofía Del Carmen
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departmento de Patología, Hospital Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain
| | - Mar Abad
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departmento de Patología, Hospital Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain
| | - Elena Arconada-Luque
- Universidad de Castilla-La Mancha, Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad asociada al CSIC, Albacete, Spain
| | - Jaime Jiménez-Suárez
- Universidad de Castilla-La Mancha, Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad asociada al CSIC, Albacete, Spain
| | - Luis Miguel Chinchilla-Tábora
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departmento de Patología, Hospital Universitario de Salamanca, Universidad de Salamanca, Salamanca, Spain
| | - Mª José Ruíz-Hidalgo
- Universidad de Castilla-La Mancha, Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad asociada al CSIC, Albacete, Spain
- Universidad de Castilla-La Mancha, Departamento de Química Inorgánica, Orgánica y Bioquímica, Área de Bioquímica y Biología Molecular. Facultad de Medicina, Albacete, Spain
| | - Ricardo Sánchez-Prieto
- Universidad de Castilla-La Mancha, Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad asociada al CSIC, Albacete, Spain
- Universidad de Castilla-La Mancha, Departamento de Ciencias Médicas, Facultad de Medicina, Albacete, Spain
- Departamento de Biología del Cáncer, Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Madrid, Spain
- Instituto de Investigaciones Biomédicas 'Alberto Sols', Consejo Superior de Investigaciones Científicas (IIBM-CSIC)-Universidad de Castilla-La Mancha (UCLM), Albacete, Spain
| | - Atanasio Pandiella
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC)-CSIC, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-Universidad de Salamanca, Salamanca, Spain
| | - Azucena Esparís-Ogando
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC)-CSIC, Salamanca, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-Universidad de Salamanca, Salamanca, Spain.
| |
Collapse
|
5
|
Neuregulin modulates hormone receptor levels in breast cancer through concerted action on multiple signaling pathways. Clin Sci (Lond) 2023; 137:1-15. [PMID: 36511917 PMCID: PMC9805957 DOI: 10.1042/cs20220472] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/05/2022] [Accepted: 12/13/2022] [Indexed: 12/15/2022]
Abstract
The Neuregulins (NRGs) are growth factors that bind and activate ErbB/HER receptor tyrosine kinases. Some reports have described an interplay between this ligand-receptor system and hormonal receptors in breast cancer. However, the mechanisms by which NRGs regulate hormonal receptor signaling have not been sufficiently described. Here, we show that in breast cancer cells the activation of NRG receptors down-regulated ERα through a double mechanism that included post-transcriptional and transcriptional effects. This regulation required the concerted participation of three signaling routes: the PI3K/AKT/mTOR, ERK1/2, and ERK5 pathways. Moreover, these three routes were also involved in the phosphorylation of ERα at serines 118 and 167, two residues implicated in resistance to endocrine therapies. On the other hand, NRGs conferred resistance to fulvestrant in breast cancer cells and this resistance could be reversed when the three pathways activated by NRGs were simultaneously inhibited. Our results indicate that estrogen receptor-positive (ER+) breast tumors that can have access to NRGs may be resistant to fulvestrant. This resistance could be overcome if strategies to target the three main pathways involved in the interplay between NRG receptors and ERα could be developed.
Collapse
|
6
|
You I, Donovan KA, Krupnick NM, Boghossian AS, Rees MG, Ronan MM, Roth JA, Fischer ES, Wang ES, Gray NS. Acute pharmacological degradation of ERK5 does not inhibit cellular immune response or proliferation. Cell Chem Biol 2022; 29:1630-1638.e7. [PMID: 36220104 PMCID: PMC9675722 DOI: 10.1016/j.chembiol.2022.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/26/2022] [Accepted: 09/17/2022] [Indexed: 01/31/2023]
Abstract
Recent interest in the role that extracellular signal-regulated kinase 5 (ERK5) plays in various diseases, particularly cancer and inflammation, has grown. Phenotypes observed from genetic knockdown or deletion of ERK5 suggested that targeting ERK5 could have therapeutic potential in various disease settings, motivating the development ATP-competitive ERK5 inhibitors. However, these inhibitors were unable to recapitulate the effects of genetic loss of ERK5, suggesting that ERK5 may have key kinase-independent roles. To investigate potential non-catalytic functions of ERK5, we report the development of INY-06-061, a potent and selective heterobifunctional degrader of ERK5. In contrast to results reported through genetic knockdown of ERK5, INY-06-061-induced ERK5 degradation did not induce anti-proliferative effects in multiple cancer cell lines or suppress inflammatory responses in primary endothelial cells. Thus, we developed and characterized a chemical tool useful for validating phenotypes reported to be associated with genetic ERK5 ablation and for guiding future ERK5-directed drug discovery efforts.
Collapse
Affiliation(s)
- Inchul You
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Katherine A Donovan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Noah M Krupnick
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | - Matthew G Rees
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Melissa M Ronan
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jennifer A Roth
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Eric S Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Eric S Wang
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| | - Nathanael S Gray
- Department of Chemical and Systems Biology, Chem-H and Stanford Cancer Institute, Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
7
|
The ERK5/NF-κB signaling pathway targets endometrial cancer proliferation and survival. Cell Mol Life Sci 2022; 79:524. [PMID: 36123565 PMCID: PMC9485191 DOI: 10.1007/s00018-022-04541-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/20/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022]
Abstract
Endometrial cancer (EC) is the most common type of gynecologic cancer in women of developed countries. Despite surgery combined with chemo-/radiotherapy regimens, overall survival of patients with high-risk EC tumors is poor, indicating a need for novel therapies. The MEK5-ERK5 pathway is activated in response to growth factors and to different stressors, including oxidative stress and cytokines. Previous evidence supports a role for the MEK5-ERK5 pathway in the pathology of several cancers. We investigated the role of ERK5 in EC. In silico analysis of the PanCancer Atlas dataset showed alterations in components of the MEK5-ERK5 pathway in 48% of EC patients. Here, we show that ERK5 inhibition or silencing decreased EGF-induced EC cell proliferation, and that genetic deletion of MEK5 resulted in EC impaired proliferation and reduced tumor growth capacity in nude mice. Pharmacologic inhibition or ERK5 silencing impaired NF-kB pathway in EC cells and xenografts. Furthermore, we found a positive correlation between ERK5 and p65/RELA protein levels in human EC tumor samples. Mechanistically, genetic or pharmacologic impairment of ERK5 resulted in downregulation of NEMO/IKKγ expression, leading to impaired p65/RELA activity and to apoptosis in EC cells and xenografts, which was rescued by NEMO/IKKγ overexpression. Notably, ERK5 inhibition, MEK5 deletion or NF-kB inhibition sensitized EC cells to standard EC chemotherapy (paclitaxel/carboplatin) toxicity, whereas ERK5 inhibition synergized with paclitaxel to reduce tumor xenograft growth in mice. Together, our results suggest that the ERK5-NEMO-NF-κB pathway mediates EC cell proliferation and survival. We propose the ERK5/NF-κB axis as new target for EC treatment.
Collapse
|
8
|
Arconada-Luque E, Jiménez-Suarez J, Pascual-Serra R, Nam-Cha SH, Moline T, Cimas FJ, Fliquete G, Ortega-Muelas M, Roche O, Fernández-Aroca DM, Muñoz Velasco R, García-Flores N, Garnés-García C, Sánchez-Fdez A, Matilla-Almazán S, Sánchez-Arévalo Lobo VJ, Hernández-Losa J, Belandia B, Pandiella A, Esparís-Ogando A, Ramón y Cajal S, del Peso L, Sánchez-Prieto R, Ruiz-Hidalgo MJ. ERK5 Is a Major Determinant of Chemical Sarcomagenesis: Implications in Human Pathology. Cancers (Basel) 2022; 14:cancers14143509. [PMID: 35884568 PMCID: PMC9316148 DOI: 10.3390/cancers14143509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/11/2022] [Accepted: 07/16/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Sarcoma is a heterogeneous group of tumors poorly studied with few therapeutic opportunities. Interestingly, the role of MAPKs still remains unclear in sarcomatous pathology. Here, we describe for the first time the critical role of ERK5 in the biology of soft tissue sarcoma by using in vitro and in vivo approaches in a murine experimental model of chemical sarcomagenesis. Indeed, our observations were extrapolated to a short series of human leiomyosarcoma and rhabdomyosarcomas. Furthermore, transcriptome analysis allows us to demonstrate the critical role of KLF2 in the biological effects of ERK5. Therefore, the data presented here open new windows in the diagnosis and therapy of soft tissue sarcomas. Abstract Sarcomas are a heterogeneous group of tumors in which the role of ERK5 is poorly studied. To clarify the role of this MAPK in sarcomatous pathology, we used a murine 3-methyl-cholanthrene (3MC)-induced sarcoma model. Our data show that 3MC induces pleomorphic sarcomas with muscle differentiation, showing an increased expression of ERK5. Indeed, this upregulation was also observed in human sarcomas of muscular origin, such as leiomyosarcoma or rhabdomyosarcoma. Moreover, in cell lines derived from these 3MC-induced tumors, abrogation of Mapk7 expression by using specific shRNAs decreased in vitro growth and colony-forming capacity and led to a marked loss of tumor growth in vivo. In fact, transcriptomic profiling in ERK5 abrogated cell lines by RNAseq showed a deregulated gene expression pattern for key biological processes such as angiogenesis, migration, motility, etc., correlating with a better prognostic in human pathology. Finally, among the various differentially expressed genes, Klf2 is a key mediator of the biological effects of ERK5 as indicated by its specific interference, demonstrating that the ERK5–KLF2 axis is an important determinant of sarcoma biology that should be further studied in human pathology.
Collapse
Affiliation(s)
- Elena Arconada-Luque
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
| | - Jaime Jiménez-Suarez
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
| | - Raquel Pascual-Serra
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
| | - Syong Hyun Nam-Cha
- Servicio de Anatomía Patológica, Hospital General de Albacete, 02008 Albacete, Spain;
| | - Teresa Moline
- Grupo de Patología Molecular Traslacional, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona Centro de Investigación Biomédica en RED de Cancer CIBERONC, 08035 Barcelona, Spain; (T.M.); (G.F.); (J.H.-L.); (S.R.y.C.)
| | - Francisco J. Cimas
- Unidad de Bioquímica y Biología Molecular, Servicio de Instrumentación Biomédica, Universidad de Castilla-La Mancha, 02008 Albacete, Spain;
| | - Germán Fliquete
- Grupo de Patología Molecular Traslacional, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona Centro de Investigación Biomédica en RED de Cancer CIBERONC, 08035 Barcelona, Spain; (T.M.); (G.F.); (J.H.-L.); (S.R.y.C.)
| | - Marta Ortega-Muelas
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
| | - Olga Roche
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
- Departamento de Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| | - Diego M. Fernández-Aroca
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
| | - Raúl Muñoz Velasco
- Grupo de Oncología Molecular, Facultad de Ciencias Experimentales, Instituto de Investigación Biosanitaria, Universidad Francisco de Vitoria, Pozuelo de Alarcón, 28223 Madrid, Spain; (R.M.V.); (V.J.S.-A.L.)
- Departamento de Anatomía Patológica, Instituto de Investigación Hospital 12 de Octubre, Av. Córdoba, s/n, 28041 Madrid, Spain
| | - Natalia García-Flores
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
| | - Cristina Garnés-García
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
| | - Adrián Sánchez-Fdez
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, 37007 Salamanca, Spain; (A.S.-F.); (S.M.-A.); (A.P.); (A.E.-O.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, CSIC, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en RED de Cancer CIBERONC, 37007 Salamanca, Spain
| | - Sofía Matilla-Almazán
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, 37007 Salamanca, Spain; (A.S.-F.); (S.M.-A.); (A.P.); (A.E.-O.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, CSIC, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en RED de Cancer CIBERONC, 37007 Salamanca, Spain
| | - Víctor J. Sánchez-Arévalo Lobo
- Grupo de Oncología Molecular, Facultad de Ciencias Experimentales, Instituto de Investigación Biosanitaria, Universidad Francisco de Vitoria, Pozuelo de Alarcón, 28223 Madrid, Spain; (R.M.V.); (V.J.S.-A.L.)
- Departamento de Anatomía Patológica, Instituto de Investigación Hospital 12 de Octubre, Av. Córdoba, s/n, 28041 Madrid, Spain
| | - Javier Hernández-Losa
- Grupo de Patología Molecular Traslacional, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona Centro de Investigación Biomédica en RED de Cancer CIBERONC, 08035 Barcelona, Spain; (T.M.); (G.F.); (J.H.-L.); (S.R.y.C.)
| | - Borja Belandia
- Departamento de Biología del Cáncer, Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, 28029 Madrid, Spain;
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, 37007 Salamanca, Spain; (A.S.-F.); (S.M.-A.); (A.P.); (A.E.-O.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, CSIC, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en RED de Cancer CIBERONC, 37007 Salamanca, Spain
| | - Azucena Esparís-Ogando
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, 37007 Salamanca, Spain; (A.S.-F.); (S.M.-A.); (A.P.); (A.E.-O.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, Universidad de Salamanca, CSIC, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en RED de Cancer CIBERONC, 37007 Salamanca, Spain
| | - Santiago Ramón y Cajal
- Grupo de Patología Molecular Traslacional, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona Centro de Investigación Biomédica en RED de Cancer CIBERONC, 08035 Barcelona, Spain; (T.M.); (G.F.); (J.H.-L.); (S.R.y.C.)
| | - Luis del Peso
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM) and Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), 28029 Madrid, Spain;
- Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias CIBERES, 28029 Madrid, Spain
| | - Ricardo Sánchez-Prieto
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
- Departamento de Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Departamento de Biología del Cáncer, Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, 28029 Madrid, Spain;
- Instituto de Investigaciones Biomédicas ‘Alberto Sols’, Consejo Superior de Investigaciones Científicas (IIBM-CSIC)-Universidad de Castilla-La Mancha, 02008 Albacete, Spain
- Correspondence:
| | - María José Ruiz-Hidalgo
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, Unidad Asociada de Biomedicina UCLM, Unidad Asociada al CSIC, Universidad de Castilla-La Mancha, 02008 Albacete, Spain; (E.A.-L.); (J.J.-S.); (R.P.-S.); (M.O.-M.); (O.R.); (D.M.F.-A.); (N.G.-F.); (C.G.-G.); (M.J.R.-H.)
- Departamento de Química Inorgánica, Orgánica y Bioquímica, Área de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Castilla-La Mancha, 02008 Albacete, Spain
| |
Collapse
|
9
|
Montero JC, Calvo-Jiménez E, Del Carmen S, Abad M, Ocaña A, Pandiella A. Surfaceome analyses uncover CD98hc as an antibody drug-conjugate target in triple negative breast cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:106. [PMID: 35317825 PMCID: PMC8941813 DOI: 10.1186/s13046-022-02330-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/12/2022] [Indexed: 11/10/2022]
Abstract
Background Despite the incorporation of novel therapeutics, advanced triple negative breast cancer (TNBC) still represents a relevant clinical problem. Considering this, as well as the clinical efficacy of antibody-drug conjugates (ADCs), we aimed at identifying novel ADC targets that could be used to treat TNBC. Methods Transcriptomic analyses were performed on TNBC and normal samples from three different studies. Plasma membrane proteins of three cell lines representative of the TNBC subtype were identified by cell surface biotinylation or plasma membrane isolation, followed by analyses of cell surface proteins using the Surfaceome online tool. Immunofluorescence and western studies were used to characterize the action of a CD98hc-directed ADC, which was prepared by in house coupling of emtansine to an antibody that recognized the ectodomain of CD98hc. Xenografted TNBC cells were used to analyze the antitumoral properties of the anti-CD98hc ADC. Results Comparative genomic studies between normal breast and TNBC tissues, together with proteomic and bioinformatic analyses resulted in the elaboration of a catalog of potential ADC targets. One of them, the CD98hc transmembrane protein, was validated as an ADC target. An antibody recognizing the ectodomain of CD98hc efficiently internalized and reached the lysosomal compartment. An emtansine-based ADC derived from such antibody was prepared and showed antitumoral properties in TNBC in vitro and in vivo models. Mechanistically, the anti-CD98hc ADC blocked cell cycle progression, that was followed by cell death caused by mitotic catastrophe. Conclusions This work describes a list of potential ADC targets in TNBC and validates one of them, the transmembrane protein CD98hc. The studies presented here also demonstrate the robustness of the multiomic approach herewith described to identify novel potential ADC targets. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02330-4.
Collapse
Affiliation(s)
- Juan Carlos Montero
- Institute of Biomedical Research of Salamanca (IBSAL), Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Salamanca, Spain. .,Department of Pathology and IBSAL, University Hospital of Salamanca, University of Salamanca, 37007, Salamanca, Spain.
| | - Elisa Calvo-Jiménez
- Institute of Biomedical Research of Salamanca (IBSAL), Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Salamanca, Spain
| | - Sofía Del Carmen
- Department of Pathology and IBSAL, University Hospital of Salamanca, University of Salamanca, 37007, Salamanca, Spain
| | - Mar Abad
- Department of Pathology and IBSAL, University Hospital of Salamanca, University of Salamanca, 37007, Salamanca, Spain
| | | | - Atanasio Pandiella
- Institute of Biomedical Research of Salamanca (IBSAL), Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Salamanca, Spain
| |
Collapse
|
10
|
Zhang J, Pearson AJ, Sabherwal N, Telfer BA, Ali N, Kan K, Xu Q, Zhang W, Chen F, Li S, Wang J, Gray NS, Risa-Ebrí B, Finegan KG, Cross MJ, Giurisato E, Whitmarsh AJ, Tournier C. Inhibiting ERK5 overcomes breast cancer resistance to anti-HER2 therapy by targeting the G1/S cell cycle transition. CANCER RESEARCH COMMUNICATIONS 2022; 2:131-145. [PMID: 36466034 PMCID: PMC7613885 DOI: 10.1158/2767-9764.crc-21-0089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Targeting the human epidermal growth factor receptor 2 (HER2) became a landmark in the treatment of HER2-driven breast cancer. Nonetheless, the clinical efficacy of anti-HER2 therapies can be short-lived and a significant proportion of patients ultimately develop metastatic disease and die. One striking consequence of oncogenic activation of HER2 in breast cancer cells is the constitutive activation of the extracellular-regulated protein kinase 5 (ERK5) through its hyperphosphorylation. In this study, we sought to decipher the significance of this unique molecular signature in promoting therapeutic resistance to anti-HER2 agents. We found that a small-molecule inhibitor of ERK5 suppressed the phosphorylation of the retinoblastoma protein (RB) in HER2 positive breast cancer cells. As a result, ERK5 inhibition enhanced the anti-proliferative activity of single-agent anti-HER2 therapy in resistant breast cancer cell lines by causing a G1 cell cycle arrest. Moreover, ERK5 knockdown restored the anti-tumor activity of the anti-HER2 agent lapatinib in human breast cancer xenografts. Taken together, these findings support the therapeutic potential of ERK5 inhibitors to improve the clinical benefit that patients receive from targeted HER2 therapies.
Collapse
Affiliation(s)
- Jingwei Zhang
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health (FBMH), University of Manchester, UK
| | - Adam J Pearson
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health (FBMH), University of Manchester, UK
| | - Nitin Sabherwal
- Division of Developmental Biology and Medicine, School of Medical Sciences, FBMH, University of Manchester, UK
| | - Brian A Telfer
- Division of Pharmacy and Optometry, School of Health Sciences, FBMH, University of Manchester, UK
| | - Nisha Ali
- Manchester University NHS FT, Wythenshawe hospital, UK
| | - Karmern Kan
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health (FBMH), University of Manchester, UK
| | - Qiuping Xu
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health (FBMH), University of Manchester, UK
| | - Wei Zhang
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health (FBMH), University of Manchester, UK
| | - Fuhui Chen
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health (FBMH), University of Manchester, UK
| | - Shiyang Li
- Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, FBMH, University of Manchester, UK
| | - Jinhua Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, USA
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, USA
| | - Blanca Risa-Ebrí
- Division of Pharmacy and Optometry, School of Health Sciences, FBMH, University of Manchester, UK
| | - Katherine G Finegan
- Division of Pharmacy and Optometry, School of Health Sciences, FBMH, University of Manchester, UK
| | - Michael J Cross
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology (ISMIB), University of Liverpool, UK
| | - Emanuele Giurisato
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health (FBMH), University of Manchester, UK,Department of Biotechnology Chemistry and Pharmacy, University of Siena, Italy
| | - Alan J Whitmarsh
- Division of Molecular and Cellular Function, School of Biological Sciences, FBMH, University of Manchester, UK
| | - Cathy Tournier
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health (FBMH), University of Manchester, UK,Corresponding author: Cathy Tournier, University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK, Tel: +44 161 275 5417,
| |
Collapse
|
11
|
Díaz-Gil L, Brasó-Maristany F, Locatelli C, Centa A, Győrffy B, Ocaña A, Prat A, Pandiella A. Modelling hypersensitivity to trastuzumab defines biomarkers of response in HER2 positive breast cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:313. [PMID: 34620206 PMCID: PMC8496101 DOI: 10.1186/s13046-021-02098-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/07/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND Trastuzumab-based therapies are the therapeutic option for HER2 positive (HER2+) breast cancer. HER2 amplification is the only biomarker validated for trastuzumab-based therapies. However, a proportion of tumors become refractory during treatment course. For this reason, the finding of new biomarkers beyond HER2 overexpression to identify patients who would benefit most from trastuzumab regimens is of outstanding importance. METHODS Models of trastuzumab-resistant or hypersensitive cells were generated by exposure to trastuzumab. Cell surface, total HER2, and analyses of proteins involved in cell cycle or apoptosis were analyzed by western blotting. Cell proliferation was analyzed by cell counting, cell cycle and apoptosis was evaluated by FACS. Transcriptomic characterization of the cellular models was performed using bioinformatic online tools, and clinico-genomic analyses were performed using the PAMELA clinical trial data. RESULTS Besides differing in sensitivities to trastuzumab, the different cellular models also showed distinct response to other anti-HER2 drugs (lapatinib, neratinib, pertuzumab and T-DM1) used in the clinic. That differential effect was not due to changes in cell surface, total or activated HER2. Trastuzumab caused important induction of cell death in hypersensitive cells but not in parental or resistant cells. Transcriptomic analyses of these cellular models together with querying of online databases allowed the identification of individual genes and gene signatures that predicted prognosis and trastuzumab response in HER2+ breast cancer patients. CONCLUSION The identification of trastuzumab response biomarkers may be used to select patients particularly sensitive to facilitate the use of trastuzumab-based therapies and refine follow-up guidelines in patients with HER2+ tumors.
Collapse
Affiliation(s)
- Laura Díaz-Gil
- Instituto de Biología Molecular y Celular del Cáncer, CSIC and CIBERONC, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Fara Brasó-Maristany
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.,Department of Medical Oncology, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Claudriana Locatelli
- Programa de Pós-Graduação em Desenvolvimento e Sociedade, Universidade Alto Vale do Rio do Peixe - UNIARP, Caçador, SC, Brazil
| | - Ariana Centa
- Instituto de Biología Molecular y Celular del Cáncer, CSIC and CIBERONC, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Programa de Pós-Graduação em Desenvolvimento e Sociedade, Universidade Alto Vale do Rio do Peixe - UNIARP, Caçador, SC, Brazil
| | - Balász Győrffy
- Department of Bioinformatics and 2nd Department of Pediatrics, Semmelweis University and TTK Cancer Biomarker Research Group, Budapest, Hungary
| | | | - Aleix Prat
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.,Department of Medical Oncology, Hospital Clínic of Barcelona, Barcelona, Spain.,SOLTI cooperative group, Barcelona, Spain.,Department of Oncology, Quironsalud Group, IOB Institute of Oncology, Barcelona, Spain.,Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer, CSIC and CIBERONC, Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.
| |
Collapse
|
12
|
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
|
13
|
Sánchez-Fdez A, Re-Louhau MF, Rodríguez-Núñez P, Ludeña D, Matilla-Almazán S, Pandiella A, Esparís-Ogando A. Clinical, genetic and pharmacological data support targeting the MEK5/ERK5 module in lung cancer. NPJ Precis Oncol 2021; 5:78. [PMID: 34404896 PMCID: PMC8371118 DOI: 10.1038/s41698-021-00218-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 07/20/2021] [Indexed: 11/09/2022] Open
Abstract
Despite advances in its treatment, lung cancer still represents the most common and lethal tumor. Because of that, efforts to decipher the pathophysiological actors that may promote lung tumor generation/progression are being made, with the final aim of establishing new therapeutic options. Using a transgenic mouse model, we formerly demonstrated that the sole activation of the MEK5/ERK5 MAPK route had a pathophysiological role in the onset of lung adenocarcinomas. Given the prevalence of that disease and its frequent dismal prognosis, our findings opened the possibility of targeting the MEK5/ERK5 route with therapeutic purposes. Here we have explored such possibility. We found that increased levels of MEK5/ERK5 correlated with poor patient prognosis in lung cancer. Moreover, using genetic as well as pharmacological tools, we show that targeting the MEK5/ERK5 route is therapeutically effective in lung cancer. Not only genetic disruption of ERK5 by CRISPR/Cas9 caused a relevant inhibition of tumor growth in vitro and in vivo; such ERK5 deficit augmented the antitumoral effect of agents normally used in the lung cancer clinic. The clinical correlation studies together with the pharmacological and genetic results establish the basis for considering the targeting of the MEK5/ERK5 route in the therapy for lung cancer.
Collapse
Affiliation(s)
- Adrián Sánchez-Fdez
- Institute of Molecular and Cellular Biology of Cancer (IBMCC)-CSIC, Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Cancer Network Research (CIBERONC), Salamanca, Spain
| | - María Florencia Re-Louhau
- Institute of Molecular and Cellular Biology of Cancer (IBMCC)-CSIC, Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Pablo Rodríguez-Núñez
- Institute of Molecular and Cellular Biology of Cancer (IBMCC)-CSIC, Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
| | - Dolores Ludeña
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Pathology Service, University Hospital, Salamanca, Spain
| | - Sofía Matilla-Almazán
- Institute of Molecular and Cellular Biology of Cancer (IBMCC)-CSIC, Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Cancer Network Research (CIBERONC), Salamanca, Spain
| | - Atanasio Pandiella
- Institute of Molecular and Cellular Biology of Cancer (IBMCC)-CSIC, Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Cancer Network Research (CIBERONC), Salamanca, Spain
| | - Azucena Esparís-Ogando
- Institute of Molecular and Cellular Biology of Cancer (IBMCC)-CSIC, Salamanca, Spain. .,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain. .,Cancer Network Research (CIBERONC), Salamanca, Spain.
| |
Collapse
|
14
|
Xu Q, Zhang J, Telfer BA, Zhang H, Ali N, Chen F, Risa B, Pearson AJ, Zhang W, Finegan KG, Ucar A, Giurisato E, Tournier C. The extracellular-regulated protein kinase 5 (ERK5) enhances metastatic burden in triple-negative breast cancer through focal adhesion protein kinase (FAK)-mediated regulation of cell adhesion. Oncogene 2021; 40:3929-3941. [PMID: 33981002 PMCID: PMC8195737 DOI: 10.1038/s41388-021-01798-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 03/23/2021] [Accepted: 04/14/2021] [Indexed: 12/22/2022]
Abstract
There is overwhelming clinical evidence that the extracellular-regulated protein kinase 5 (ERK5) is significantly dysregulated in human breast cancer. However, there is no definite understanding of the requirement of ERK5 in tumor growth and metastasis due to very limited characterization of the pathway in disease models. In this study, we report that a high level of ERK5 is a predictive marker of metastatic breast cancer. Mechanistically, our in vitro data revealed that ERK5 was critical for maintaining the invasive capability of triple-negative breast cancer (TNBC) cells through focal adhesion protein kinase (FAK) activation. Specifically, we found that phosphorylation of FAK at Tyr397 was controlled by a kinase-independent function of ERK5. Accordingly, silencing ERK5 in mammary tumor grafts impaired FAK phosphorylation at Tyr397 and suppressed TNBC cell metastasis to the lung without preventing tumor growth. Collectively, these results establish a functional relationship between ERK5 and FAK signaling in promoting malignancy. Thus, targeting the oncogenic ERK5-FAK axis represents a promising therapeutic strategy for breast cancer exhibiting aggressive clinical behavior.
Collapse
Affiliation(s)
- Qiuping Xu
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jingwei Zhang
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Brian A Telfer
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Hao Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Nisha Ali
- Manchester University NHS FT, Wythenshawe hospital, Manchester, UK
| | - Fuhui Chen
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Blanca Risa
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Adam J Pearson
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Wei Zhang
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Katherine G Finegan
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Ahmet Ucar
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Emanuele Giurisato
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Department of Biotechnology Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Cathy Tournier
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
15
|
Montero JC, Pandiella A. PDCD4 limits prooncogenic neuregulin-ErbB signaling. Cell Mol Life Sci 2021; 78:1799-1815. [PMID: 32804243 PMCID: PMC11073242 DOI: 10.1007/s00018-020-03617-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 07/21/2020] [Accepted: 08/07/2020] [Indexed: 11/26/2022]
Abstract
The neuregulins and their ErbB/HER receptors play essential roles in mammalian development and tissue homeostasis. In addition, deregulation of their function has been linked to the pathogenesis of diseases such as cancer or schizophrenia. These circumstances have stimulated research into the biology of this ligand-receptor system. Here we show the identification of programmed cell death protein-4 (PDCD4) as a novel neuregulin-ErbB signaling mediator. Phosphoproteomic analyses identified PDCD4 as protein whose phosphorylation increased in cells treated with neuregulin. Mutagenesis experiments defined serine 67 of PDCD4 as a site whose phosphorylation increased upon activation of neuregulin receptors. Phosphorylation of that site promoted degradation of PDCD4 by the proteasome, which depended on exit of PDCD4 from the nucleus to the cytosol. Mechanistic studies defined mTORC1 and ERK1/2 as routes implicated in neuregulin-induced serine 67 phosphorylation and PDCD4 degradation. Functionally, PDCD4 regulated several important biological functions of neuregulin, such as proliferation, migration, or invasion.
Collapse
Affiliation(s)
- Juan Carlos Montero
- Institute of Biomedical Research of Salamanca (IBSAL), Instituto de Biología Molecular y Celular del Cáncer (CSIC) and CIBERONC, Campus Miguel de Unamuno, 37007, Salamanca, Spain.
| | - Atanasio Pandiella
- Institute of Biomedical Research of Salamanca (IBSAL), Instituto de Biología Molecular y Celular del Cáncer (CSIC) and CIBERONC, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| |
Collapse
|
16
|
Malavia N, Kuche K, Ghadi R, Jain S. A bird's eye view of the advanced approaches and strategies for overshadowing triple negative breast cancer. J Control Release 2020; 330:72-100. [PMID: 33321156 DOI: 10.1016/j.jconrel.2020.12.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022]
Abstract
Triple negative breast cancer (TNBC) is one of the most aggressive form of breast cancer. It is characterized by the absence of estrogen, progesterone and human epidermal growth factor receptors. The main issue with TNBC is that it exhibits poor prognosis, high risk of relapse, short progression-free survival and low overall survival in patients. This is because the conventional therapy used for managing TNBC has issues pertaining to poor bioavailability, lower cellular uptake, increased off-target effects and development of resistance. To overcome such pitfalls, several other approaches are explored. In this context, the present manuscript showcases three of the most widely used approaches which are (i) nanotechnology-based approach; (ii) gene therapy approach and (iii) Phytochemical-based approach. The ultimate focus is to present and explain the insightful reports based on these approaches. Further, the review also expounds on the identified molecular targets and novel targeting ligands which are explored for managing TNBC effectively. Thus, in a nutshell, the review tries to highlight these existing treatment approaches which might inspire for future development of novel therapies with a potential of overshadowing TNBC.
Collapse
Affiliation(s)
- Nilesh Malavia
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, India
| | - Kaushik Kuche
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, India
| | - Rohan Ghadi
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, India
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, India.
| |
Collapse
|
17
|
Efficient Suppression of NRAS-Driven Melanoma by Co-Inhibition of ERK1/2 and ERK5 MAPK Pathways. J Invest Dermatol 2020; 140:2455-2465.e10. [PMID: 32376279 DOI: 10.1016/j.jid.2020.03.972] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/24/2020] [Accepted: 03/18/2020] [Indexed: 12/19/2022]
Abstract
Cutaneous melanoma is a highly malignant tumor typically driven by somatic mutation in the oncogenes BRAF or NRAS, leading to uncontrolled activation of the MEK/ERK MAPK pathway. Despite the availability of immunotherapy, MAPK pathway‒targeting regimens are still a valuable treatment option for BRAF-mutant melanoma. Unfortunately, patients with NRAS mutation do not benefit from such therapies owing to the lack of targetable BRAF mutations and a high degree of intrinsic and acquired resistance toward MEK inhibition. Here, we demonstrate that concomitant inhibition of ERK5 removes this constraint and effectively sensitizes NRAS-mutant melanoma cells for MAPK pathway‒targeting therapy. Using approved MEK inhibitors or a pharmacologic ERK inhibitor, we demonstrate that MAPK inhibition triggers a delayed activation of ERK5 through a PDGFR inhibitor-sensitive pathway in NRAS-mutant melanoma cells, resulting in sustained proliferation and survival. ERK5 phosphorylation also occurred naturally in NRAS-mutant melanoma cells and correlated with nuclear localization of its stem cell-associated effector KLF2. Importantly, MEK/ERK5 co-inhibition prevented long-term growth of human NRAS-mutant melanoma cells in vitro and effectively repressed tumor progression in a xenotransplant mouse model. Our findings suggest MEK/ERK5 cotargeting as a potential treatment option for NRAS-mutant melanoma, which currently is not amenable for targeted therapies.
Collapse
|
18
|
Targeted Avenues for Cancer Treatment: The MEK5-ERK5 Signaling Pathway. Trends Mol Med 2020; 26:394-407. [PMID: 32277933 DOI: 10.1016/j.molmed.2020.01.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/20/2019] [Accepted: 01/21/2020] [Indexed: 12/13/2022]
Abstract
Twenty years have passed since extracellular signal-regulated kinase 5 (ERK5) and its upstream activator, mitogen-activated protein kinase 5 (MEK5), first emerged onto the cancer research scene. Although we have come a long way in defining the liaison between dysregulated MEK5-ERK5 signaling and the pathogenesis of epithelial and nonepithelial malignancies, selective targeting of this unique pathway remains elusive. Here, we provide an updated review of the existing evidence for a correlation between aberrant MEK5-ERK5 (phospho)proteomic/transcriptomic profiles, aggressive cancer states, and poor patient outcomes. We then focus on emerging insights from preclinical models regarding the relevance of upregulated ERK5 activity in promoting tumor growth, metastasis, therapy resistance, undifferentiated traits, and immunosuppression, highlighting the opportunities, prospects, and challenges of selectively blocking this cascade for antineoplastic treatment and chemosensitization.
Collapse
|
19
|
Pearson AJ, Fullwood P, Toro Tapia G, Prise I, Smith MP, Xu Q, Jordan A, Giurisato E, Whitmarsh AJ, Francavilla C, Tournier C. Discovery of a Gatekeeper Residue in the C-Terminal Tail of the Extracellular Signal-Regulated Protein Kinase 5 (ERK5). Int J Mol Sci 2020; 21:E929. [PMID: 32023819 PMCID: PMC7037328 DOI: 10.3390/ijms21030929] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/23/2020] [Accepted: 01/29/2020] [Indexed: 01/17/2023] Open
Abstract
The extracellular signal-regulated protein kinase 5 (ERK5) is a non-redundant mitogen-activated protein kinase (MAPK) that exhibits a unique C-terminal extension which comprises distinct structural and functional properties. Here, we sought to elucidate the significance of phosphoacceptor sites in the C-terminal transactivation domain of ERK5. We have found that Thr732 acted as a functional gatekeeper residue controlling C-terminal-mediated nuclear translocation and transcriptional enhancement. Consistently, using a non-bias quantitative mass spectrometry approach, we demonstrated that phosphorylation at Thr732 conferred selectivity for binding interactions of ERK5 with proteins related to chromatin and RNA biology, whereas a number of metabolic regulators were associated with full-length wild type ERK5. Additionally, our proteomic analysis revealed that phosphorylation of the Ser730-Glu-Thr732-Pro motif could occur independently of dual phosphorylation at Thr218-Glu-Tyr220 in the activation loop. Collectively, our results firmly establish the significance of C-terminal phosphorylation in regulating ERK5 function. The post-translational modification of ERK5 on its C-terminal tail might be of particular relevance in cancer cells where ERK5 has be found to be hyperphosphoryated.
Collapse
Affiliation(s)
- Adam J. Pearson
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (A.J.P.); (Q.X.); (E.G.)
| | - Paul Fullwood
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (P.F.); (G.T.T.); (M.P.S.); (A.J.W.); (C.F.)
| | - Gabriela Toro Tapia
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (P.F.); (G.T.T.); (M.P.S.); (A.J.W.); (C.F.)
| | - Ian Prise
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK;
| | - Michael P. Smith
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (P.F.); (G.T.T.); (M.P.S.); (A.J.W.); (C.F.)
| | - Qiuping Xu
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (A.J.P.); (Q.X.); (E.G.)
| | - Allan Jordan
- Drug Discovery Unit, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M13 9PT, UK;
| | - Emanuele Giurisato
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (A.J.P.); (Q.X.); (E.G.)
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Alan J. Whitmarsh
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (P.F.); (G.T.T.); (M.P.S.); (A.J.W.); (C.F.)
| | - Chiara Francavilla
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (P.F.); (G.T.T.); (M.P.S.); (A.J.W.); (C.F.)
| | - Cathy Tournier
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (A.J.P.); (Q.X.); (E.G.)
| |
Collapse
|
20
|
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
|
21
|
Paclitaxel-Trastuzumab Mixed Nanovehicle to Target HER2-Overexpressing Tumors. NANOMATERIALS 2019; 9:nano9070948. [PMID: 31261957 PMCID: PMC6669497 DOI: 10.3390/nano9070948] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 06/26/2019] [Accepted: 06/28/2019] [Indexed: 12/13/2022]
Abstract
Paclitaxel is one of the most widely used chemotherapeutic agents thanks to its effectiveness and broad spectrum of antitumor activity. However, it has a very poor aqueous solubility and a limited specificity. To solve these handicaps, a novel paclitaxel-trastuzumab targeted transport nanosystem has been developed and characterized in this work to specifically treat cancer cells that overexpress the human epidermal growth factor receptor-2 (HER2). Methods: Alginate and piperazine nanoparticles were synthetized and conjugated with paclitaxel:β-cyclodextrins complexes and trastuzumab. Conjugated nanoparticles (300 nm) were characterized and their internalization in HER2-overexpressing tumor cells was analyzed by immunofluorescence. Its specific antitumor activity was studied in vitro using human cell lines with different levels of HER2-expression. Results: In comparison with free paclitaxel:β-cyclodextrins complexes, the developed conjugated nanovehicle presented specificity for the treatment of HER2-overpressing cells, in which it was internalized by endocytosis. Conclusions: It seems that potentially avoiding the conventional adverse effects of paclitaxel treatment could be possible with the use of the proposed mixed nanovehicle, which improves its bioavailability and targets HER2-positive cancer cells.
Collapse
|
22
|
Wu C, Chen M, Zhang Q, Yu L, Zhu J, Gao X. Genomic and GeneChip expression profiling reveals the inhibitory effects of Amorphophalli Rhizoma in TNBC cells. JOURNAL OF ETHNOPHARMACOLOGY 2019; 235:206-218. [PMID: 30731183 DOI: 10.1016/j.jep.2019.02.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/29/2019] [Accepted: 02/03/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Amorphophalli Rhizoma has been widely used as an adjuvant treatment for advanced or metastatic breast cancer, pancreatic cancer, hepatoma, and malignant lymphoma, but its molecular mechanism of action for treatment of metastatic triple-negative breast cancer (TNBC) is generally poorly understood. AIM OF THE STUDY To investigate genomic changes related to the inhibitory effect of Amorphophalli Rhizoma and to elucidate the molecular mechanism of this inhibition in MDA-MB-231 TNBC cells. MATERIALS AND METHODS Gene chip analysis was employed to explore genomic changes caused by Amorphophalli Rhizoma in TNBC cells. Potential classical signaling pathways, upstream regulators, functions, regulatory effects and gene interaction networks were analyzed by Ingenuity Pathway Analysis (IPA). Real-time quantitative PCR (RT-qPCR) and RNA interference (RNAi) assays were used to clarify the roles of potential target genes. RESULTS In total, 536 significantly upregulated and 648 significantly downregulated genes were identified between the group treated with Amorphophalli Rhizoma extract and that treated with vehicle. Many of these differentially expressed genes (DEGs) in TNBC cells are involved in DNA replication, recombination and repair, the cell cycle, and cellular assembly and organization. Attenuation of KNL1, OLFML2A, RTKN2 and SGO1 gene expression by Amorphophalli Rhizoma significantly induced cell cycle arrest and suppressed cell proliferation and migration. CONCLUSIONS The inhibitory effects of Amorphophalli Rhizoma in TNBC cells likely occur through regulation of the spindle checkpoint, chromosomal and centrosomal instability, and cell membrane stability.
Collapse
Affiliation(s)
- Chunyu Wu
- Department of Breast Surgery (Integrated Traditional and Western Medicine), Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai 200032, China
| | - Mingcang Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiuhua Zhang
- Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang 310053, China
| | - Linghong Yu
- Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang 310053, China
| | - Jiayan Zhu
- Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang 310053, China
| | - Xiufei Gao
- The First Affiliated Hospital of Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang 310006, China.
| |
Collapse
|
23
|
Díaz-Rodríguez E, Pérez-Peña J, Ríos-Luci C, Arribas J, Ocaña A, Pandiella A. TRAIL receptor activation overcomes resistance to trastuzumab in HER2 positive breast cancer cells. Cancer Lett 2019; 453:34-44. [PMID: 30928382 DOI: 10.1016/j.canlet.2019.03.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/20/2019] [Accepted: 03/22/2019] [Indexed: 12/22/2022]
Abstract
The appearance of resistance to the anti-HER2 targeted drug trastuzumab constitutes, nowadays, an important challenge in the oncology clinic. To fight such resistance, we searched for potential vulnerabilities in cells resistant to that drug. To that end, we used cell lines primary resistant to trastuzumab, as well as cells made secondarily resistant to the drug upon continuous exposure. Using genomic and proteomic approaches, a deregulation in cell death pathways was identified in trastuzumab-resistant cells. More precisely, an increased response to the death factor TRAIL, caused by an increase in the cellular receptors for this factor, was observed. In parallel, a decrease in inhibitory components of the pathway was detected. This combination produces a more efficient assembly of the functional complex in the trastuzumab-resistant cells that translates in the observed increased response to TRAIL. Analysis of HER2 positive patient samples confirmed deregulation of this pathway in trastuzumab-resistant patients. Taken together our data identify a vulnerability of trastuzumab-resistant cells that could be used to design new targeted therapies in that context.
Collapse
Affiliation(s)
- Elena Díaz-Rodríguez
- Instituto de Biología Molecular y Celular del Cáncer. CSIC-IBSAL, Salamanca, Spain; CIBERONC, Spain.
| | - Javier Pérez-Peña
- Instituto de Biología Molecular y Celular del Cáncer. CSIC-IBSAL, Salamanca, Spain; Translational Research Unit, Albacete University Hospital and Centro Regional de Investigaciones Biomédicas (CRIB), Universidad de Castilla la Mancha, Albacete, Spain, and; CIBERONC, Spain
| | - Carla Ríos-Luci
- Instituto de Biología Molecular y Celular del Cáncer. CSIC-IBSAL, Salamanca, Spain; CIBERONC, Spain
| | - Joaquín Arribas
- Vall d´Hebron Institute of Oncology, Barcelona, Spain; CIBERONC, Spain
| | - Alberto Ocaña
- Translational Research Unit, Albacete University Hospital and Centro Regional de Investigaciones Biomédicas (CRIB), Universidad de Castilla la Mancha, Albacete, Spain, and; CIBERONC, Spain
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer. CSIC-IBSAL, Salamanca, Spain; CIBERONC, Spain.
| |
Collapse
|
24
|
Benito-Jardón L, Díaz-Martínez M, Arellano-Sánchez N, Vaquero-Morales P, Esparís-Ogando A, Teixidó J. Resistance to MAPK Inhibitors in Melanoma Involves Activation of the IGF1R-MEK5-Erk5 Pathway. Cancer Res 2019; 79:2244-2256. [PMID: 30833419 DOI: 10.1158/0008-5472.can-18-2762] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 01/14/2019] [Accepted: 02/27/2019] [Indexed: 11/16/2022]
Abstract
Combined treatment of metastatic melanoma with BRAF and MEK inhibitors has improved survival, but the emergence of resistance represents an important clinical challenge. Targeting ERK is a suitable strategy currently being investigated in melanoma and other cancers. To anticipate possible resistance to ERK inhibitors (ERKi), we used SCH772984 (SCH) as a model ERKi to characterize resistance mechanisms in two BRAF V600E melanoma cell lines. The ERKi-resistant cells were also resistant to vemurafenib (VMF), trametinib (TMT), and combined treatment with either VMF and SCH or TMT and SCH. Resistance to SCH involved stimulation of the IGF1R-MEK5-Erk5 signaling pathway, which counteracted inhibition of Erk1/2 activation and cell growth. Inhibition of IGF1R with linsitinib blocked Erk5 activation in SCH-resistant cells and decreased their growth in 3D spheroid growth assays as well as in NOD scid gamma (NSG) mice. Cells doubly resistant to VMF and TMT or to VMF and SCH also exhibited downregulated Erk1/2 activation linked to stimulation of the IGF1R-MEK5-Erk5 pathway, which accounted for resistance. In addition, we found that the decreased Erk1/2 activation in SCH-resistant cells involved reduced expression and function of TGFα. These data reveal an escape signaling route that melanoma cells use to bypass Erk1/2 blockade during targeted melanoma treatment and offer several possible targets whose disruption may circumvent resistance. SIGNIFICANCE: Activation of the IGF1R-MEK5-Erk5 signaling pathway opposes pharmacologic inhibition of Erk1/2 in melanoma, leading to the reactivation of cell proliferation and acquired resistance.
Collapse
Affiliation(s)
- Lucía Benito-Jardón
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - Marta Díaz-Martínez
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - Nohemi Arellano-Sánchez
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - Paloma Vaquero-Morales
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - Azucena Esparís-Ogando
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, IBSAL, and CIBERONC, Salamanca, Spain
| | - Joaquin Teixidó
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain.
| |
Collapse
|
25
|
Sánchez-Fdez A, Ortiz-Ruiz MJ, Re-Louhau MF, Ramos I, Blanco-Múñez Ó, Ludeña D, Abad M, Sánchez-Martín M, Pandiella A, Esparís-Ogando A. MEK5 promotes lung adenocarcinoma. Eur Respir J 2019; 53:13993003.01327-2018. [PMID: 30442718 PMCID: PMC6393765 DOI: 10.1183/13993003.01327-2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 11/05/2018] [Indexed: 11/05/2022]
Abstract
Lung cancer represents the leading cause of cancer death worldwide [1]. Because of that, intense efforts are being devoted to the development of novel therapeutic strategies to fight the disease [2]. In this respect, identification of new oncogenic drivers offers therapeutic opportunities in tumours in which those molecules or other cooperating elements play a pathophysiological role. Here, we show that the MEK5 mitogen-activated protein kinase kinase has a pivotal role in lung cancer. MEK5 acts as an oncogenic driver in mice lung cancer and is pivotal for human lung adenocarcinomahttp://ow.ly/M9e830mZb8N
Collapse
Affiliation(s)
- Adrián Sánchez-Fdez
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, Salamanca, Spain.,IBSAL, Salamanca, Spain.,CIBERONC, Salamanca, Spain
| | - María Jesús Ortiz-Ruiz
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, Salamanca, Spain.,IBSAL, Salamanca, Spain
| | - María Florencia Re-Louhau
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, Salamanca, Spain.,IBSAL, Salamanca, Spain
| | - Isabel Ramos
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, Salamanca, Spain
| | - Óscar Blanco-Múñez
- IBSAL, Salamanca, Spain.,Pathology Service, University Hospital, Salamanca, Spain
| | - Dolores Ludeña
- IBSAL, Salamanca, Spain.,Pathology Service, University Hospital, Salamanca, Spain
| | - Mar Abad
- IBSAL, Salamanca, Spain.,Pathology Service, University Hospital, Salamanca, Spain
| | - Manuel Sánchez-Martín
- CIBERONC, Salamanca, Spain.,Transgenic Facility of the Nucleus Platform, University of Salamanca, Salamanca, Spain
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, Salamanca, Spain.,IBSAL, Salamanca, Spain.,CIBERONC, Salamanca, Spain
| | - Azucena Esparís-Ogando
- Instituto de Biología Molecular y Celular del Cáncer-CSIC, Salamanca, Spain.,IBSAL, Salamanca, Spain.,CIBERONC, Salamanca, Spain
| |
Collapse
|
26
|
Jiang W, Jin G, Cai F, Chen X, Cao N, Zhang X, Liu J, Chen F, Wang F, Dong W, Zhuang H, Hua ZC. Extracellular signal-regulated kinase 5 increases radioresistance of lung cancer cells by enhancing the DNA damage response. Exp Mol Med 2019; 51:1-20. [PMID: 30804322 PMCID: PMC6389946 DOI: 10.1038/s12276-019-0209-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 10/26/2018] [Accepted: 10/29/2018] [Indexed: 12/18/2022] Open
Abstract
Radiotherapy is a frequent mode of cancer treatment, although the development of radioresistance limits its effectiveness. Extensive investigations indicate the diversity of the mechanisms underlying radioresistance. Here, we aimed to explore the effects of extracellular signal-regulated kinase 5 (ERK5) on lung cancer radioresistance and the associated mechanisms. Our data showed that ERK5 is activated during solid lung cancer development, and ectopic expression of ERK5 promoted cell proliferation and G2/M cell cycle transition. In addition, we found that ERK5 is a potential regulator of radiosensitivity in lung cancer cells. Mechanistic investigations revealed that ERK5 could trigger IR-induced activation of Chk1, which has been implicated in DNA repair and cell cycle arrest in response to DNA double-strand breaks (DSBs). Subsequently, ERK5 knockdown or pharmacological inhibition selectively inhibited colony formation of lung cancer cells and enhanced IR-induced G2/M arrest and apoptosis. In vivo, ERK5 knockdown strongly radiosensitized A549 and LLC tumor xenografts to inhibition, with a higher apoptotic response and reduced tumor neovascularization. Taken together, our data indicate that ERK5 is a novel potential target for the treatment of lung cancer, and its expression might be used as a biomarker to predict radiosensitivity in NSCLC patients. Resistance to radiotherapy in patients with lung cancer may be countered by targeting a protein involved in promoting DNA repair. Radiotherapy causes DNA double-stranded breaks in lung cancer cells in order to kill them. However, cancer cells can show improved DNA repair and responses to damage, resulting in resistance to treatment. Zi-Chun Hua, Hongqin Zhuang at Nanjing University in China and co-workers examined the activity of the extracellular signal-related kinase 5 (ERK5) protein in response to the stress of ionizing radiation. They found that after radiation exposure ERK5 increased expression of another protein involved in DNA repair, facilitating cancer cell recovery. Knocking out ERK5 suppressed this resistance to radiotherapy. ERK5 could be a valuable target for treating lung cancer, and ERK5 expression level could be used as a biomarker for patient sensitivity to radiotherapy.
Collapse
Affiliation(s)
- Weiwei Jiang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Guanghui Jin
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China.,Department of Basic Medical Sciences, Medical College, Xiamen University, Xiamen, PR China
| | - Fangfang Cai
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Xiao Chen
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Nini Cao
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Xiangyu Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Jia Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Fei Chen
- Department of Nuclear Medicine, The Affiliated Nanjing First Hospital, Nanjing Medical University, Nanjing, PR China
| | - Feng Wang
- Department of Nuclear Medicine, The Affiliated Nanjing First Hospital, Nanjing Medical University, Nanjing, PR China
| | - Wei Dong
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China
| | - Hongqin Zhuang
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China.
| | - Zi-Chun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, PR China. .,Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories Inc., Changzhou, 213164, PR China.
| |
Collapse
|
27
|
Nguyen D, Lemos C, Wortmann L, Eis K, Holton SJ, Boemer U, Moosmayer D, Eberspaecher U, Weiske J, Lechner C, Prechtl S, Suelzle D, Siegel F, Prinz F, Lesche R, Nicke B, Nowak-Reppel K, Himmel H, Mumberg D, von Nussbaum F, Nising CF, Bauser M, Haegebarth A. Discovery and Characterization of the Potent and Highly Selective (Piperidin-4-yl)pyrido[3,2- d]pyrimidine Based in Vitro Probe BAY-885 for the Kinase ERK5. J Med Chem 2019; 62:928-940. [PMID: 30563338 DOI: 10.1021/acs.jmedchem.8b01606] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The availability of a chemical probe to study the role of a specific domain of a protein in a concentration- and time-dependent manner is of high value. Herein, we report the identification of a highly potent and selective ERK5 inhibitor BAY-885 by high-throughput screening and subsequent structure-based optimization. ERK5 is a key integrator of cellular signal transduction, and it has been shown to play a role in various cellular processes such as proliferation, differentiation, apoptosis, and cell survival. We could demonstrate that inhibition of ERK5 kinase and transcriptional activity with a small molecule did not translate into antiproliferative activity in different relevant cell models, which is in contrast to the results obtained by RNAi technology.
Collapse
Affiliation(s)
- Duy Nguyen
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Clara Lemos
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Lars Wortmann
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Knut Eis
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Simon J Holton
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Ulf Boemer
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Dieter Moosmayer
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Uwe Eberspaecher
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Joerg Weiske
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Christian Lechner
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Stefan Prechtl
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Detlev Suelzle
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Franziska Siegel
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Florian Prinz
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Ralf Lesche
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Barbara Nicke
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | | | - Herbert Himmel
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Dominik Mumberg
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Franz von Nussbaum
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Carl F Nising
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Marcus Bauser
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| | - Andrea Haegebarth
- Research & Development, Pharmaceuticals , Bayer AG , 13353 Berlin , Germany
| |
Collapse
|
28
|
Vaseva AV, Blake DR, Gilbert TSK, Ng S, Hostetter G, Azam SH, Ozkan-Dagliyan I, Gautam P, Bryant KL, Pearce KH, Herring LE, Han H, Graves LM, Witkiewicz AK, Knudsen ES, Pecot CV, Rashid N, Houghton PJ, Wennerberg K, Cox AD, Der CJ. KRAS Suppression-Induced Degradation of MYC Is Antagonized by a MEK5-ERK5 Compensatory Mechanism. Cancer Cell 2018; 34:807-822.e7. [PMID: 30423298 PMCID: PMC6321749 DOI: 10.1016/j.ccell.2018.10.001] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 07/03/2018] [Accepted: 10/01/2018] [Indexed: 12/20/2022]
Abstract
Our recent ERK1/2 inhibitor analyses in pancreatic ductal adenocarcinoma (PDAC) indicated ERK1/2-independent mechanisms maintaining MYC protein stability. To identify these mechanisms, we determined the signaling networks by which mutant KRAS regulates MYC. Acute KRAS suppression caused rapid proteasome-dependent loss of MYC protein, through both ERK1/2-dependent and -independent mechanisms. Surprisingly, MYC degradation was independent of PI3K-AKT-GSK3β signaling and the E3 ligase FBWX7. We then established and applied a high-throughput screen for MYC protein degradation and performed a kinome-wide proteomics screen. We identified an ERK1/2-inhibition-induced feedforward mechanism dependent on EGFR and SRC, leading to ERK5 activation and phosphorylation of MYC at S62, preventing degradation. Concurrent inhibition of ERK1/2 and ERK5 disrupted this mechanism, synergistically causing loss of MYC and suppressing PDAC growth.
Collapse
Affiliation(s)
- Angelina V Vaseva
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; The Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Devon R Blake
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Thomas S K Gilbert
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; UNC Michael Hooker Proteomics Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Serina Ng
- Department of Molecular and Cellular Biology, Roswell Park Cancer Center, Buffalo, NY 14203, USA
| | - Galen Hostetter
- Pathology and Biorepository Core, The Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Salma H Azam
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Irem Ozkan-Dagliyan
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Prson Gautam
- Institute for Molecular Medicine Finland, University of Helsinki, 00290 Helsinki, Finland
| | - Kirsten L Bryant
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kenneth H Pearce
- Center for Integrative Chemical Biology and Drug Discovery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Laura E Herring
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; UNC Michael Hooker Proteomics Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Haiyong Han
- Molecular Medicine Division, Translational Genomic Research Institute, Phoenix, AZ 85004, USA
| | - Lee M Graves
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Erik S Knudsen
- Department of Molecular and Cellular Biology, Roswell Park Cancer Center, Buffalo, NY 14203, USA
| | - Chad V Pecot
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Naim Rashid
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Peter J Houghton
- The Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Krister Wennerberg
- Institute for Molecular Medicine Finland, University of Helsinki, 00290 Helsinki, Finland
| | - Adrienne D Cox
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Channing J Der
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
29
|
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
|
30
|
Centa A, Rodríguez-Barrueco R, Montero JC, Pandiella A. The immunoglobulin-like domain of neuregulins potentiates ErbB3/HER3 activation and cellular proliferation. Mol Oncol 2018; 12:1061-1076. [PMID: 29683256 PMCID: PMC6026874 DOI: 10.1002/1878-0261.12310] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/04/2018] [Accepted: 04/09/2018] [Indexed: 12/03/2022] Open
Abstract
The neuregulins (NRGs) represent a large family of membrane‐anchored growth factors, whose deregulation may contribute to the pathogenesis of several tumors. In fact, targeting of NRG‐activated pathways has demonstrated clinical benefit. To improve the efficacy of anti‐NRG therapies, it is essential to gain insights into the regions of NRGs that favor their pro‐oncogenic properties. Here, we have addressed the protumorigenic impact of different NRG domains. To do this, deletion mutants affecting different NRG domains were expressed in 293 and MCF7 cells. Of the five forms studied, only the wild‐type and a mutant lacking the Ig‐like domain (NRGΔIg) were properly sorted to the plasma membrane. Both forms were released as soluble forms to the culture media. However, the mutant NRGΔIg failed to efficiently activate HER2 and HER3 receptors, signaling pathways, and cell proliferation when compared to wild‐type NRG. Treatment with trastuzumab, a humanized antibody used in the breast cancer clinic, inhibited the constitutive activation of HER2, HER3, and downstream signaling in MCF7 cells constitutively expressing wild‐type NRG. In contrast, this treatment had a marginal effect on MCF7‐NRGΔIg cells. This study demonstrates that the Ig‐like region of NRGs exerts an important role in their capability to activate ErbB/HER receptors and mitogenic responses. Strategies aimed at targeting NRGs should consider that fact to improve neutralization of the pro‐oncogenic properties of NRGs.
Collapse
Affiliation(s)
- Ariana Centa
- Instituto de Biología Molecular y Celular del Cáncer, IBSAL, CSIC and CIBERONC, Salamanca, Spain
| | - Ruth Rodríguez-Barrueco
- Instituto de Biología Molecular y Celular del Cáncer, IBSAL, CSIC and CIBERONC, Salamanca, Spain
| | - Juan Carlos Montero
- Instituto de Biología Molecular y Celular del Cáncer, IBSAL, CSIC and CIBERONC, Salamanca, Spain
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer, IBSAL, CSIC and CIBERONC, Salamanca, Spain
| |
Collapse
|
31
|
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
|
32
|
Orive-Ramos A, Seoane S, Ocaña A, Pandiella A, Montero JC. Regulation of the prometastatic neuregulin-MMP13 axis by SRC family kinases: therapeutic implications. Mol Oncol 2017; 11:1788-1805. [PMID: 29032615 PMCID: PMC5709617 DOI: 10.1002/1878-0261.12145] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 09/12/2017] [Accepted: 10/03/2017] [Indexed: 11/15/2022] Open
Abstract
Metastatic dissemination of tumor cells is responsible for the fatal outcome of breast cancer. Therefore, understanding the mechanisms involved in dissemination is essential for the development of new therapeutic strategies to prevent metastasis. One mechanism involved in metastatic dissemination of breast cancer cells is dependent on control of the production of matrix metalloproteinases by the neuregulins (NRGs). The NRGs are polypeptide factors that act by binding to the ErbB/HER subfamily of receptor tyrosine kinases. NRG‐mediated activation of HER receptors causes an increase in the production of metalloprotease 13 (MMP13, also termed collagenase‐3), which facilitates metastatic dissemination of breast tumors. In this context, we aimed to explore whether the clinically approved tyrosine kinase inhibitor dasatinib was able to neutralize this mechanism of metastatic dissemination. Here, we show that dasatinib restricted NRG‐induced MMP13 upregulation, both in vitro and in vivo, and in vivo metastatic dissemination of breast cancer cells. Chemical proteomics studies showed that the main cellular targets of dasatinib were SRC family kinases (SFKs). Moreover, genetic studies showed that knockdown of SRC or YES strongly inhibited NRG‐induced MMP13 upregulation in vitro. Mechanistically, dasatinib treatment or knockdown of SRC also inhibited ERK1/2 kinases in vitro, which were required for NRG‐induced MMP13 upregulation. These results open the possibility of clinically exploring the antitumoral action of dasatinib in those tumors in which the NRG–MMP13 signaling axis may play a relevant role in the control of tumor cell dissemination.
Collapse
Affiliation(s)
- Ana Orive-Ramos
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), CSIC, Salamanca, Spain
| | - Samuel Seoane
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), CSIC, Salamanca, Spain
| | - Alberto Ocaña
- Unidad de Investigación Traslacional, Hospital Universitario de Albacete, Universidad de Castilla La Mancha, Albacete, Spain.,Centro Regional de Investigaciones Biomédicas (CRIB), Universidad de Castilla La Mancha, Albacete, Spain.,CIBERONC, Madrid, Spain
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), CSIC, Salamanca, Spain.,CIBERONC, Madrid, Spain
| | - Juan Carlos Montero
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), CSIC, Salamanca, Spain.,CIBERONC, Madrid, Spain
| |
Collapse
|
33
|
Zhang X, Maity T, Kashyap MK, Bansal M, Venugopalan A, Singh S, Awasthi S, Marimuthu A, Charles Jacob HK, Belkina N, Pitts S, Cultraro CM, Gao S, Kirkali G, Biswas R, Chaerkady R, Califano A, Pandey A, Guha U. Quantitative Tyrosine Phosphoproteomics of Epidermal Growth Factor Receptor (EGFR) Tyrosine Kinase Inhibitor-treated Lung Adenocarcinoma Cells Reveals Potential Novel Biomarkers of Therapeutic Response. Mol Cell Proteomics 2017; 16:891-910. [PMID: 28331001 PMCID: PMC5417828 DOI: 10.1074/mcp.m117.067439] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 02/24/2017] [Indexed: 02/05/2023] Open
Abstract
Mutations in the Epidermal growth factor receptor (EGFR) kinase domain, such as the L858R missense mutation and deletions spanning the conserved sequence 747LREA750, are sensitive to tyrosine kinase inhibitors (TKIs). The gatekeeper site residue mutation, T790M accounts for around 60% of acquired resistance to EGFR TKIs. The first generation EGFR TKIs, erlotinib and gefitinib, and the second generation inhibitor, afatinib are FDA approved for initial treatment of EGFR mutated lung adenocarcinoma. The predominant biomarker of EGFR TKI responsiveness is the presence of EGFR TKI-sensitizing mutations. However, 30-40% of patients with EGFR mutations exhibit primary resistance to these TKIs, underscoring the unmet need of identifying additional biomarkers of treatment response. Here, we sought to characterize the dynamics of tyrosine phosphorylation upon EGFR TKI treatment of mutant EGFR-driven human lung adenocarcinoma cell lines with varying sensitivity to EGFR TKIs, erlotinib and afatinib. We employed stable isotope labeling with amino acids in cell culture (SILAC)-based quantitative mass spectrometry to identify and quantify tyrosine phosphorylated peptides. The proportion of tyrosine phosphorylated sites that had reduced phosphorylation upon erlotinib or afatinib treatment correlated with the degree of TKI-sensitivity. Afatinib, an irreversible EGFR TKI, more effectively inhibited tyrosine phosphorylation of a majority of the substrates. The phosphosites with phosphorylation SILAC ratios that correlated with the TKI-sensitivity of the cell lines include sites on kinases, such as EGFR-Y1197 and MAPK7-Y221, and adaptor proteins, such as SHC1-Y349/350, ERRFI1-Y394, GAB1-Y689, STAT5A-Y694, DLG3-Y705, and DAPP1-Y139, suggesting these are potential biomarkers of TKI sensitivity. DAPP1, is a novel target of mutant EGFR signaling and Y-139 is the major site of DAPP1 tyrosine phosphorylation. We also uncovered several off-target effects of these TKIs, such as MST1R-Y1238/Y1239 and MET-Y1252/1253. This study provides unique insight into the TKI-mediated modulation of mutant EGFR signaling, which can be applied to the development of biomarkers of EGFR TKI response.
Collapse
Affiliation(s)
- Xu Zhang
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Tapan Maity
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Manoj K Kashyap
- §Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205
| | - Mukesh Bansal
- ¶Department of System Biology, Columbia University, New York, New York, 10032
- ‖PsychoGenics Inc., Tarrytown, New York, 10591
| | - Abhilash Venugopalan
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Sahib Singh
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Shivangi Awasthi
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | | | | | - Natalya Belkina
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Stephanie Pitts
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Constance M Cultraro
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Shaojian Gao
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Guldal Kirkali
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Romi Biswas
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Raghothama Chaerkady
- §Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205
- **Medimmune LLC, Gaithersburg, Maryland, 20878
| | - Andrea Califano
- ¶Department of System Biology, Columbia University, New York, New York, 10032
| | - Akhilesh Pandey
- §Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205
| | - Udayan Guha
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892;
| |
Collapse
|
34
|
Hoang VT, Yan TJ, Cavanaugh JE, Flaherty PT, Beckman BS, Burow ME. Oncogenic signaling of MEK5-ERK5. Cancer Lett 2017; 392:51-59. [PMID: 28153789 PMCID: PMC5901897 DOI: 10.1016/j.canlet.2017.01.034] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 01/17/2017] [Accepted: 01/23/2017] [Indexed: 12/17/2022]
Abstract
Mitogen-activated protein kinases (MAPKs) regulate diverse cellular processes including proliferation, cell survival, differentiation, and apoptosis. While conventional MAPK constituents have well-defined roles in oncogenesis, the MEK5 pathway has only recently emerged in cancer research. In this review, we consider the MEK5 signaling cascade, focusing specifically on its involvement in drug resistance and regulation of aggressive cancer phenotypes. Moreover, we explore the role of MEK5/ERK5 in tumorigenesis and metastatic progression, discussing the discrepancies in preclinical studies and assessing its viability as a therapeutic target for anti-cancer agents.
Collapse
Affiliation(s)
- Van T Hoang
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA, USA
| | - Thomas J Yan
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA, USA
| | - Jane E Cavanaugh
- Department of Pharmacological Sciences, Division of Medicinal Chemistry, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | - Patrick T Flaherty
- Department of Pharmacological Sciences, Division of Medicinal Chemistry, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | | | - Matthew E Burow
- Department of Medicine-Section of Hematology and Medical Oncology, Tulane University, New Orleans, LA, USA; Department of Pharmacology, Tulane University, New Orleans, LA, USA; Tulane Cancer Center, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
35
|
Targeting BMK1 Impairs the Drug Resistance to Combined Inhibition of BRAF and MEK1/2 in Melanoma. Sci Rep 2017; 7:46244. [PMID: 28387310 PMCID: PMC5384194 DOI: 10.1038/srep46244] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 03/14/2017] [Indexed: 01/20/2023] Open
Abstract
Combined inhibition of BRAF and MEK1/2 (CIBM) improves therapeutic efficacy of BRAF-mutant melanoma. However, drug resistance to CIBM is inevitable and the drug resistance mechanisms still remain to be elucidated. Here, we show that BMK1 pathway contributes to the drug resistance to CIBM. Considering that ERK1/2 pathway regulates cellular processes by phosphorylating, we first performed a SILAC phosphoproteomic profiling of CIBM. Phosphorylation of 239 proteins was identified to be downregulated, while phosphorylation of 47 proteins was upregulated. Following siRNA screening of 47 upregulated proteins indicated that the knockdown of BMK1 showed the most significant ability to inhibit the proliferation of CIBM resistant cells. It was found that phosphorylation of BMK1 was enhanced in resistant cells, which suggested an association of BMK1 with drug resistance. Further study indicated that phospho-activation of BMK1 by MEK5D enhanced the resistance to CIBM. Conversely, inhibition of BMK1 by shRNAi or BMK1 inhibitor (XMD8-92) impaired not only the acquirement of resistance to CIBM, but also the proliferation of CIBM resistant cells. Further kinome-scale siRNA screening demonstrated that SRC\MEK5 cascade promotes the phospho-activation of BMK1 in response to CIBM. Our study not only provides a global phosphoproteomic view of CIBM in melanoma, but also demonstrates that inhibition of BMK1 has therapeutic potential for the treatment of melanoma.
Collapse
|
36
|
Zheng R, Studzinski GP. Nuclear ERK5 inhibits progression of leukemic monocytes to macrophages by regulating the transcription factor PU.1 and heat shock protein HSP70. Leuk Lymphoma 2016; 58:1468-1480. [PMID: 27748139 DOI: 10.1080/10428194.2016.1243675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Differentiation therapy can supplement the therapy of APL, but other subtypes of AML are treated principally with cytotoxic agents, with few lasting remissions. While the induction of monocyte followed by macrophage differentiation by vitamin D derivatives (VDDs) is dramatic in cultured AML cells of all subtypes, attempts to translate this to the clinic have not been effective. Thus, better understanding of the mechanisms underlying VDD-induced differentiation may improve this approach. The key events in this form of differentiation include increased expression of CD11b, and the transcription factor PU.1 is known to be a part of this process. We show here that in the transition of monocytes to macrophages induced by a VDD, ERK5, a member of the MAPK family of signaling molecules, prevents PU.1 expression. However, upon ERK5 inhibition PU.1 protein is stabilized by HSP70.Thus, ERK5 may be a target for manipulation of the immunoregulatory actions of macrophages in cancer.
Collapse
Affiliation(s)
- Ruifang Zheng
- a Department of Pathology and Laboratory Medicine , New Jersey Medical School, Rutgers University , Newark , NJ , USA
| | - George P Studzinski
- a Department of Pathology and Laboratory Medicine , New Jersey Medical School, Rutgers University , Newark , NJ , USA
| |
Collapse
|
37
|
Angulo-Ibáñez M, Rovira-Clavé X, Granados-Jaén A, Downs B, Kim YC, Wang SM, Reina M, Espel E. Erk5 contributes to maintaining the balance of cellular nucleotide levels and erythropoiesis. Cell Cycle 2016; 14:3864-76. [PMID: 26697837 DOI: 10.1080/15384101.2015.1120914] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
An adequate supply of nucleotides is essential for accurate DNA replication, and inappropriate deoxyribonucleotide triphosphate (dNTP) concentrations can lead to replication stress, a common source of DNA damage, genomic instability and tumourigenesis. Here, we provide evidence that Erk5 is necessary for correct nucleotide supply during erythroid development. Mice with Erk5 knockout in the haematopoietic lineage showed impaired erythroid development in bone marrow, accompanied by altered dNTP levels and increased DNA mutagenesis in erythroid progenitors as detected by exome sequencing. Moreover, Erk5-depleted leukemic Jurkat cells presented a marked sensitivity to thymidine-induced S phase stalling, as evidenced by increased H2AX phosphorylation and apoptosis. The increase in thymidine sensitivity correlated with a higher dTTP/dCTP ratio. These results indicate that Erk5 is necessary to maintain the balance of nucleotide levels, thus preventing dNTP misincorporation and DNA damage in proliferative erythroid progenitors and leukemic Jurkat T cells.
Collapse
Affiliation(s)
- Maria Angulo-Ibáñez
- a Department of Cell Biology ; Celltec-UB; Facultat de Biologia; Universitat de Barcelona ; Barcelona , Spain
| | - Xavier Rovira-Clavé
- a Department of Cell Biology ; Celltec-UB; Facultat de Biologia; Universitat de Barcelona ; Barcelona , Spain
| | - Alba Granados-Jaén
- a Department of Cell Biology ; Celltec-UB; Facultat de Biologia; Universitat de Barcelona ; Barcelona , Spain
| | - Bradley Downs
- b University of Nebraska Medical Center; Nebraska Medical Center ; Omaha , NE USA
| | - Yeong C Kim
- b University of Nebraska Medical Center; Nebraska Medical Center ; Omaha , NE USA
| | - San Ming Wang
- b University of Nebraska Medical Center; Nebraska Medical Center ; Omaha , NE USA
| | - Manuel Reina
- a Department of Cell Biology ; Celltec-UB; Facultat de Biologia; Universitat de Barcelona ; Barcelona , Spain
| | - Enric Espel
- a Department of Cell Biology ; Celltec-UB; Facultat de Biologia; Universitat de Barcelona ; Barcelona , Spain.,c Department of Physiology and Immunology ; Facultat de Biologia; Universitat de Barcelona ; Barcelona ; Spain
| |
Collapse
|
38
|
Song C, Xu Q, Jiang K, Zhou G, Yu X, Wang L, Zhu Y, Fang L, Yu Z, Lee JD, Yu SC, Yang Q. Inhibition of BMK1 pathway suppresses cancer stem cells through BNIP3 and BNIP3L. Oncotarget 2016; 6:33279-89. [PMID: 26432836 PMCID: PMC4741765 DOI: 10.18632/oncotarget.5337] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/17/2015] [Indexed: 12/22/2022] Open
Abstract
Cancer stem cells (CSCs) possess many characteristics associated with stem cells and are believed to drive tumor initiation. Although targeting of CSCs offers great promise for the new generation of therapeutics, lack of the effective drugable target and appropriate pharmacological reagents significantly impedes the development of chemotherapies. Here, we show that the phosphorylation of BMK1 was significantly correlated with not only embryonic and induced pluripotent stem (iPS) cells, but also the CSCs. It was showed that activation of BMK1 by the expression of MEK5D enhanced the self-renew (sphere formation), proliferation (clone formation) and tumorigenic capacity of CSCs. While BMK1 inhibitor, XMD8-92, suppressed these capacities. RNA-seq and microarray analysis revealed that inhibition of BMK1 significantly enhanced the expression of BNIP3 and BNIP3L, which play important roles in cell death. Further study indicated that shRNA-mediated knock down of BNIP3 and BNIP3L impairs the BMK1 inhibitor, XMD8-92-induced suppression of sphere formation and clone formation of CSC. Collectively, these results not only indicate that BMK1 plays an important role in maintaining "stemness" of CSCs, but also implicate that BMK1 might be a potential drug target for CSCs.
Collapse
Affiliation(s)
- Chengli Song
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| | - Qiang Xu
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| | - Kui Jiang
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| | - Guangyu Zhou
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| | - Xuebin Yu
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| | - Lina Wang
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| | - Yuting Zhu
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| | - Liping Fang
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| | - Zhe Yu
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| | - Jiing-Dwan Lee
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Shi-Cang Yu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Qingkai Yang
- Department of Oncology, The Second Affiliated Hospital of DaLian Medical University, Institute of Cancer Stem Cell, DaLian Medical University, Dalian, Liaoning 116044, China
| |
Collapse
|
39
|
Novel targets for paclitaxel nano formulations: Hopes and hypes in triple negative breast cancer. Pharmacol Res 2016; 111:577-591. [PMID: 27461138 DOI: 10.1016/j.phrs.2016.07.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 07/18/2016] [Accepted: 07/19/2016] [Indexed: 12/30/2022]
Abstract
Triple negative breast cancer is defined as one of the utmost prevailing breast cancers worldwide, possessing an inadequate prognosis and treatment option limited to chemotherapy and radiotherapy, creating a challenge for researchers as far as developing a specific targeted therapy is concerned. The past research era has shown several promising outcomes for TNBC such as nano-formulations of the chemotherapeutic agents already used for the management of the malignant tumor. Taking a glance at paclitaxel nano formulations, it has been proven beneficial in several researches in the past decade; nevertheless its solubility is often a challenge to scientists in achieving success. We have henceforth discussed the basic heterogeneity of triple negative breast cancer along with the current management options as well as a brief outlook on pros and cons of paclitaxel, known as the most widely used chemotherapeutic agent for the treatment of the disease. We further analyzed the need of nanotechnology pertaining to the problems encountered with the current paclitaxel formulations available discussing the strategic progress in various nano-formulations till date taking into account the basic research strategies required in terms of solubility, permeability, physicochemical properties, active and passive targeting. A thorough review in recent advances in active targeting for TNBC was carried out whereby the various ligands which are at present finding its way into TNBC research such as hyaluronic acid, folic acid, transferrin, etc. were discussed. These ligands have specific receptor affinity to TNBC tumor cells hence can be beneficial for novel drug targeting approaches. Conversely, there are currently several novel strategies in the research pipeline whose targeting ligands have not yet been studied. Therefore, we reviewed upon the numerous novel receptor targets along with the respective nano-formulation aspects which have not yet been fully researched upon and could be exemplified as outstanding target strategies for TNBC which is currently an urgent requirement.
Collapse
|
40
|
Tsioumpekou M, Papadopoulos N, Burovic F, Heldin CH, Lennartsson J. Platelet-derived growth factor (PDGF)-induced activation of Erk5 MAP-kinase is dependent on Mekk2, Mek1/2, PKC and PI3-kinase, and affects BMP signaling. Cell Signal 2016; 28:1422-1431. [PMID: 27339033 DOI: 10.1016/j.cellsig.2016.06.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 06/16/2016] [Accepted: 06/16/2016] [Indexed: 02/09/2023]
Abstract
Platelet-derived growth factor-BB (PDGF-BB) binds to its tyrosine kinase receptors (PDGFRs) and stimulates mitogenicity and survival of cells of mesenchymal origin. Activation of PDGFRs initiates a number of downstream signaling pathways, including phosphatidyl 3'-inositol kinase (PI3-kinase), phospholipase Cγ and MAP kinase pathways. In this report, we show that Erk5 MAP kinase is activated in response to PDGF-BB in the smooth muscle cell line MOVAS in a manner dependent on Mekk2, Mek1/2, Mek5, PI3-kinase and protein kinase C (PKC). The co-operation of Mek1/2 and Mekk2 in the activation of Erk5, suggests a close co-regulation between the Erk1/2 and Erk5 MAP kinase pathways. Furthermore, we found that classical PKCs are important for Erk5 activation. In addition, we found that PKCζ interacts with Erk5 and may exert a negative feed-back effect. We observed no nuclear accumulation of Erk5 in response to PDGF-BB stimulation, however, we identified a mechanism by which cytoplasmic Erk5 influences gene expression; Erk5 was essential for PDGF-BB-mediated Smad1/5/8 signaling by stimulating release and/or activation of bone morphogenetic protein(s) (BMPs). Thus, PDGF-BB-induced Erk5 activation involves parallel stimulatory and inhibitory pathways and promotes Smad1/5/8 signaling.
Collapse
Affiliation(s)
- Maria Tsioumpekou
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE-751 24 Uppsala, Sweden
| | - Natalia Papadopoulos
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE-751 24 Uppsala, Sweden
| | - Fatima Burovic
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE-751 24 Uppsala, Sweden
| | - Carl-Henrik Heldin
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE-751 24 Uppsala, Sweden
| | - Johan Lennartsson
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Box 595, SE-751 24 Uppsala, Sweden.
| |
Collapse
|
41
|
ERK5 signalling rescues intestinal epithelial turnover and tumour cell proliferation upon ERK1/2 abrogation. Nat Commun 2016; 7:11551. [PMID: 27187615 PMCID: PMC4873670 DOI: 10.1038/ncomms11551] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 04/07/2016] [Indexed: 12/14/2022] Open
Abstract
The ERK1/2 MAPK signalling module integrates extracellular cues that induce proliferation and differentiation of epithelial lineages, and is an established oncogenic driver, particularly in the intestine. However, the interrelation of the ERK1/2 module relative to other signalling pathways in intestinal epithelial cells and colorectal cancer (CRC) is unclear. Here we show that loss of Erk1/2 in intestinal epithelial cells results in defects in nutrient absorption, epithelial cell migration and secretory cell differentiation. However, intestinal epithelial cell proliferation is not impeded, implying compensatory mechanisms. Genetic deletion of Erk1/2 or pharmacological targeting of MEK1/2 results in supraphysiological activity of the ERK5 pathway. Furthermore, targeting both pathways causes a more effective suppression of cell proliferation in murine intestinal organoids and human CRC lines. These results suggest that ERK5 provides a common bypass route in intestinal epithelial cells, which rescues cell proliferation upon abrogation of ERK1/2 signalling, with therapeutic implications in CRC. It is unclear how the extracellular signal-regulated kinases 1 and 2 (ERK1/2) pathways interact with other signals in intestinal epithelial cells. Here, the authors show that upon loss of Erk1/2, or pharmacological inhibition of MEK1/2, the ERK5 pathway is upregulated to maintain epithelial cell proliferation.
Collapse
|
42
|
Fulford L, Milewski D, Ustiyan V, Ravishankar N, Cai Y, Le T, Masineni S, Kasper S, Aronow B, Kalinichenko VV, Kalin TV. The transcription factor FOXF1 promotes prostate cancer by stimulating the mitogen-activated protein kinase ERK5. Sci Signal 2016; 9:ra48. [DOI: 10.1126/scisignal.aad5582] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
43
|
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
|
44
|
Lochhead PA, Clark J, Wang LZ, Gilmour L, Squires M, Gilley R, Foxton C, Newell DR, Wedge SR, Cook SJ. Tumor cells with KRAS or BRAF mutations or ERK5/MAPK7 amplification are not addicted to ERK5 activity for cell proliferation. Cell Cycle 2016; 15:506-18. [PMID: 26959608 PMCID: PMC5056618 DOI: 10.1080/15384101.2015.1120915] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 10/08/2015] [Accepted: 11/12/2015] [Indexed: 10/24/2022] Open
Abstract
ERK5, encoded by MAPK7, has been proposed to play a role in cell proliferation, thus attracting interest as a cancer therapeutic target. While oncogenic RAS or BRAF cause sustained activation of the MEK1/2-ERK1/2 pathway, ERK5 is directly activated by MEK5. It has been proposed that RAS and RAF proteins can also promote ERK5 activation. Here we investigated the interplay between RAS-RAF-MEK-ERK and ERK5 signaling and studied the role of ERK5 in tumor cell proliferation in 2 disease-relevant cell models. We demonstrate that although an inducible form of CRAF (CRAF:ER*) can activate ERK5 in fibroblasts, the response is delayed and reflects feed-forward signaling. Additionally, oncogenic KRAS and BRAF do not activate ERK5 in epithelial cells. Although KRAS and BRAF do not couple directly to MEK5-ERK5, ERK5 signaling might still be permissive for proliferation. However, neither the selective MEK5 inhibitor BIX02189 or ERK5 siRNA inhibited proliferation of colorectal cancer cells harbouring KRAS(G12C/G13D) or BRAF(V600E). Furthermore, there was no additive or synergistic effect observed when BIX02189 was combined with the MEK1/2 inhibitor Selumetinib (AZD6244), suggesting that ERK5 was neither required for proliferation nor a driver of innate resistance to MEK1/2 inhibitors. Finally, even cancer cells with MAPK7 amplification were resistant to BIX02189 and ERK5 siRNA, showing that ERK5 amplification does not confer addiction to ERK5 for cell proliferation. Thus ERK5 signaling is unlikely to play a role in tumor cell proliferation downstream of KRAS or BRAF or in tumor cells with ERK5 amplification. These results have important implications for the role of ERK5 as an anti-cancer drug target.
Collapse
Affiliation(s)
| | - Jonathan Clark
- Biological Chemistry Facility; The Babraham Institute; Cambridge, UK
| | - Lan-Zhen Wang
- The Northern Institute for Cancer Research; University of Newcastle upon Tyne, Newcastle, UK
| | - Lesley Gilmour
- Cancer Research Technology; The Beatson Institute for Cancer Research; Garscube Estate; Glasgow, UK
- Current address: Translational Radiation Biology; The Beatson Institute for Cancer Research; Garscube Estate; Glasgow, UK
| | - Matthew Squires
- Signalling Laboratory; The Babraham Institute; Cambridge, UK
- Current address: Novartis; Basel, Switzerland
| | - Rebecca Gilley
- Signalling Laboratory; The Babraham Institute; Cambridge, UK
| | - Caroline Foxton
- Cancer Research Technology; CRT Discovery Laboratories; London Bioscience Innovation Centre; London, UK
- Current address: Centre for Drug Development; Cancer Research UK; London, UK
| | - David R. Newell
- The Northern Institute for Cancer Research; University of Newcastle upon Tyne, Newcastle, UK
| | - Stephen R. Wedge
- The Northern Institute for Cancer Research; University of Newcastle upon Tyne, Newcastle, UK
| | - Simon J. Cook
- Signalling Laboratory; The Babraham Institute; Cambridge, UK
| |
Collapse
|
45
|
Xiong Y, Zhang L, Wang T. Phosphorylation of BMK1 induces prostatic carcinoma cell proliferation by promoting entry into the S phase of the cell cycle. Oncol Lett 2015; 11:99-104. [PMID: 26870175 PMCID: PMC4727042 DOI: 10.3892/ol.2015.3909] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 09/29/2015] [Indexed: 01/03/2023] Open
Abstract
Big mitogen-activated protein kinase 1 (BMK1) is activated by mitogens and oncogenic signals, and is strongly implicated in tumorigenesis. In the present study, it was demonstrated that the activation of BMK1, but not extracellular signal-regulated kinase (ERK)1/2, can induce proliferation in prostate cancer cells. It was found that the proliferation of epidermal growth factor (EGF)-treated cells was accelerated by 40% compared with non-treated cells using a CCK8 assay. In addition, cell cycle analysis using flow cytometry showed that the proportion of cells in the S phase increased significantly in the BMK1-activated PC-3 cells, suggesting that the activation of BMK1 promotes entry into the S phase of the cell cycle in prostate cancer cells. Furthermore, western blot analysis indicated that EGF-mediated activation of BMK1, but not ERK1/2, participates in the proliferation and cell cycle regulation in prostate cancer cells. Furthermore, the effects of cell cycle regulation by the activation of BMK1 were associated with the increased expression levels of cyclin A and cyclin E, whereas the expression of cyclin B and cyclin D1 was unchanged in this process. Therefore, the present study demonstrated that the activation of BMK1 can induce proliferation by promoting entry into the S phase through the upregulation of cyclin A and cyclin E expression levels in prostate cancer cells.
Collapse
Affiliation(s)
- Ying Xiong
- Department of Urology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Luying Zhang
- Division of Anatomy, Hubei College of Chinese Medicine, Jingzhou, Hubei 434100, P.R. China
| | - Tao Wang
- Department of Urology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| |
Collapse
|
46
|
Ortiz-Ruiz MJ, Álvarez-Fernández S, Parrott T, Zaknoen S, Burrows FJ, Ocaña A, Pandiella A, Esparís-Ogando A. Therapeutic potential of ERK5 targeting in triple negative breast cancer. Oncotarget 2015; 5:11308-18. [PMID: 25350956 PMCID: PMC4294347 DOI: 10.18632/oncotarget.2324] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 08/06/2014] [Indexed: 12/14/2022] Open
Abstract
Triple negative breast cancers (TNBCs) account for 15% of all breast cancers, and represent one of the most aggressive forms of the disease, exhibiting short relapse-free survival. In contrast to other breast cancer subtypes, the absence of knowledge about the etiopathogenic alterations that cause TNBCs force the use of chemotherapeutics to treat these tumors. Because of this, efforts have been devoted with the aim of incorporating novel therapies into the clinical setting. Kinases play important roles in the pathophysiology of several tumors, including TNBC. Since expression of the MAP kinase ERK5 has been linked to patient outcome in breast cancer, we analyzed the potential value of its targeting in TNBC. ERK5 was frequently overexpressed and active in samples from patients with TNBC, as well as in explants from mice carrying genetically-defined TNBC tumors. Moreover, expression of ERK5 was linked to a worse prognosis in TNBC patients. Knockdown experiments demonstrated that ERK5 supported proliferation of TNBC cells. Pharmacological inhibition of ERK5 with TG02, a clinical stage inhibitor which targets ERK5 and other kinases, inhibited cell proliferation by blocking passage of cells through G1 and G2, and also triggered apoptosis in certain TNBC cell lines. TG02 had significant antitumor activity in a TNBC xenograft model in vivo, and also augmented the activity of chemotherapeutic agents commonly used to treat TNBC. Together, these data indicate that ERK5 targeting may represent a valid strategy against TNBC, and support the development of trials aimed at evaluating the clinical effectiveness of drugs that block this kinase.
Collapse
Affiliation(s)
- María Jesús Ortiz-Ruiz
- Instituto de Biología Molecular y Celular del Cáncer. CSIC-IBSAL-Universidad de Salamanca. Spain
| | - Stela Álvarez-Fernández
- Instituto de Biología Molecular y Celular del Cáncer. CSIC-IBSAL-Universidad de Salamanca. Spain
| | | | | | | | - Alberto Ocaña
- Hospital Universitario de Albacete, and AECC Unit, Spain
| | - Atanasio Pandiella
- Instituto de Biología Molecular y Celular del Cáncer. CSIC-IBSAL-Universidad de Salamanca. Spain
| | - Azucena Esparís-Ogando
- Instituto de Biología Molecular y Celular del Cáncer. CSIC-IBSAL-Universidad de Salamanca. Spain
| |
Collapse
|
47
|
Wilhelmsen K, Xu F, Farrar K, Tran A, Khakpour S, Sundar S, Prakash A, Wang J, Gray NS, Hellman J. Extracellular signal-regulated kinase 5 promotes acute cellular and systemic inflammation. Sci Signal 2015; 8:ra86. [PMID: 26307013 DOI: 10.1126/scisignal.aaa3206] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Inflammatory critical illness is a syndrome that is characterized by acute inflammation and organ injury, and it is triggered by infections and noninfectious tissue injury, both of which activate innate immune receptors and pathways. Although reports suggest an anti-inflammatory role for the mitogen-activated protein kinase (MAPK) extracellular signal-regulated kinase 5 (ERK5), we previously found that ERK5 mediates proinflammatory responses in primary human cells in response to stimulation of Toll-like receptor 2 (TLR2). We inhibited the kinase activities and reduced the abundances of ERK5 and MEK5, a MAPK kinase directly upstream of ERK5, in primary human vascular endothelial cells and monocytes, and found that ERK5 promoted inflammation induced by a broad range of microbial TLR agonists and by the proinflammatory cytokines interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α). Furthermore, we found that inhibitors of MEK5 or ERK5 reduced the plasma concentrations of proinflammatory cytokines in mice challenged with TLR ligands or heat-killed Staphylococcus aureus, as well as in mice that underwent sterile lung ischemia-reperfusion injury. Finally, we found that inhibition of ERK5 protected endotoxemic mice from death. Together, our studies support a proinflammatory role for ERK5 in primary human endothelial cells and monocytes, and suggest that ERK5 is a potential therapeutic target in diverse disorders that cause inflammatory critical illness.
Collapse
Affiliation(s)
- Kevin Wilhelmsen
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Fengyun Xu
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Katherine Farrar
- Graduate Program in Biomedical Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alphonso Tran
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Samira Khakpour
- Graduate Program in Biomedical Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Shirin Sundar
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arun Prakash
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jinhua Wang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Nathanael S Gray
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Judith Hellman
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94143, USA. Division of Critical Care Medicine and Biomedical Sciences Program, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
48
|
Hernández-García S, González V, Sanz E, Pandiella A. Effect of Oncoxin Oral Solution in HER2-Overexpressing Breast Cancer. Nutr Cancer 2015; 67:1159-69. [PMID: 26241555 DOI: 10.1080/01635581.2015.1068819] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
One of the most aggressive breast cancer subtypes includes tumors with high expression of HER2. Gene expression and functional studies have shown a link between HER2 overexpression and oxidative stress. Because of this, we hypothesized that Oncoxin Oral Solution (OOS), a composite product that contains several antioxidants, could have an antitumoral effect against HER2+ tumors. Dose-response studies, biochemical and cytometric assessment of the effect of OOS on cell cycle and apoptosis, and drug combination analyses were performed on BT474 and SKBR3 cells, 2 HER2-overexpressing breast cancer cell lines. OOS reduced the proliferation of these cells, and augmented the action of lapatinib, a HER2 inhibitor used in the breast cancer clinic. Moreover, OOS decreased growth of HER2+ tumors in mice. Mechanistically, OOS provoked cell cycle blockade through upregulation of p27 expression and downregulation of cyclin D levels. OOS also caused apoptotic cell death in HER2+ breast cancer cells, as indicated by increases in PARP cleavage as well as upregulation of caspase 8 and caspase 3 activities. These results demonstrate an antitumoral action of OOS in preclinical models of HER2+ breast cancer and suggest that it can be used with anti-HER2 therapies currently adopted as standard of care in the oncology clinic.
Collapse
Affiliation(s)
- Susana Hernández-García
- a Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca , Salamanca , Spain
| | - Verena González
- a Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca , Salamanca , Spain
| | | | - Atanasio Pandiella
- a Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca , Salamanca , Spain
| |
Collapse
|
49
|
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
|
50
|
Cameron SJ, Ture SK, Mickelsen D, Chakrabarti E, Modjeski KL, McNitt S, Seaberry M, Field DJ, Le NT, Abe JI, Morrell CN. Platelet Extracellular Regulated Protein Kinase 5 Is a Redox Switch and Triggers Maladaptive Platelet Responses and Myocardial Infarct Expansion. Circulation 2015; 132:47-58. [PMID: 25934838 DOI: 10.1161/circulationaha.115.015656] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 04/27/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND Platelets have a pathophysiologic role in the ischemic microvascular environment of acute coronary syndromes. In comparison with platelet activation in normal healthy conditions, less attention is given to mechanisms of platelet activation in diseased states. Platelet function and mechanisms of activation in ischemic and reactive oxygen species-rich environments may not be the same as in normal healthy conditions. Extracellular regulated protein kinase 5 (ERK5) is a mitogen-activated protein kinase family member activated in hypoxic, reactive oxygen species-rich environments and in response to receptor-signaling mechanisms. Prior studies suggest a protective effect of ERK5 in endothelial and myocardial cells after ischemia. We present evidence that platelets express ERK5 and that platelet ERK5 has an adverse effect on platelet activation via selective receptor-dependent and receptor-independent reactive oxygen species-mediated mechanisms in ischemic myocardium. METHODS AND RESULTS Using isolated human platelets and a mouse model of myocardial infarction (MI), we found that platelet ERK5 is activated post-MI and that platelet-specific ERK5(-/-) mice have less platelet activation, reduced MI size, and improved post-MI heart function. Furthermore, the expression of downstream ERK5-regulated proteins is reduced in ERK5(-/-) platelets post-MI. CONCLUSIONS ERK5 functions as a platelet activator in ischemic conditions, and platelet ERK5 maintains the expression of some platelet proteins after MI, leading to infarct expansion. This demonstrates that platelet function in normal healthy conditions is different from platelet function in chronic ischemic and inflammatory conditions. Platelet ERK5 may be a target for acute therapeutic intervention in the thrombotic and inflammatory post-MI environment.
Collapse
Affiliation(s)
- Scott J Cameron
- From Aab Cardiovascular Research Institute, University of Rochester School of Medicine, NY (S.J.C., S.K.T., D.M., E.C., K.L.M., M.S., D.J.F., C.N.M.); Department of Medicine (S.J.C., C.N.M.) and Heart Research Follow-Up Program (S.M.), Division of Cardiology, University of Rochester School of Medicine, NY; and Department of Cardiology Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston (N.-T.L., J.-i.A.)
| | - Sara K Ture
- From Aab Cardiovascular Research Institute, University of Rochester School of Medicine, NY (S.J.C., S.K.T., D.M., E.C., K.L.M., M.S., D.J.F., C.N.M.); Department of Medicine (S.J.C., C.N.M.) and Heart Research Follow-Up Program (S.M.), Division of Cardiology, University of Rochester School of Medicine, NY; and Department of Cardiology Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston (N.-T.L., J.-i.A.)
| | - Deanne Mickelsen
- From Aab Cardiovascular Research Institute, University of Rochester School of Medicine, NY (S.J.C., S.K.T., D.M., E.C., K.L.M., M.S., D.J.F., C.N.M.); Department of Medicine (S.J.C., C.N.M.) and Heart Research Follow-Up Program (S.M.), Division of Cardiology, University of Rochester School of Medicine, NY; and Department of Cardiology Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston (N.-T.L., J.-i.A.)
| | - Enakshi Chakrabarti
- From Aab Cardiovascular Research Institute, University of Rochester School of Medicine, NY (S.J.C., S.K.T., D.M., E.C., K.L.M., M.S., D.J.F., C.N.M.); Department of Medicine (S.J.C., C.N.M.) and Heart Research Follow-Up Program (S.M.), Division of Cardiology, University of Rochester School of Medicine, NY; and Department of Cardiology Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston (N.-T.L., J.-i.A.)
| | - Kristina L Modjeski
- From Aab Cardiovascular Research Institute, University of Rochester School of Medicine, NY (S.J.C., S.K.T., D.M., E.C., K.L.M., M.S., D.J.F., C.N.M.); Department of Medicine (S.J.C., C.N.M.) and Heart Research Follow-Up Program (S.M.), Division of Cardiology, University of Rochester School of Medicine, NY; and Department of Cardiology Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston (N.-T.L., J.-i.A.)
| | - Scott McNitt
- From Aab Cardiovascular Research Institute, University of Rochester School of Medicine, NY (S.J.C., S.K.T., D.M., E.C., K.L.M., M.S., D.J.F., C.N.M.); Department of Medicine (S.J.C., C.N.M.) and Heart Research Follow-Up Program (S.M.), Division of Cardiology, University of Rochester School of Medicine, NY; and Department of Cardiology Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston (N.-T.L., J.-i.A.)
| | - Michael Seaberry
- From Aab Cardiovascular Research Institute, University of Rochester School of Medicine, NY (S.J.C., S.K.T., D.M., E.C., K.L.M., M.S., D.J.F., C.N.M.); Department of Medicine (S.J.C., C.N.M.) and Heart Research Follow-Up Program (S.M.), Division of Cardiology, University of Rochester School of Medicine, NY; and Department of Cardiology Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston (N.-T.L., J.-i.A.)
| | - David J Field
- From Aab Cardiovascular Research Institute, University of Rochester School of Medicine, NY (S.J.C., S.K.T., D.M., E.C., K.L.M., M.S., D.J.F., C.N.M.); Department of Medicine (S.J.C., C.N.M.) and Heart Research Follow-Up Program (S.M.), Division of Cardiology, University of Rochester School of Medicine, NY; and Department of Cardiology Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston (N.-T.L., J.-i.A.)
| | - Nhat-Tu Le
- From Aab Cardiovascular Research Institute, University of Rochester School of Medicine, NY (S.J.C., S.K.T., D.M., E.C., K.L.M., M.S., D.J.F., C.N.M.); Department of Medicine (S.J.C., C.N.M.) and Heart Research Follow-Up Program (S.M.), Division of Cardiology, University of Rochester School of Medicine, NY; and Department of Cardiology Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston (N.-T.L., J.-i.A.)
| | - Jun-Ichi Abe
- From Aab Cardiovascular Research Institute, University of Rochester School of Medicine, NY (S.J.C., S.K.T., D.M., E.C., K.L.M., M.S., D.J.F., C.N.M.); Department of Medicine (S.J.C., C.N.M.) and Heart Research Follow-Up Program (S.M.), Division of Cardiology, University of Rochester School of Medicine, NY; and Department of Cardiology Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston (N.-T.L., J.-i.A.)
| | - Craig N Morrell
- From Aab Cardiovascular Research Institute, University of Rochester School of Medicine, NY (S.J.C., S.K.T., D.M., E.C., K.L.M., M.S., D.J.F., C.N.M.); Department of Medicine (S.J.C., C.N.M.) and Heart Research Follow-Up Program (S.M.), Division of Cardiology, University of Rochester School of Medicine, NY; and Department of Cardiology Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston (N.-T.L., J.-i.A.).
| |
Collapse
|