CDK4 Regulates Lysosomal Function and Mtorc1 Activation to Promote Cancer Cell Survival

Total Page:16

File Type:pdf, Size:1020Kb

CDK4 Regulates Lysosomal Function and Mtorc1 Activation to Promote Cancer Cell Survival Author Manuscript Published OnlineFirst on August 8, 2019; DOI: 10.1158/0008-5472.CAN-19-0708 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. CDK4 regulates lysosomal function and mTORC1 activation to promote cancer cell survival Laia Martínez-Carreres1, Julien Puyal2, Lucía C. Leal-Esteban1, Meritxell Orpinell3, Judit Castillo-Armengol1, Albert Giralt1, Oleksandr Dergai1, Catherine Moret1, Valentin Barquissau1, Anita Nasrallah1, Angélique Pabois4,5, Lianjun Zhang4,6,7, Pedro Romero4, Isabel C. Lopez-Mejia1 and Lluis Fajas1* 1Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland. 2Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland. 3Department of Physiology, University of Lausanne, 1005 Lausanne, Switzerland 4Department of Fundamental Oncology, University of Lausanne, 1066 Epalinges, Switzerland. 5Ludwig Institute for Cancer Research, University of Lausanne, 1066 Epalinges, Switzerland. 6Present address: Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, 100005 Beijing, China. 7Present address: Suzhou Institute of Systems Medicine, Suzhou, Jiangsu 215123, China. *Corresponding author: Lluis Fajas, University of Lausanne, Center for Integrative Genomics, Quartier UNIL-Sorge, Bat. Genopode, CH-1015 Lausanne, Switzerland. Phone: +41.21.692.4111; E-mail: [email protected]. Declaration of interests The authors declare no competing interests. Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 8, 2019; DOI: 10.1158/0008-5472.CAN-19-0708 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Abstract Cyclin-dependent kinase 4 (CDK4) is well-known for its role in regulating the cell cycle, however, its role in cancer metabolism, especially mTOR signaling, is undefined. In this study, we established a connection between CDK4 and lysosomes, an emerging metabolic organelle crucial for mTORC1 activation. On the one hand, CDK4 phosphorylated the tumor suppressor FLCN, regulating mTORC1 recruitment to the lysosomal surface in response to amino acids. On the other hand, CDK4 directly regulated lysosomal function and was essential for lysosomal degradation, ultimately regulating mTORC1 activity. Pharmacological inhibition or genetic inactivation of CDK4, other than retaining FLCN at the lysosomal surface, led to the accumulation of undigested material inside lysosomes, which impaired the autophagic flux and induced cancer cell senescence in vitro and in xenograft models. Importantly, the use of CDK4 inhibitors in therapy is known to cause senescence but not cell death. To overcome this phenomenon and based on our findings, we increased the autophagic flux in cancer cells by using an AMPK activator in combination with a CDK4 inhibitor. The cotreatment induced autophagy (AMPK activation), and impaired lysosomal function (CDK4 inhibition), resulting in cell death and tumor regression. Altogether, we uncover a previously unknown role for CDK4 in lysosomal biology and propose a novel therapeutic strategy to target cancer cells. Statement of significance Findings uncover a novel function of CDK4 in lysosomal biology which promotes cancer progression by activating mTORC1, targeting this function offers a new therapeutic strategy for cancer treatment. 2 Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 8, 2019; DOI: 10.1158/0008-5472.CAN-19-0708 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Introduction Cyclin-dependent kinase 4 (CDK4) has a well-established role in cell cycle control (1) and CDK4-cyclin complexes are commonly deregulated in tumorigenesis (2). These complexes are of great interest as therapeutic targets, and the FDA has approved the specific CDK4/6 kinase inhibitors PD0332991 (palbociclib), LEE011 (ribociclib) and LY2835219 (abemaciclib) for treating advanced or metastatic hormone receptor (HR)-positive and HER2- negative breast cancer. Clinical studies using CDK4/6 inhibitors to treat other malignancies are being conducted (3). Research from our group and others has shown that the role of CDK4 is not limited to the control of the cell cycle. Indeed CDK4 is also a major regulator of energy homeostasis (4-6) through E2F1-RB complex (7), AMPK (8) and IRS2 (9). Importantly, the CDK4 pathway has been shown to cross-talk with the mTOR pathway, which is a major regulator of cell growth and metabolism (10,11). CDK4/6 inhibition attenuates mTOR Complex 1 (mTORC1) activity in some cancer models (12,13), yet the effects of CDK4/6 inhibitors on mTORC1 seem to be cell-type specific since opposite results were observed in other cancer types (14). The exact mechanism underlying the CDK4-mTOR cross-talk in mammals is unknown, although in Drosophila it occurs via the phosphorylation of TSC2 (15). Given that mTOR activity is increased in numerous cancers and participates in the translational regulation of several oncogenic proteins, mTOR inactivation constitutes an attractive strategy for cancer treatment (16). Lysosomes, considered for years as only the digestive system of the cell, have emerged as key effectors in cell metabolism, due to their role as platforms in the activation of mTOR pathway (17-19). mTORC1 is recruited to the surface of lysosomes in a complex amino acid (AA)-dependent manner (17). Among the multiple regulators of this process, we focused on FLCN, a tumor suppressor which functions as a complex with FNIP. The FLCN- FNIP complex interacts with Rag GTPases in the absence of AAs repressing their activity. When AAs are sensed, FLCN-FNIP complexes dissociate from Rag GTPases eliciting their activation. The activation of Rag GTPases is crucial for mTORC1 recruitment to lysosomes (20). Importantly, mTORC1 activation is also triggered by the accumulation of AAs in the lysosomal lumen (21). Therefore, alterations in the lysosomal function directly impact 3 Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 8, 2019; DOI: 10.1158/0008-5472.CAN-19-0708 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. mTORC1 activity (22,23). Additionally, these organelles play roles in cell survival and cell proliferation, thus becoming emerging targets for cancer therapy (24-26). In this study, we demonstrate that CDK4 is capable of modulating mTORC1 activity in a direct manner, through the phosphorylation of FLCN, and indirectly, by promoting lysosomal function. When CDK4 inhibitors are used, the lack of lysosomal function induces senescence in triple-negative breast cancer (TNBC) cells and impairs tumor growth in a mouse xenograft model. Moreover, a combination of AMPK activation and CDK4 inhibition was used in an attempt to trigger autophagy in conditions when lysosomes are dysfunctional and resulted in cell death and tumor regression. This finding is of high relevance in TNBC, a highly invasive and aggressive cancer type that does not have a clear therapeutic strategy yet (27). Materials and Methods Materials. LY2835219, PD0332991 and LEE011 were purchased from MedChem Express and used at 0.5 µM, unless otherwise indicated. R3 human IGF-1 (I11146, Sigma) was used at 30 ng/ml. Minimal Essential Media (MEM) Amino Acids Solution (50X, Gibco) was used at the indicated concentrations. Rapamycin (LC Laboratories) was kindly provided by Pedro Romero (UNIL) and used at the indicated concentrations. Bafilomycin A1 (BafA1) was purchased from Enzo Life Sciences (ALX-380-030-M001) and used at 0.3 µM. Cell permeable αKG analog DMKG (349631, Sigma) was used at 5mM. Cell culture and transfection. MDA-MB-231, CCRF-CEM, HTC116, IB115, HT29, SKOV, MCF7 and PC3 cell lines, all obtained from ATCC, were cultured in RPMI 1640 + GlutaMAX containing, 10mM HEPES, 1mM sodium pyruvate (All from Gibco) and 10% fetal bovine serum (FBS) (PAA Laboratories). Thawed cells were allowed one passage to reach exponential growth phase before being used. Cells were used during maximum of ten passages in the experiments performed in this study. PCR-based mycoplasma tests were done routinely using specific primers: 5’-TGCACCATCTGTCACTCTGTTAACCTC-3’ and 3- GGGAGCAAACAGGATTAGATACCCT-5’, the last test was performed in June 2019. MDA-MB-231 CDK4 knockout (KO), E2F1 KO and FLCN KO stable cell lines were generated with CRISPR/Cas9 technology. The lentiCRISPR v2 plasmid was a gift from Feng Zhang (MIT; Addgene plasmid 52961)(28). The pMD2.G plasmid was a gift from Didier Trono (EPFL; Addgene 12259). The pCMV-dR8.91 plasmid and the guide RNA for FLCN were 4 Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 8, 2019; DOI: 10.1158/0008-5472.CAN-19-0708 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. gifts from Christian Widmann (UNIL). The target sequences for the guide RNAs are shown in Supplementary Table S1. Synthetic oligonucleotides were purchased and cloned into the digested LentiCrispR vector as described in Shalem et al (28).
Recommended publications
  • Role of Cyclin-Dependent Kinase 1 in Translational Regulation in the M-Phase
    cells Review Role of Cyclin-Dependent Kinase 1 in Translational Regulation in the M-Phase Jaroslav Kalous *, Denisa Jansová and Andrej Šušor Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Rumburska 89, 27721 Libechov, Czech Republic; [email protected] (D.J.); [email protected] (A.Š.) * Correspondence: [email protected] Received: 28 April 2020; Accepted: 24 June 2020; Published: 27 June 2020 Abstract: Cyclin dependent kinase 1 (CDK1) has been primarily identified as a key cell cycle regulator in both mitosis and meiosis. Recently, an extramitotic function of CDK1 emerged when evidence was found that CDK1 is involved in many cellular events that are essential for cell proliferation and survival. In this review we summarize the involvement of CDK1 in the initiation and elongation steps of protein synthesis in the cell. During its activation, CDK1 influences the initiation of protein synthesis, promotes the activity of specific translational initiation factors and affects the functioning of a subset of elongation factors. Our review provides insights into gene expression regulation during the transcriptionally silent M-phase and describes quantitative and qualitative translational changes based on the extramitotic role of the cell cycle master regulator CDK1 to optimize temporal synthesis of proteins to sustain the division-related processes: mitosis and cytokinesis. Keywords: CDK1; 4E-BP1; mTOR; mRNA; translation; M-phase 1. Introduction 1.1. Cyclin Dependent Kinase 1 (CDK1) Is a Subunit of the M Phase-Promoting Factor (MPF) CDK1, a serine/threonine kinase, is a catalytic subunit of the M phase-promoting factor (MPF) complex which is essential for cell cycle control during the G1-S and G2-M phase transitions of eukaryotic cells.
    [Show full text]
  • Analysis of 3-Phosphoinositide-Dependent Kinase-1 Signaling and Function in ES Cells
    EXPERIMENTAL CELL RESEARCH 314 (2008) 2299– 2312 available at www.sciencedirect.com www.elsevier.com/locate/yexcr Research Article Analysis of 3-phosphoinositide-dependent kinase-1 signaling and function in ES cells Tanja Tamgüneya,b, Chao Zhangc, Dorothea Fiedlerc, Kevan Shokatc, David Stokoea,⁎ aUCSF Cancer Research Institute, USA bMolecular Medicine Program, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany cDepartment of Cellular and Molecular Pharmacology, University of California, San Francisco, 2340 Sutter Street, San Francisco, CA 94115, USA ARTICLE INFORMATION ABSTRACT Article Chronology: 3-Phosphoinositide-dependent kinase-1 (PDK1) phosphorylates and activates several Received 5 February 2008 kinases in the cAMP-dependent, cGMP-dependent and protein kinase C (AGC) family. Revised version received Many putative PDK1 substrates have been identified, but have not been analyzed following 15 April 2008 transient and specific inhibition of PDK1 activity. Here, we demonstrate that a previously Accepted 16 April 2008 characterized PDK1 inhibitor, BX-795, shows biological effects that are not consistent with Available online 23 April 2008 PDK1 inhibition. Therefore, we describe the creation and characterization of a PDK1 mutant, L159G, which can bind inhibitor analogues containing bulky groups that hinder access to the − − Keywords: ATP binding pocket of wild type (WT) kinases. When expressed in PDK1 / ES cells, PDK1 PDK1 L159G restored phosphorylation of PDK1 targets known to be hypophosphorylated in these PKB/Akt cells. Screening of multiple inhibitor analogues showed that 1-NM-PP1 and 3,4-DMB-PP1 − − PI3K optimally inhibited the phosphorylation of PDK1 targets in PDK1 / ES cells expressing PDK1 Chemical genetics L159G but not WT PDK1. These compounds confirmed previously assumed PDK1 substrates, AGC kinases but revealed distinct dephosphorylation kinetics.
    [Show full text]
  • Dynamic Modelling of the Mtor Signalling Network Reveals Complex
    www.nature.com/scientificreports OPEN Dynamic modelling of the mTOR signalling network reveals complex emergent behaviours conferred by Received: 24 August 2017 Accepted: 1 December 2017 DEPTOR Published: xx xx xxxx Thawfeek M. Varusai1,4 & Lan K. Nguyen2,3,4 The mechanistic Target of Rapamycin (mTOR) signalling network is an evolutionarily conserved network that controls key cellular processes, including cell growth and metabolism. Consisting of the major kinase complexes mTOR Complex 1 and 2 (mTORC1/2), the mTOR network harbours complex interactions and feedback loops. The DEP domain-containing mTOR-interacting protein (DEPTOR) was recently identifed as an endogenous inhibitor of both mTORC1 and 2 through direct interactions, and is in turn degraded by mTORC1/2, adding an extra layer of complexity to the mTOR network. Yet, the dynamic properties of the DEPTOR-mTOR network and the roles of DEPTOR in coordinating mTORC1/2 activation dynamics have not been characterised. Using computational modelling, systems analysis and dynamic simulations we show that DEPTOR confers remarkably rich and complex dynamic behaviours to mTOR signalling, including abrupt, bistable switches, oscillations and co-existing bistable/oscillatory responses. Transitions between these distinct modes of behaviour are enabled by modulating DEPTOR expression alone. We characterise the governing conditions for the observed dynamics by elucidating the network in its vast multi-dimensional parameter space, and develop strategies to identify core network design motifs underlying these dynamics. Our fndings provide new systems-level insights into the complexity of mTOR signalling contributed by DEPTOR. Discovered in the early 1990s as an anti-fungal agent produced by the soil bacterium Streptomyces hygroscopicus, rapamycin has continually surprised scientists with its diverse clinical efects including potent immunosuppres- sive and anti-tumorigenic properties1–3.
    [Show full text]
  • Title Mtorc1 Upregulation Via ERK-Dependent Gene Expression Change Confers Intrinsic Resistance to MEK Inhibitors in Oncogenic Kras-Mutant Cancer Cells
    mTORC1 upregulation via ERK-dependent gene expression Title change confers intrinsic resistance to MEK inhibitors in oncogenic KRas-mutant cancer cells. Komatsu, Naoki; Fujita, Yoshihisa; Matsuda, Michiyuki; Aoki, Author(s) Kazuhiro Citation Oncogene (2015), 34(45): 5607-5616 Issue Date 2015-11-05 URL http://hdl.handle.net/2433/207613 This is the accepted manuscrip of the article is available at http://dx.doi.org/10.1038/onc.2015.16.; The full-text file will be made open to the public on 5 May 2016 in accordance with Right publisher's 'Terms and Conditions for Self-Archiving'.; この論 文は出版社版でありません。引用の際には出版社版をご 確認ご利用ください。; This is not the published version. Please cite only the published version. Type Journal Article Textversion author Kyoto University 1 Title mTORC1 upregulation via ERK-dependent gene expression change confers intrinsic resistance to MEK inhibitors in oncogenic KRas-mutant cancer cells. Authors Naoki Komatsu1, Yoshihisa Fujita2, Michiyuki Matsuda1,2, and Kazuhiro Aoki3 Affiliations 1. Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University, Japan 2. Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Japan 3. Imaging Platform for Spatio-Temporal Information, Graduate School of Medicine, Kyoto University, Japan To whom correspondence should be addressed Kazuhiro Aoki, Imaging Platform for Spatio-Temporal Information, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan; Tel.: 81-75-753-9450; Fax: 81-75-753-4698; E-mail: [email protected] Running title (less than 50 letters and spaces): Transcriptional control of mTORC1 activity by ERK 2 Abstract Cancer cells harboring oncogenic BRaf mutants, but not oncogenic KRas mutants, are sensitive to MEK inhibitors (MEKi).
    [Show full text]
  • A Pathway of Targeted Autophagy Is Induced by DNA Damage in Budding Yeast
    A pathway of targeted autophagy is induced by DNA damage in budding yeast Vinay V. Eapena,1, David P. Watermana, Amélie Bernardb, Nathan Schiffmannc, Enrich Sayasd,e, Roarke Kamberf, Brenda Lemosa, Gonen Memisoglua, Jessie Anga, Allison Mazellaa, Silvia G. Chuartzmanc, Robbie J. Loewithd,e, Maya Schuldinerc, Vladimir Denicf, Daniel J. Klionskyb, and James E. Habera,2 aDepartment of Biology, Brandeis University, Waltham, MA 02454; bLife Sciences Institute, University of Michigan, Ann Arbor, MI 48109; cDepartment of Molecular Genetics, Weizmann Institute of Sciences, Rehovot, Israel 7610001; dDepartment of Molecular Biology, University of Geneva, CH-1211 Geneva 4, Switzerland; eInstitute of Genetics and Genomics in Geneva, University of Geneva, CH-1211 Geneva 4, Switzerland; and fMolecular and Cell Biology, Harvard University, Cambridge, MA 02138 Contributed by James E. Haber, December 5, 2016 (sent for review August 31, 2016; reviewed by Eric H. Baehrecke and Stephen J. Elledge) Autophagy plays a central role in the DNA damage response (DDR) by conditions. For example, in response to nutrient starvation, cells controlling the levels of various DNA repair and checkpoint proteins; recycle internal reserves of basic metabolites by the degradation however, how the DDR communicates with the autophagy pathway of proteins, or even whole organelles, by their digestion in the remains unknown. Using budding yeast, we demonstrate that global vacuole/lysosome. This process is initiated by the formation of genotoxic damage or even a single unrepaired double-strand break double-membraned cytosolic sequestering vesicles, termed phag- (DSB) initiates a previously undescribed and selective pathway of ophores, which can engulf either random portions of the cyto- autophagy that we term genotoxin-induced targeted autophagy plasm, organelles, or specific proteins (10).
    [Show full text]
  • Targeting Mtor Signaling Overcomes Acquired Resistance to Combined BRAF and MEK Inhibition in BRAF-Mutant Melanoma
    www.nature.com/onc Oncogene ARTICLE OPEN Targeting mTOR signaling overcomes acquired resistance to combined BRAF and MEK inhibition in BRAF-mutant melanoma Beike Wang1,11, Wei Zhang1,11, Gao Zhang 2,10,11, Lawrence Kwong3, Hezhe Lu4, Jiufeng Tan2, Norah Sadek2, Min Xiao2, Jie Zhang 5, 6 2 2 2 2 5 7 6 Marilyne Labrie , Sergio Randell , Aurelie Beroard , Eric Sugarman✉ , Vito W. Rebecca✉ , Zhi Wei , Yiling Lu , Gordon B. Mills , Jeffrey Field8, Jessie Villanueva2, Xiaowei Xu9, Meenhard Herlyn 2 and Wei Guo 1 © The Author(s) 2021 Targeting MAPK pathway using a combination of BRAF and MEK inhibitors is an efficient strategy to treat melanoma harboring BRAF-mutation. The development of acquired resistance is inevitable due to the signaling pathway rewiring. Combining western blotting, immunohistochemistry, and reverse phase protein array (RPPA), we aim to understanding the role of the mTORC1 signaling pathway, a center node of intracellular signaling network, in mediating drug resistance of BRAF-mutant melanoma to the combination of BRAF inhibitor (BRAFi) and MEK inhibitor (MEKi) therapy. The mTORC1 signaling pathway is initially suppressed by BRAFi and MEKi combination in melanoma but rebounds overtime after tumors acquire resistance to the combination therapy (CR) as assayed in cultured cells and PDX models. In vitro experiments showed that a subset of CR melanoma cells was sensitive to mTORC1 inhibition. The mTOR inhibitors, rapamycin and NVP-BEZ235, induced cell cycle arrest and apoptosis in CR cell lines. As a proof-of-principle, we demonstrated that rapamycin and NVP-BEZ235 treatment reduced tumor growth in CR xenograft models. Mechanistically, AKT or ERK contributes to the activation of mTORC1 in CR cells, depending on PTEN status of these cells.
    [Show full text]
  • Novel Regulation of Mtor Complex 1 Signaling by Site-Specific Mtor Phosphorylation
    Novel Regulation of mTOR Complex 1 Signaling by Site-Specific mTOR Phosphorylation by Bilgen Ekim Üstünel A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy (Cell and Developmental Biology) in The University of Michigan 2012 Doctoral Committee: Assistant Professor Diane C. Fingar, Chair Associate Professor Billy Tsai Associate Professor Anne B. Vojtek Assistant Professor Patrick J. Hu Assistant Professor Ken Inoki “Our true mentor in life is science.” (“Hayatta en hakiki mürşit ilimdir.”) Mustafa Kemal Atatürk, the founder of Turkish Republic © Bilgen Ekim Üstünel 2012 Acknowledgements This thesis would not have been possible without the enormous support and encouragement of my Ph.D. advisor Diane C. Fingar. I am sincerely thankful for her research insight and guidance during my Ph.D. training. I would like to express my great appreciation to Billy Tsai, Anne B. Vojtek, Ken Inoki, and Patrick J. Hu for serving on my thesis committee, whose advice and help have been valuable. I would like to thank all members of the Fingar, Tsai, and Verhey labs for the discussion in our group meetings. I also would like to thank the CDB administrative staff, especillay Kristen Hug, for their help. I thank Ed Feener for performing the liquid chromatography tandem mass spectrometry analysis to identify novel phosphorylation sites on mTOR and Steve Riddle for performing the in vitro kinome screen to identify candidate kinases for mTOR S2159 phosphorylation site. I thank Brian Magnuson, Hugo A. Acosta-Jaquez, and Jennifer A. Keller for contributing to my first-author paper published in Molecular and Cellular Biology Journal in 2011.
    [Show full text]
  • Inhibition of Mtorc1 Leads to MAPK Pathway Activation Through a PI3K-Dependent Feedback Loop in Human Cancer
    Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer Arkaitz Carracedo, … , Jose Baselga, Pier Paolo Pandolfi J Clin Invest. 2008;118(9):3065-3074. https://doi.org/10.1172/JCI34739. Research Article Numerous studies have established a causal link between aberrant mammalian target of rapamycin (mTOR) activation and tumorigenesis, indicating that mTOR inhibition may have therapeutic potential. In this study, we show that rapamycin and its analogs activate the MAPK pathway in human cancer, in what represents a novel mTORC1-MAPK feedback loop. We found that tumor samples from patients with biopsy-accessible solid tumors of advanced disease treated with RAD001, a rapamycin derivative, showed an administration schedule–dependent increase in activation of the MAPK pathway. RAD001 treatment also led to MAPK activation in a mouse model of prostate cancer. We further show that rapamycin-induced MAPK activation occurs in both normal cells and cancer cells lines and that this feedback loop depends on an S6K-PI3K-Ras pathway. Significantly, pharmacological inhibition of the MAPK pathway enhanced the antitumoral effect of mTORC1 inhibition by rapamycin in cancer cells in vitro and in a xenograft mouse model. Taken together, our findings identify MAPK activation as a consequence of mTORC1 inhibition and underscore the potential of a combined therapeutic approach with mTORC1 and MAPK inhibitors, currently employed as single agents in the clinic, for the treatment of human cancers. Find the latest version: https://jci.me/34739/pdf Related Commentary, page 3003 Research article Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer Arkaitz Carracedo,1,2,3 Li Ma,2,3,4 Julie Teruya-Feldstein,3 Federico Rojo,5,6 Leonardo Salmena,1,2,3 Andrea Alimonti,1,2,3 Ainara Egia,1,2,3 Atsuo T.
    [Show full text]
  • Inhibition of Mtorc1/2 Overcomes Resistance to MAPK Pathway Inhibitors Mediated by Pgc1a and Oxidative Phosphorylation in Melanoma
    Published OnlineFirst October 8, 2014; DOI: 10.1158/0008-5472.CAN-14-1392 Cancer Therapeutics, Targets, and Chemical Biology Research Inhibition of mTORC1/2 Overcomes Resistance to MAPK Pathway Inhibitors Mediated by PGC1a and Oxidative Phosphorylation in Melanoma Y.N. Vashisht Gopal1, Helen Rizos2, Guo Chen1, Wanleng Deng1, Dennie T. Frederick3, Zachary A. Cooper4, Richard A. Scolyer2, Gulietta Pupo2, Kakajan Komurov5, Vasudha Sehgal6, Jiexin Zhang7, Lalit Patel8, Cristiano G. Pereira1, Bradley M. Broom7, Gordon B. Mills6, Prahlad Ram6, Paul D. Smith9, Jennifer A. Wargo4, Georgina V. Long2, and Michael A. Davies1,6 Abstract Metabolic heterogeneity is a key factor in cancer pathogenesis. We found that a subset of BRAF-andNRAS- mutant human melanomas resistant to the MEK inhibitor selumetinib displayed increased oxidative phosphorylation (OxPhos) mediated by the transcriptional coactivator PGC1a. Notably, all selumetinib- resistant cells with elevated OxPhos could be resensitized by cotreatment with the mTORC1/2 inhibitor AZD8055,whereasthiscombinationwasineffectiveinresistantcelllineswithlowOxPhos.InbothBRAF-and NRAS-mutant melanoma cells, MEK inhibition increased MITFexpression,whichinturnelevatedlevelsof PGC1a. In contrast, mTORC1/2 inhibition triggered cytoplasmic localization of MITF, decreasing PGC1a expression and inhibiting OxPhos. Analysis of tumor biopsies from patients with BRAF-mutant melanoma progressing on BRAF inhibitor Æ MEK inhibitor revealed that PGC1a levels were elevated in approximately half of the resistant tumors. Overall, our findings highlight the significance of OxPhos in melanoma and suggest that combined targeting of the MAPK and mTORC pathways may offer an effective therapeutic strategy to treat melanomas with this metabolic phenotype. Cancer Res; 74(23); 1–11. Ó2014 AACR. Introduction mechanisms of de novo and acquired resistance to MAPK The identification of frequent activating mutations in pathway inhibitors.
    [Show full text]
  • Regulation of the Error-Prone DNA Polymerase Polκ by Oncogenic Signaling and Its Contribution to Drug Resistance
    bioRxiv preprint doi: https://doi.org/10.1101/316729; this version posted May 11, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Title: Regulation of the error-prone DNA polymerase polκ by oncogenic signaling and its contribution to drug resistance 1,2 3 4 5 Authors: Kelsey Temprine , Erin M Langdon , Krisha Mehta , Averill Clapp , Richard M White1* Affiliations: 1Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. 2Gerstner Sloan Kettering Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. 3University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA. 4Division of General Internal Medicine, Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA. 5University of Chicago, Chicago, IL 60637, USA. * Corresponding author. Email: [email protected] One Sentence Summary: Cancer cells under stress from oncogene or mTOR inhibition dysregulate the error-prone DNA polymerase polk, which contributes to drug resistance in melanoma cells. 1 bioRxiv preprint doi: https://doi.org/10.1101/316729; this version posted May 11, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Abstract Mutations in the proofreading domains of the replicative DNA polymerases polδ and polε are associated with elevated mutation rates in cancer, but the roles of other DNA polymerases in tumorigenesis remain poorly understood.
    [Show full text]
  • Mtorc1 Is a Mechanosensor That Regulates Surfactant Function and Lung Compliance During Ventilator-Induced Lung Injury
    RESEARCH ARTICLE mTORC1 is a mechanosensor that regulates surfactant function and lung compliance during ventilator-induced lung injury Hyunwook Lee,1,2 Qinqin Fei,1,2 Adam Streicher,1,2 Wenjuan Zhang,1,2 Colleen Isabelle,3 Pragi Patel,1,2 Hilaire C. Lam,3 Antonio Arciniegas-Rubio,3 Miguel Pinilla-Vera,3 Diana P. Amador-Munoz,3 Diana Barragan-Bradford,3 Angelica Higuera-Moreno,3 Rachel K. Putman,3 Lynette M. Sholl,4 Elizabeth P. Henske,3 Christopher M. Bobba,1,2,5 Natalia Higuita-Castro,2,5 Emily M. Shalosky,1,2 R. Duncan Hite,6 John W. Christman,1,2 Samir N. Ghadiali,2,5 Rebecca M. Baron,3 and Joshua A. Englert1,2 1Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, and 2The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State Wexner Medical Center, Columbus, Ohio, USA. 3Division of Pulmonary and Critical Care Medicine, Department of Medicine, and 4Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA. 5Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA. 6Division of Pulmonary, Critical Care, and Sleep Medicine, The University of Cincinnati College of Medicine, Cincinnati, Ohio, USA. The acute respiratory distress syndrome (ARDS) is a highly lethal condition that impairs lung function and causes respiratory failure. Mechanical ventilation (MV) maintains gas exchange in patients with ARDS but exposes lung cells to physical forces that exacerbate injury. Our data demonstrate that mTOR complex 1 (mTORC1) is a mechanosensor in lung epithelial cells and that activation of this pathway during MV impairs lung function.
    [Show full text]
  • MTOR Signaling and Metabolism in Early T Cell Development
    G C A T T A C G G C A T genes Review MTOR Signaling and Metabolism in Early T Cell Development Guy Werlen, Ritika Jain and Estela Jacinto * Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA; [email protected] (G.W.); [email protected] (R.J.) * Correspondence: [email protected]; Tel.: +1-732-235-4476 Abstract: The mechanistic target of rapamycin (mTOR) controls cell fate and responses via its func- tions in regulating metabolism. Its role in controlling immunity was unraveled by early studies on the immunosuppressive properties of rapamycin. Recent studies have provided insights on how metabolic reprogramming and mTOR signaling impact peripheral T cell activation and fate. The contribution of mTOR and metabolism during early T-cell development in the thymus is also emerging and is the subject of this review. Two major T lineages with distinct immune functions and peripheral homing organs diverge during early thymic development; the αβ- and γδ-T cells, which are defined by their respective TCR subunits. Thymic T-regulatory cells, which have immunosup- pressive functions, also develop in the thymus from positively selected αβ-T cells. Here, we review recent findings on how the two mTOR protein complexes, mTORC1 and mTORC2, and the signaling molecules involved in the mTOR pathway are involved in thymocyte differentiation. We discuss emerging views on how metabolic remodeling impacts early T cell development and how this can be mediated via mTOR signaling. Keywords: mTOR; mTORC1; mTORC2; thymocytes; T lymphocytes; early T cell development; T-cell metabolism Citation: Werlen, G.; Jain, R.; Jacinto, E.
    [Show full text]