1
|
Stulpinas A, Sereika M, Vitkeviciene A, Imbrasaite A, Krestnikova N, Kalvelyte AV. Crosstalk between protein kinases AKT and ERK1/2 in human lung tumor-derived cell models. Front Oncol 2023; 12:1045521. [PMID: 36686779 PMCID: PMC9848735 DOI: 10.3389/fonc.2022.1045521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/28/2022] [Indexed: 01/06/2023] Open
Abstract
There is no doubt that cell signaling manipulation is a key strategy for anticancer therapy. Furthermore, cell state determines drug response. Thus, establishing the relationship between cell state and therapeutic sensitivity is essential for the development of cancer therapies. In the era of personalized medicine, the use of patient-derived ex vivo cell models is a promising approach in the translation of key research findings into clinics. Here, we were focused on the non-oncogene dependencies of cell resistance to anticancer treatments. Signaling-related mechanisms of response to inhibitors of MEK/ERK and PI3K/AKT pathways (regulators of key cellular functions) were investigated using a panel of patients' lung tumor-derived cell lines with various stemness- and EMT-related markers, varying degrees of ERK1/2 and AKT phosphorylation, and response to anticancer treatment. The study of interactions between kinases was the goal of our research. Although MEK/ERK and PI3K/AKT interactions are thought to be cell line-specific, where oncogenic mutations have a decisive role, we demonstrated negative feedback loops between MEK/ERK and PI3K/AKT signaling pathways in all cell lines studied, regardless of genotype and phenotype differences. Our work showed that various and distinct inhibitors of ERK signaling - selumetinib, trametinib, and SCH772984 - increased AKT phosphorylation, and conversely, inhibitors of AKT - capivasertib, idelalisib, and AKT inhibitor VIII - increased ERK phosphorylation in both control and cisplatin-treated cells. Interaction between kinases, however, was dependent on cellular state. The feedback between ERK and AKT was attenuated by the focal adhesion kinase inhibitor PF573228, and in cells grown in suspension, showing the possible role of extracellular contacts in the regulation of crosstalk between kinases. Moreover, studies have shown that the interplay between MEK/ERK and PI3K/AKT signaling pathways may be dependent on the strength of the chemotherapeutic stimulus. The study highlights the importance of spatial location of the cells and the strength of the treatment during anticancer therapy.
Collapse
|
2
|
Sommariva S, Caviglia G, Ravera S, Frassoni F, Benvenuto F, Tortolina L, Castagnino N, Parodi S, Piana M. Computational quantification of global effects induced by mutations and drugs in signaling networks of colorectal cancer cells. Sci Rep 2021; 11:19602. [PMID: 34599254 PMCID: PMC8486743 DOI: 10.1038/s41598-021-99073-7] [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: 04/29/2021] [Accepted: 09/13/2021] [Indexed: 11/09/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most deadly and commonly diagnosed tumors worldwide. Several genes are involved in its development and progression. The most frequent mutations concern APC, KRAS, SMAD4, and TP53 genes, suggesting that CRC relies on the concomitant alteration of the related pathways. However, with classic molecular approaches, it is not easy to simultaneously analyze the interconnections between these pathways. To overcome this limitation, recently these pathways have been included in a huge chemical reaction network (CRN) describing how information sensed from the environment by growth factors is processed by healthy colorectal cells. Starting from this CRN, we propose a computational model which simulates the effects induced by single or multiple concurrent mutations on the global signaling network. The model has been tested in three scenarios. First, we have quantified the changes induced on the concentration of the proteins of the network by a mutation in APC, KRAS, SMAD4, or TP53. Second, we have computed the changes in the concentration of p53 induced by up to two concurrent mutations affecting proteins upstreams in the network. Third, we have considered a mutated cell affected by a gain of function of KRAS, and we have simulated the action of Dabrafenib, showing that the proposed model can be used to determine the most effective amount of drug to be delivered to the cell. In general, the proposed approach displays several advantages, in that it allows to quantify the alteration in the concentration of the proteins resulting from a single or multiple given mutations. Moreover, simulations of the global signaling network of CRC may be used to identify new therapeutic targets, or to disclose unexpected interactions between the involved pathways.
Collapse
Affiliation(s)
- Sara Sommariva
- Dipartimento di Matematica, Università di Genova, via Dodecaneso 35, 16146, Genoa, Italy.
| | - Giacomo Caviglia
- Dipartimento di Matematica, Università di Genova, via Dodecaneso 35, 16146, Genoa, Italy
| | - Silvia Ravera
- Dipartimento di Medicina Sperimentale, Università di Genova, Via De Toni 14, 16132, Genoa, Italy
| | - Francesco Frassoni
- Dipartimento di Matematica, Università di Genova, via Dodecaneso 35, 16146, Genoa, Italy
| | - Federico Benvenuto
- Dipartimento di Matematica, Università di Genova, via Dodecaneso 35, 16146, Genoa, Italy
| | - Lorenzo Tortolina
- Dipartimento di Medicina Interna, Università di Genova, via Leon Battista Alberti 2, 16132, Genoa, Italy
| | - Nicoletta Castagnino
- Dipartimento di Medicina Interna, Università di Genova, via Leon Battista Alberti 2, 16132, Genoa, Italy
| | - Silvio Parodi
- Dipartimento di Medicina Interna, Università di Genova, via Leon Battista Alberti 2, 16132, Genoa, Italy
| | - Michele Piana
- Dipartimento di Matematica, Università di Genova, via Dodecaneso 35, 16146, Genoa, Italy
| |
Collapse
|
3
|
Vališ K, Novák P. Targeting ERK-Hippo Interplay in Cancer Therapy. Int J Mol Sci 2020; 21:ijms21093236. [PMID: 32375238 PMCID: PMC7247570 DOI: 10.3390/ijms21093236] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023] Open
Abstract
Extracellular signal-regulated kinase (ERK) is a part of the mitogen-activated protein kinase (MAPK) signaling pathway which allows the transduction of various cellular signals to final effectors and regulation of elementary cellular processes. Deregulation of the MAPK signaling occurs under many pathological conditions including neurodegenerative disorders, metabolic syndromes and cancers. Targeted inhibition of individual kinases of the MAPK signaling pathway using synthetic compounds represents a promising way to effective anti-cancer therapy. Cross-talk of the MAPK signaling pathway with other proteins and signaling pathways have a crucial impact on clinical outcomes of targeted therapies and plays important role during development of drug resistance in cancers. We discuss cross-talk of the MAPK/ERK signaling pathway with other signaling pathways, in particular interplay with the Hippo/MST pathway. We demonstrate the mechanism of cell death induction shared between MAPK/ERK and Hippo/MST signaling pathways and discuss the potential of combination targeting of these pathways in the development of more effective anti-cancer therapies.
Collapse
Affiliation(s)
- Karel Vališ
- Correspondence: (K.V.); (P.N.); Tel.: +420-325873610 (P.N.)
| | - Petr Novák
- Correspondence: (K.V.); (P.N.); Tel.: +420-325873610 (P.N.)
| |
Collapse
|
4
|
Marková I, Koníčková R, Vaňková K, Leníček M, Kolář M, Strnad H, Hradilová M, Šáchová J, Rasl J, Klímová Z, Vomastek T, Němečková I, Nachtigal P, Vítek L. Anti-angiogenic effects of the blue-green alga Arthrospira platensis on pancreatic cancer. J Cell Mol Med 2020; 24:2402-2415. [PMID: 31957261 PMCID: PMC7028863 DOI: 10.1111/jcmm.14922] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 12/06/2019] [Accepted: 12/10/2019] [Indexed: 02/06/2023] Open
Abstract
Arthrospira platensis, a blue-green alga, is a popular nutraceutical substance having potent antioxidant properties with potential anti-carcinogenic activities. The aim of our study was to assess the possible anti-angiogenic effects of A platensis in an experimental model of pancreatic cancer. The effects of an A platensis extract were investigated on human pancreatic cancer cells (PA-TU-8902) and immortalized endothelial-like cells (Ea.hy926). PA-TU-8902 pancreatic tumours xenografted to athymic mice were also examined. In vitro migration and invasiveness assays were performed on the tested cells. Multiple angiogenic factors and signalling pathways were analysed in the epithelial, endothelial and cancer cells, and tumour tissue. The A platensis extract exerted inhibitory effects on both migration and invasion of pancreatic cancer as well as endothelial-like cells. Tumours of mice treated with A platensis exhibited much lesser degrees of vascularization as measured by CD31 immunostaining (P = .004). Surprisingly, the VEGF-A mRNA and protein expressions were up-regulated in pancreatic cancer cells. A platensis inhibited ERK activation upstream of Raf and suppressed the expression of ERK-regulated proteins. Treatment of pancreatic cancer with A platensis was associated with suppressive effects on migration and invasiveness with various anti-angiogenic features, which might account for the anticancer effects of this blue-green alga.
Collapse
Affiliation(s)
- Ivana Marková
- Institute of Medical Biochemistry and Laboratory DiagnosticsFaculty General Hospital and 1st Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Renata Koníčková
- Institute of Medical Biochemistry and Laboratory DiagnosticsFaculty General Hospital and 1st Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Kateřina Vaňková
- Institute of Medical Biochemistry and Laboratory DiagnosticsFaculty General Hospital and 1st Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Martin Leníček
- Institute of Medical Biochemistry and Laboratory DiagnosticsFaculty General Hospital and 1st Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Michal Kolář
- Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
- Department of Informatics and ChemistryFaculty of Chemical TechnologyUniversity of Chemistry and TechnologyPragueCzech Republic
| | - Hynek Strnad
- Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Miluše Hradilová
- Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Jana Šáchová
- Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Jan Rasl
- Institute of Microbiology of the Czech Academy of SciencesPragueCzech Republic
- Department of Cell BiologyFaculty of ScienceCharles UniversityPragueCzech Republic
| | - Zuzana Klímová
- Institute of Microbiology of the Czech Academy of SciencesPragueCzech Republic
| | - Tomáš Vomastek
- Institute of Microbiology of the Czech Academy of SciencesPragueCzech Republic
| | - Ivana Němečková
- Department of Biological and Medical SciencesFaculty of Pharmacy in Hradec KraloveCharles UniversityHradec KrálovéCzech Republic
| | - Petr Nachtigal
- Department of Biological and Medical SciencesFaculty of Pharmacy in Hradec KraloveCharles UniversityHradec KrálovéCzech Republic
| | - Libor Vítek
- Institute of Medical Biochemistry and Laboratory DiagnosticsFaculty General Hospital and 1st Faculty of MedicineCharles UniversityPragueCzech Republic
- 4th Department of Internal MedicineFaculty General Hospital and 1st Faculty of MedicineCharles UniversityPragueCzech Republic
| |
Collapse
|
5
|
Baur F, Nietzer SL, Kunz M, Saal F, Jeromin J, Matschos S, Linnebacher M, Walles H, Dandekar T, Dandekar G. Connecting Cancer Pathways to Tumor Engines: A Stratification Tool for Colorectal Cancer Combining Human In Vitro Tissue Models with Boolean In Silico Models. Cancers (Basel) 2019; 12:cancers12010028. [PMID: 31861874 PMCID: PMC7017315 DOI: 10.3390/cancers12010028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/13/2019] [Accepted: 12/16/2019] [Indexed: 02/06/2023] Open
Abstract
To improve and focus preclinical testing, we combine tumor models based on a decellularized tissue matrix with bioinformatics to stratify tumors according to stage-specific mutations that are linked to central cancer pathways. We generated tissue models with BRAF-mutant colorectal cancer (CRC) cells (HROC24 and HROC87) and compared treatment responses to two-dimensional (2D) cultures and xenografts. As the BRAF inhibitor vemurafenib is-in contrast to melanoma-not effective in CRC, we combined it with the EGFR inhibitor gefitinib. In general, our 3D models showed higher chemoresistance and in contrast to 2D a more active HGFR after gefitinib and combination-therapy. In xenograft models murine HGF could not activate the human HGFR, stressing the importance of the human microenvironment. In order to stratify patient groups for targeted treatment options in CRC, an in silico topology with different stages including mutations and changes in common signaling pathways was developed. We applied the established topology for in silico simulations to predict new therapeutic options for BRAF-mutated CRC patients in advanced stages. Our in silico tool connects genome information with a deeper understanding of tumor engines in clinically relevant signaling networks which goes beyond the consideration of single drivers to improve CRC patient stratification.
Collapse
Affiliation(s)
- Florentin Baur
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany; (F.B.); (S.L.N.); (H.W.)
| | - Sarah L. Nietzer
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany; (F.B.); (S.L.N.); (H.W.)
- Fraunhofer Institute for Silicate Research (ISC), Translational Center Regenerative Therapies, Röntgenring 11, 97070 Würzburg, Germany
| | - Meik Kunz
- Chair of Medical Informatics, Friedrich-Alexander University of Erlangen-Nürnberg, 91058 Erlangen, Germany;
- Department of Bioinformatics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany; (F.S.); (J.J.)
| | - Fabian Saal
- Department of Bioinformatics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany; (F.S.); (J.J.)
| | - Julian Jeromin
- Department of Bioinformatics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany; (F.S.); (J.J.)
| | - Stephanie Matschos
- Department of Surgery, Molecular Oncology and Immunotherapy, University Medical Center Rostock, Schillingallee 35, 18057 Rostock, Germany; (S.M.); (M.L.)
| | - Michael Linnebacher
- Department of Surgery, Molecular Oncology and Immunotherapy, University Medical Center Rostock, Schillingallee 35, 18057 Rostock, Germany; (S.M.); (M.L.)
| | - Heike Walles
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany; (F.B.); (S.L.N.); (H.W.)
- Fraunhofer Institute for Silicate Research (ISC), Translational Center Regenerative Therapies, Röntgenring 11, 97070 Würzburg, Germany
| | - Thomas Dandekar
- Department of Bioinformatics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany; (F.S.); (J.J.)
- EMBL Heidelberg, Structural and Computational Biology, Meyerhofstraße 1, 69117 Heidelberg, Germany
- Correspondence: (T.D.); (G.D.); Tel.: +49-931-3184551 (T.D.); +49-931-3182597 (G.D.)
| | - Gudrun Dandekar
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany; (F.B.); (S.L.N.); (H.W.)
- Fraunhofer Institute for Silicate Research (ISC), Translational Center Regenerative Therapies, Röntgenring 11, 97070 Würzburg, Germany
- Correspondence: (T.D.); (G.D.); Tel.: +49-931-3184551 (T.D.); +49-931-3182597 (G.D.)
| |
Collapse
|
6
|
Lan L, Wang H, Yang R, Liu F, Bi Q, Wang S, Wei X, Yan H, Su R. R2-8018 reduces the proliferation and migration of non-small cell lung cancer cells by disturbing transactivation between M3R and EGFR. Life Sci 2019; 234:116742. [PMID: 31401315 DOI: 10.1016/j.lfs.2019.116742] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/03/2019] [Accepted: 08/07/2019] [Indexed: 01/23/2023]
Abstract
AIMS The M3 muscarinic acetylcholine receptor (M3R) is a G protein-coupled receptor that is expressed in cases of non-small cell lung cancer (NSCLC). Previous studies demonstrated that M3R antagonists reduce the proliferation of NSCLC. However, how antagonists inhibit the NSCLC proliferation and migration is still little known. This study aims to investigate the mechanism of M3R involved in the growth of NSCLC. MAIN METHODS The CRISPR/Cas9 was used to knock out (KO) the M3R gene. A real-time cell analyzer (RTCA) was used to record the proliferation of NSCLC cells. The migration and cell cycle of NSCLC cells were evaluated with scratch test and flow cytometry (FCM), respectively. Antibody microarray analysis was performed to detect the expression of proteins after antagonizing M3R and knocking out of M3R, subsequently some of these important proteins were verified by western blot. KEY FINDINGS The proliferation and migration of NSCLC cells were inhibited by M3R antagonist R2-8018 and knocking out of M3R. Antagonism or knocking out of M3R reduced the phosphorylation of EGFR. Moreover, c-Src and β-arrestin-1 are involved in the mechanism of how the inhibition of M3R affects EGFR in NSCLC. Further study demonstrated that PI3K/AKT and MEK/ERK signal pathways are involved in M3R-induced EGFR transactivation in NSCLC, and the molecules involved in the cell cycle progression and migration of NSCLC cells were identified. SIGNIFICANCE This further understanding of the relationship between M3R and NSCLC facilitates the design of therapeutic strategy with M3R antagonist as an adjuvant drug for NSCLC treatment.
Collapse
Affiliation(s)
- Liting Lan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China
| | - Hua Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China; 69242 Force Health Center, No. 1, Hongxing Road, Turpan, Xinjiang 838000, China
| | - Rui Yang
- National Institutes for Food and Drug Control, No. 31, Huatuo Road, Daxing District, Beijing 102629, China
| | - Fengqi Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China; Department of Medical Laboratory, Changzhi Medical College, No.161 Jiefang East Street, Changzhi, Shanxi, 046000, China
| | - Qingshang Bi
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China; Department of Medical Laboratory, Changzhi Medical College, No.161 Jiefang East Street, Changzhi, Shanxi, 046000, China
| | - Shiqi Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China; Center for Drug Evaluation, NMPA. No. 128, Jianguo Road, Chaoyang District, Beijing 100022, China
| | - Xiaoli Wei
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China
| | - Haitao Yan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China.
| | - Ruibin Su
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Department of Biochemical Pharmacology, Beijing Institute of Pharmacology and Toxicology, 27th Taiping Road, Beijing 100850, China.
| |
Collapse
|
7
|
Bankoglu EE, Kodandaraman G, Stopper H. A systematic review of the use of the alkaline comet assay for genotoxicity studies in human colon-derived cells. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2019; 845:402976. [DOI: 10.1016/j.mrgentox.2018.10.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 10/21/2018] [Accepted: 10/28/2018] [Indexed: 12/18/2022]
|
8
|
Perspectives of RAS and RHEB GTPase Signaling Pathways in Regenerating Brain Neurons. Int J Mol Sci 2018; 19:ijms19124052. [PMID: 30558189 PMCID: PMC6321366 DOI: 10.3390/ijms19124052] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/05/2018] [Accepted: 12/13/2018] [Indexed: 12/29/2022] Open
Abstract
Cellular activation of RAS GTPases into the GTP-binding “ON” state is a key switch for regulating brain functions. Molecular protein structural elements of rat sarcoma (RAS) and RAS homolog protein enriched in brain (RHEB) GTPases involved in this switch are discussed including their subcellular membrane localization for triggering specific signaling pathways resulting in regulation of synaptic connectivity, axonal growth, differentiation, migration, cytoskeletal dynamics, neural protection, and apoptosis. A beneficial role of neuronal H-RAS activity is suggested from cellular and animal models of neurodegenerative diseases. Recent experiments on optogenetic regulation offer insights into the spatiotemporal aspects controlling RAS/mitogen activated protein kinase (MAPK) or phosphoinositide-3 kinase (PI3K) pathways. As optogenetic manipulation of cellular signaling in deep brain regions critically requires penetration of light through large distances of absorbing tissue, we discuss magnetic guidance of re-growing axons as a complementary approach. In Parkinson’s disease, dopaminergic neuronal cell bodies degenerate in the substantia nigra. Current human trials of stem cell-derived dopaminergic neurons must take into account the inability of neuronal axons navigating over a large distance from the grafted site into striatal target regions. Grafting dopaminergic precursor neurons directly into the degenerating substantia nigra is discussed as a novel concept aiming to guide axonal growth by activating GTPase signaling through protein-functionalized intracellular magnetic nanoparticles responding to external magnets.
Collapse
|
9
|
Park J, Jang JH, Kim JH. Mediatory role of BLT2 in the proliferation of KRAS mutant colorectal cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:329-336. [PMID: 30553812 DOI: 10.1016/j.bbamcr.2018.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/22/2018] [Accepted: 12/09/2018] [Indexed: 02/02/2023]
Abstract
Inflammatory lipid mediators play various roles in colorectal cancer progression through complex pathways. However, the mechanism by which lipoxygenase-derived inflammatory lipid mediators contribute to colorectal cancer progression remains elusive. In this study, we found that BLT2, a cell surface GPCR for leukotriene B4 and 12‑hydroxyeicosatetraenoic acid, is highly upregulated in KRAS mutant LOVO and SW480 colorectal cancer cells and plays critical roles in mediating proliferation through activation of phosphatidylinositol 3‑kinase (PI3K)/protein kinase B (Akt) and subsequent upregulation of cyclin D1. Exposure to BLT2 siRNA or LY255283, a specific BLT2 inhibitor, clearly suppressed the proliferation of KRAS mutant colorectal cancer cells and markedly increased cell cycle arrest by downregulating the PI3K/Akt-cyclin D1 cascade. Xenograft tumor formation by LOVO and SW480 cells in athymic mice was also substantially reduced by treatment with the BLT2 inhibitor in vivo. Together, our study demonstrates that BLT2 is necessary for the proliferation of LOVO and SW480 cells and thus may be a potential therapeutic target for the treatment of KRAS mutant colorectal cancer.
Collapse
Affiliation(s)
- JaeIn Park
- School of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Republic of Korea
| | - Jae-Hyun Jang
- School of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Republic of Korea
| | - Jae-Hong Kim
- School of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Republic of Korea.
| |
Collapse
|
10
|
Fritsche-Guenther R, Zasada C, Mastrobuoni G, Royla N, Rainer R, Roßner F, Pietzke M, Klipp E, Sers C, Kempa S. Alterations of mTOR signaling impact metabolic stress resistance in colorectal carcinomas with BRAF and KRAS mutations. Sci Rep 2018; 8:9204. [PMID: 29907857 PMCID: PMC6003911 DOI: 10.1038/s41598-018-27394-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/23/2018] [Indexed: 11/18/2022] Open
Abstract
Metabolic reprogramming is as a hallmark of cancer, and several studies have reported that BRAF and KRAS tumors may be accompanied by a deregulation of cellular metabolism. We investigated how BRAFV600E and KRASG12V affect cell metabolism, stress resistance and signaling in colorectal carcinoma cells driven by these mutations. KRASG12V expressing cells are characterized by the induction of glycolysis, accumulation of lactic acid and sensitivity to glycolytic inhibition. Notably mathematical modelling confirmed the critical role of MCT1 designating the survival of KRASG12V cells. Carcinoma cells harboring BRAFV600E remain resistant towards alterations of glucose supply or application of signaling or metabolic inhibitors. Altogether these data demonstrate that an oncogene-specific decoupling of mTOR from AMPK or AKT signaling accounts for alterations of resistance mechanisms and metabolic phenotypes. Indeed the inhibition of mTOR in BRAFV600E cells counteracts the metabolic predisposition and demonstrates mTOR as a potential target in BRAFV600E-driven colorectal carcinomas.
Collapse
Affiliation(s)
- Raphaela Fritsche-Guenther
- Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute of Health (BIH), Robert-Roessle-Str. 10, 13125, Berlin, Germany
| | - Christin Zasada
- Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Robert-Roessle-Str. 10, 13125, Berlin, Germany
| | - Guido Mastrobuoni
- Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Robert-Roessle-Str. 10, 13125, Berlin, Germany
| | - Nadine Royla
- Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Robert-Roessle-Str. 10, 13125, Berlin, Germany
| | - Roman Rainer
- Humboldt University Berlin, Theoretical Biophysics, Invalidenstraße 42, 10115, Berlin, Germany
| | - Florian Roßner
- Charité Universitätsmedizin, Institute of Pathology, Chariteplatz 1, 10117, Berlin, Germany
| | - Matthias Pietzke
- Beatson Institute, Switchback Road, Bearsden, Glasgow, G61 1BD, United Kingdom
| | - Edda Klipp
- Charité Universitätsmedizin, Institute of Pathology, Chariteplatz 1, 10117, Berlin, Germany
| | - Christine Sers
- Charité Universitätsmedizin, Institute of Pathology, Chariteplatz 1, 10117, Berlin, Germany
| | - Stefan Kempa
- Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute of Health (BIH), Robert-Roessle-Str. 10, 13125, Berlin, Germany. .,Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Robert-Roessle-Str. 10, 13125, Berlin, Germany.
| |
Collapse
|
11
|
KRAS mutant allele-specific expression knockdown in pancreatic cancer model with systemically delivered bi-shRNA KRAS lipoplex. PLoS One 2018; 13:e0193644. [PMID: 29851957 PMCID: PMC5979018 DOI: 10.1371/journal.pone.0193644] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 02/15/2018] [Indexed: 12/18/2022] Open
Abstract
The KRAS oncogene, present in over 90% of pancreatic ductal adenocarcinomas, is most frequently the result of one of three gain-of-function substitution mutations of codon 12 glycine. Thus far, RAS mutations have been clinically refractory to both direct and selective inhibition by systemic therapeutics. This report presents the results of pre-clinical assessment of a lipoplex comprising a plasmid-encoded, modular bi-functional shRNA (bi-shRNA), which executes selective and multi-mutant allelic KRASG12mut gene silencing, encased within a fusogenic liposome systemic delivery vehicle. Using both a dual luciferase reporter system and a Restriction Fragment Length Polymorphism (RFLP) assay, selective discrimination of KRASG12mut from KRASwt was confirmed in vitro in PANC1 cells. Subsequently, systemic administration of the bi-shRNAKRAS fusogenic lipoplex into female athymic Nu/Nu mice bearing PANC1 xenografts demonstrated intratumoral plasmid delivery, KRASG12mut knockdown, and inhibition of tumor growth, without adverse effect. Clinical trials with the bi-shRNA lipoplex have been implemented.
Collapse
|
12
|
Molnár E, Rittler D, Baranyi M, Grusch M, Berger W, Döme B, Tóvári J, Aigner C, Tímár J, Garay T, Hegedűs B. Pan-RAF and MEK vertical inhibition enhances therapeutic response in non-V600 BRAF mutant cells. BMC Cancer 2018; 18:542. [PMID: 29739364 PMCID: PMC5941622 DOI: 10.1186/s12885-018-4455-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 04/30/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Currently, there are no available targeted therapy options for non-V600 BRAF mutated tumors. The aim of this study was to investigate the effects of RAF and MEK concurrent inhibition on tumor growth, migration, signaling and apoptosis induction in preclinical models of non-V600 BRAF mutant tumor cell lines. METHODS Six BRAF mutated human tumor cell lines CRL5885 (G466 V), WM3629 (D594G), WM3670 (G469E), MDAMB231 (G464 V), CRL5922 (L597 V) and A375 (V600E as control) were investigated. Pan-RAF inhibitor (sorafenib or AZ628) and MEK inhibitor (selumetinib) or their combination were used in in vitro viability, video microscopy, immunoblot, cell cycle and TUNEL assays. The in vivo effects of the drugs were assessed in an orthotopic NSG mouse breast cancer model. RESULTS All cell lines showed a significant growth inhibition with synergism in the sorafenib/AZ628 and selumetinib combination. Combination treatment resulted in higher Erk1/2 inhibition and in increased induction of apoptosis when compared to single agent treatments. However, single selumetinib treatment could cause adverse therapeutic effects, like increased cell migration in certain cells, selumetinib and sorafenib combination treatment lowered migratory capacity in all the cell lines. Importantly, combination resulted in significantly increased tumor growth inhibition in orthotropic xenografts of MDAMB231 cells when compared to sorafenib - but not to selumetinib - treatment. CONCLUSIONS Our data suggests that combined blocking of RAF and MEK may achieve increased therapeutic response in non-V600 BRAF mutant tumors.
Collapse
Affiliation(s)
- Eszter Molnár
- 2nd Department of Pathology, Semmelweis University, Budapest, 1091, Hungary
| | - Dominika Rittler
- 2nd Department of Pathology, Semmelweis University, Budapest, 1091, Hungary
| | - Marcell Baranyi
- 2nd Department of Pathology, Semmelweis University, Budapest, 1091, Hungary
| | - Michael Grusch
- Institute of Cancer Research, Medical University of Vienna, 1090, Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research, Medical University of Vienna, 1090, Vienna, Austria
| | - Balázs Döme
- Department of Thoracic Surgery, Medical University of Vienna, 1090, Vienna, Austria.,National Korányi Institute of TB and Pulmonology, Budapest, 1085, Hungary.,Department of Thoracic Surgery, Semmelweis University-National Institute of Oncology, Budapest, 1122, Hungary
| | - József Tóvári
- Department of Experimental Pharmacology, National Institute of Oncology, Budapest, 1122, Hungary
| | - Clemens Aigner
- Department of Thoracic Surgery, Ruhrlandklinik, University Duisburg-Essen, 45239, Essen, Germany
| | - József Tímár
- 2nd Department of Pathology, Semmelweis University, Budapest, 1091, Hungary.,HAS-SE Molecular Oncology Research Group, Hungarian Academy of Sciences, Budapest, 1051, Hungary
| | - Tamás Garay
- 2nd Department of Pathology, Semmelweis University, Budapest, 1091, Hungary.,HAS-SE Molecular Oncology Research Group, Hungarian Academy of Sciences, Budapest, 1051, Hungary.,HAS Postdoctoral Fellowship Program Hungarian Academy of Sciences, Budapest, 1051, Hungary
| | - Balázs Hegedűs
- 2nd Department of Pathology, Semmelweis University, Budapest, 1091, Hungary. .,Department of Thoracic Surgery, Ruhrlandklinik, University Duisburg-Essen, 45239, Essen, Germany. .,HAS-SE Molecular Oncology Research Group, Hungarian Academy of Sciences, Budapest, 1051, Hungary.
| |
Collapse
|
13
|
Advantages, Disadvantages and Modifications of Conventional ELISA. SPRINGERBRIEFS IN APPLIED SCIENCES AND TECHNOLOGY 2018. [DOI: 10.1007/978-981-10-6766-2_5] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
14
|
Kauke MJ, Traxlmayr MW, Parker JA, Kiefer JD, Knihtila R, McGee J, Verdine G, Mattos C, Wittrup KD. An engineered protein antagonist of K-Ras/B-Raf interaction. Sci Rep 2017; 7:5831. [PMID: 28724936 PMCID: PMC5517481 DOI: 10.1038/s41598-017-05889-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/05/2017] [Indexed: 12/31/2022] Open
Abstract
Ras is at the hub of signal transduction pathways controlling cell proliferation and survival. Its mutants, present in about 30% of human cancers, are major drivers of oncogenesis and render tumors unresponsive to standard therapies. Here we report the engineering of a protein scaffold for preferential binding to K-Ras G12D. This is the first reported inhibitor to achieve nanomolar affinity while exhibiting specificity for mutant over wild type (WT) K-Ras. Crystal structures of the protein R11.1.6 in complex with K-Ras WT and K-Ras G12D offer insight into the structural basis for specificity, highlighting differences in the switch I conformation as the major defining element in the higher affinity interaction. R11.1.6 directly blocks interaction with Raf and reduces signaling through the Raf/MEK/ERK pathway. Our results support greater consideration of the state of switch I and provide a novel tool to study Ras biology. Most importantly, this work makes an unprecedented contribution to Ras research in inhibitor development strategy by revealing details of a targetable binding surface. Unlike the polar interfaces found for Ras/effector interactions, the K-Ras/R11.1.6 complex reveals an extensive hydrophobic interface that can serve as a template to advance the development of high affinity, non-covalent inhibitors of K-Ras oncogenic mutants.
Collapse
Affiliation(s)
- Monique J Kauke
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Michael W Traxlmayr
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Jillian A Parker
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, 02115, USA
| | - Jonathan D Kiefer
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Ryan Knihtila
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, 02115, USA
| | - John McGee
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Greg Verdine
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, 02138, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA.,Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Carla Mattos
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, 02115, USA
| | - K Dane Wittrup
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA. .,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA. .,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
15
|
Uhlitz F, Sieber A, Wyler E, Fritsche-Guenther R, Meisig J, Landthaler M, Klinger B, Blüthgen N. An immediate-late gene expression module decodes ERK signal duration. Mol Syst Biol 2017; 13:928. [PMID: 28468958 PMCID: PMC5448165 DOI: 10.15252/msb.20177554] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The RAF‐MEK‐ERK signalling pathway controls fundamental, often opposing cellular processes such as proliferation and apoptosis. Signal duration has been identified to play a decisive role in these cell fate decisions. However, it remains unclear how the different early and late responding gene expression modules can discriminate short and long signals. We obtained both protein phosphorylation and gene expression time course data from HEK293 cells carrying an inducible construct of the proto‐oncogene RAF. By mathematical modelling, we identified a new gene expression module of immediate–late genes (ILGs) distinct in gene expression dynamics and function. We find that mRNA longevity enables these ILGs to respond late and thus translate ERK signal duration into response amplitude. Despite their late response, their GC‐rich promoter structure suggested and metabolic labelling with 4SU confirmed that transcription of ILGs is induced immediately. A comparative analysis shows that the principle of duration decoding is conserved in PC12 cells and MCF7 cells, two paradigm cell systems for ERK signal duration. Altogether, our findings suggest that ILGs function as a gene expression module to decode ERK signal duration.
Collapse
Affiliation(s)
- Florian Uhlitz
- IRI Life Sciences & Institute for Theoretical Biology, Humboldt Universität Berlin, Berlin, Germany.,Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anja Sieber
- IRI Life Sciences & Institute for Theoretical Biology, Humboldt Universität Berlin, Berlin, Germany.,Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Raphaela Fritsche-Guenther
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Johannes Meisig
- IRI Life Sciences & Institute for Theoretical Biology, Humboldt Universität Berlin, Berlin, Germany.,Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Bertram Klinger
- IRI Life Sciences & Institute for Theoretical Biology, Humboldt Universität Berlin, Berlin, Germany.,Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nils Blüthgen
- IRI Life Sciences & Institute for Theoretical Biology, Humboldt Universität Berlin, Berlin, Germany .,Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
16
|
Riemer P, Rydenfelt M, Marks M, van Eunen K, Thedieck K, Herrmann BG, Blüthgen N, Sers C, Morkel M. Oncogenic β-catenin and PIK3CA instruct network states and cancer phenotypes in intestinal organoids. J Cell Biol 2017; 216:1567-1577. [PMID: 28442534 PMCID: PMC5461020 DOI: 10.1083/jcb.201610058] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 02/08/2017] [Accepted: 03/30/2017] [Indexed: 01/25/2023] Open
Abstract
Colorectal cancer is driven by cooperating oncogenic mutations. In this study, we use organotypic cultures derived from transgenic mice inducibly expressing oncogenic β-catenin and/or PIK3CAH1047R to follow sequential changes in cancer-related signaling networks, intestinal cell metabolism, and physiology in a three-dimensional environment mimicking tissue architecture. Activation of β-catenin alone results in the formation of highly clonogenic cells that are nonmotile and prone to undergo apoptosis. In contrast, coexpression of stabilized β-catenin and PIK3CAH1047R gives rise to intestinal cells that are apoptosis-resistant, proliferative, stem cell-like, and motile. Systematic inhibitor treatments of organoids followed by quantitative phenotyping and phosphoprotein analyses uncover key changes in the signaling network topology of intestinal cells after induction of stabilized β-catenin and PIK3CAH1047R We find that survival and motility of organoid cells are associated with 4EBP1 and AKT phosphorylation, respectively. Our work defines phenotypes, signaling network states, and vulnerabilities of transgenic intestinal organoids as a novel approach to understanding oncogene activities and guiding the development of targeted therapies.
Collapse
Affiliation(s)
- Pamela Riemer
- Laboratory of Molecular Tumor Pathology, Institute of Pathology, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany.,German Cancer Consortium, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Mattias Rydenfelt
- Integrative Research Institute Life Sciences, Humboldt University Berlin, 10099 Berlin, Germany
| | - Matthias Marks
- Department of Developmental Genetics, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Karen van Eunen
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Kathrin Thedieck
- Department of Pediatrics, Center for Liver, Digestive and Metabolic Diseases, University Medical Center Groningen, 9713 GZ Groningen, Netherlands
| | - Bernhard G Herrmann
- Department of Developmental Genetics, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Nils Blüthgen
- Laboratory of Molecular Tumor Pathology, Institute of Pathology, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany.,Integrative Research Institute Life Sciences, Humboldt University Berlin, 10099 Berlin, Germany
| | - Christine Sers
- Laboratory of Molecular Tumor Pathology, Institute of Pathology, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany.,German Cancer Consortium, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Markus Morkel
- Laboratory of Molecular Tumor Pathology, Institute of Pathology, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|