Understand KRAS and the Quest for Anti-Cancer Drugs

Total Page:16

File Type:pdf, Size:1020Kb

Understand KRAS and the Quest for Anti-Cancer Drugs cells Review Understand KRAS and the Quest for Anti-Cancer Drugs Chang Woo Han 1 , Mi Suk Jeong 2,* and Se Bok Jang 3,* 1 Institute of Systems Biology, Pusan National University, Jangjeon-dong, Geumjeong-gu, Busan 46241, Korea; [email protected] 2 Institute for Plastic Information and Energy Materials and Sustainable Utilization of Photovoltaic Energy Research Center, Pusan National University, Jangjeon-dong, Geumjeong-gu, Busan 46241, Korea 3 Department of Molecular Biology, College of Natural Sciences, Pusan National University, Jangjeon-dong, Geumjeong-gu, Busan 46241, Korea * Correspondence: [email protected] (M.S.J.); [email protected] (S.B.J.); Tel.: +82-51-510-2523 (M.S.J. & S.B.J.) Abstract: The KRAS oncogene is mutated in approximately ~30% of human cancers, and the targeting of KRAS has long been highlighted in many studies. Nevertheless, attempts to target KRAS directly have been ineffective. This review provides an overview of the structure of KRAS and its characteristic signaling pathways. Additionally, we examine the problems associated with currently available KRAS inhibitors and discuss promising avenues for drug development. Keywords: KRAS; cancer; signaling pathway; inhibitors 1. Introduction RAS HRAS KRAS NRAS genes ( , , and ) encode 21 kDa monomeric small guanosine triphosphate (GTP)-hydrolases (GTPases) that function as regulators of intracellular signal- Citation: Han, C.W.; Jeong, M.S.; ing transduction cascades involved in cell growth, differentiation, and survival. Normally, Jang, S.B. Understand KRAS and the the cycling of RAS protein by GTPases is tightly regulated by the binding of guanosine Quest for Anti-Cancer Drugs. Cells triphosphate (GTP) to nucleotide exchange factors (GEFs) and by the hydrolysis of GTP 2021, 10, 842. https://doi.org/ to guanosine diphosphate (GDP) by GTPase activating proteins (GAPs) [1,2]. KRAS is 10.3390/cells10040842 frequently mutated in pancreatic, colorectal, endometrial, biliary tract, lung, and cervical cancer. For example, oncogenic missense mutations in the KRAS gene frequently occur Academic Editor: Bor Luen Tang at glycine (Gly) 12 or 13 or glutamine (Gln) 61 codons located on the P loop and Switch II region and are related to the impairment of stable nucleotide-binding in its active site. Received: 11 March 2021 These mutations impair GTPase cycling and result in accumulations of bound and active Accepted: 6 April 2021 GTP, which result in activation of multiple downstream effectors [3,4]. In this review, Published: 8 April 2021 we examine aberrant activated KRAS signaling in tumors and place focus on promising KRAS inhibitors. Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in 2. Structure published maps and institutional affil- iations. KRAS is a 21 kDa monomeric, membrane-localized GTPase with a conserved domain, known as the G domain (residues 1–165, Figure1A), and a less conserved C-terminal tail, called the hypervariable domain (residues 166–188, Figure1A) [ 5]. KRAS consists of five α2-helixes (α1–α5) and six β-sheets (β1–β6). GTPase has two switches— switches I (residues 30–38) and II (residues 60–76)—which undergo major conformational changes in Copyright: © 2021 by the authors. response to changes in guanine nucleotide (GTP to GDP or vice versa) [6]. The phosphate- Licensee MDPI, Basel, Switzerland. binding loop (P-loop, residues 10–17) of KRAS participates in the induction of the confor- This article is an open access article distributed under the terms and mational change that occurs during its GDP-bound (inactive) and the GTP-bound (active) conditions of the Creative Commons states (Figure1B). The γ-phosphate of GTP interacts with tyrosine (Tyr) 32 and threonine Attribution (CC BY) license (https:// (Thr) 35 residues of the switch I region. Additionally, conserved Gly 60 in the switch creativecommons.org/licenses/by/ II region interacts with γ-phosphate [7]. KRAS is the most frequently mutated proto- 4.0/). oncogene in human cancers, and mutated KRAS remains locked in an active state, relaying Cells 2021, 10, 842. https://doi.org/10.3390/cells10040842 https://www.mdpi.com/journal/cells Cells 2021, 10, x 2 of 11 Cells 2021, 10, 842 2 of 10 (active) states (Figure 1B). The γ-phosphate of GTP interacts with tyrosine (Tyr) 32 and threonine (Thr) 35 residues of the switch I region. Additionally, conserved Gly 60 in the switch Ⅱ region interacts with γ-phosphate [7]. KRAS is the most frequently mutated uncontrolledproto-oncogene proliferative in human cancers, signals. and Jonathanmutated KRAS M. Ostrem remains et locked al. identified in an active that state, oncogenic mutationsrelaying uncontrolled result in conformational proliferative signals. change Jonathan and functionalM. Ostrem et activation al. identified of RAS that proteinson- by impairingcogenic mutations both intrinsic result in GTPase conformational activity change and GAP-stimulated and functional activation GTP hydrolysis of RAS [pro-6]. Recently, manyteins by studies impairing have both focused intrinsic on GTPase attempts activity that and target GAP exchange-stimulated nucleotide. GTP hydrolysis Recent [6]. studies haveRecently, focused many onstudies attempts have focus to blocked on nucleotideattempts that exchange target exchange [8,9]. nucleotide. Several structure-based Recent inhibitorsstudies have (e.g., focused SML-8-73-1, on attempts H-REV107 to block peptide, nucleotide BAY-293, exchange or BI-3406)[8,9]. Several have structure been reported- to targetbased inhibitors the guanine (e.g., nucleotide SML-8-73-1, binding H-REV107 site peptide, of the BAY KRAS-293 mutant, or BI-3406) and have inhibit been GDP re- to GTP exchangeported to target[10– 13the]. guanine The Cys-A-A-X nucleotide C-terminalbinding site of motif, the KRAS where mutant A is and isoleucine, inhibit GDP leucine, or valine,to GTP exchange and X is [10 either–13]. methionine The Cys-A-A or-X serine,C-terminal is located motif, where in the A KRAS is isoleucine, hypervariable leucine, region (HVR).or valine, KRAS and X traffickingis either methionine on the plasma or serine, membrane is located in plays the KRAS essential hypervariable roles in multiple region signal- (HVR). KRAS trafficking on the plasma membrane plays essential roles in multiple sig- ing pathways that control various processes, and the Cys-A-A-X C-terminal motif of KRAS naling pathways that control various processes, and the Cys-A-A-X C-terminal motif of undergoesKRAS undergoes several several post-translational post-translational modifications modifications (PTMs) (PTMs) that that facilitate facilitate itsits traffickingtraf- to theficking inner to the surface inner of surface the plasma of the plasma membrane membrane [14–16 [14].–16]. Cells 2021, 10, x 3 of 11 FigureFigure 1.1. DomainDomain structuresstructures and the three three-dimensional-dimensional structure structure.. (A ()A Domain) Domain structure structure of offull full-length- length KRAS. (B) Ribbon representation of the monomeric crystal structure of KRAS (PDB ID: 7C40) KRAS. (B) Ribbon representation of the monomeric crystal structure of KRAS (PDB ID: 7C40) using using the program PyMOL. The Mg2+ ion is indicated by a gray circle and guanosine diphosphate 2+ the(GDP program) by a yellow PyMOL. rod. TheThe MgGDP Mg ion molecule is indicated is color by-coded a gray as circlefollows and: C yellow, guanosine O red, diphosphate N blue, P (GDP) bypurple, a yellow and rod.Mg2+ Thegray. MgGDP molecule is color-coded as follows: C yellow, O red, N blue, P purple, and Mg2+ gray. (i) KRAS is synthesized as a biologically inactive cytosolic propeptide (Pro-Ras). (ii) The first modification of this propeptide is catalyzed by farnesyltransferase (FTase) and involves covalent addition of the 15-carbon farnesyl group (C-15) from farnesyl diphos- phate (FDP) onto the cysteine residue of the CAAX sequence. (iii) Next, the -AAX se- quence is cleaved by a CAAX protease RAS converting CAAX endopeptidase 1 also known as RAS converting enzyme 1 (RCE1), and this is followed by methylation of the carboxyl-prenylated cysteine residue through isoprenylcysteine carboxyl methyltransfer- ase (ICMT). (iv) Finally, palmitoyl transferase (PTase) catalyzes the addition of palmitic acid (a 16-carbon fatty acid) to upstream cysteine residues. These KRAS modifications enhance protein hydrophobicity and plasma membrane anchoring, and membrane-anchored KRAS proteins cycle between the GTP-bound active form and the GDP-bound inactive form. The associated conformational changes of KRAS serve as master moderators of a large number of signaling pathways involved in various cellular processes. 3. Oncogenic KRAS Signaling Pathways in Human Cancer 3.1. GEFs and GAPs KRAS-associated signaling pathways are persistently activated in many cancers, where they participate in cellular growth and proliferation, differentiation, protein syn- thesis, glucose metabolism, cell survival, and inflammation (Figure 2) [3]. KRAS is acti- vated by GEFs: SOS1 and SOS2 (two sons of sevenless 1 and 2), GRB2 (growth factor re- ceptor-bound protein 2), RASGRF2 (Ras protein-specific guanine nucleotide releasing fac- tor 2), or RasGRPs (Ras guanine nucleotide releasing proteins). GEFs are activated by Cells 2021, 10, 842 3 of 10 (i) KRAS is synthesized as a biologically inactive cytosolic propeptide (Pro-Ras). (ii) The first modification of this propeptide is catalyzed by farnesyltransferase (FTase) and involves covalent addition of the 15-carbon farnesyl group (C-15) from farnesyl diphos- phate (FDP) onto
Recommended publications
  • Adaptive Stress Signaling in Targeted Cancer Therapy Resistance
    Oncogene (2015) 34, 5599–5606 © 2015 Macmillan Publishers Limited All rights reserved 0950-9232/15 www.nature.com/onc REVIEW Adaptive stress signaling in targeted cancer therapy resistance E Pazarentzos1,2 and TG Bivona1,2 The identification of specific genetic alterations that drive the initiation and progression of cancer and the development of targeted drugs that act against these driver alterations has revolutionized the treatment of many human cancers. Although substantial progress has been achieved with the use of such targeted cancer therapies, resistance remains a major challenge that limits the overall clinical impact. Hence, despite progress, new strategies are needed to enhance response and eliminate resistance to targeted cancer therapies in order to achieve durable or curative responses in patients. To date, efforts to characterize mechanisms of resistance have primarily focused on molecular events that mediate primary or secondary resistance in patients. Less is known about the initial molecular response and adaptation that may occur in tumor cells early upon exposure to a targeted agent. Although understudied, emerging evidence indicates that the early adaptive changes by which tumor cells respond to the stress of a targeted therapy may be crucial for tumo r cell survival during treatment and the development of resistance. Here we review recent data illuminating the molecular architecture underlying adaptive stress signaling in tumor cells. We highlight how leveraging this knowledge could catalyze novel strategies to minimize
    [Show full text]
  • The Rac Gtpase in Cancer: from Old Concepts to New Paradigms Marcelo G
    Published OnlineFirst August 14, 2017; DOI: 10.1158/0008-5472.CAN-17-1456 Cancer Review Research The Rac GTPase in Cancer: From Old Concepts to New Paradigms Marcelo G. Kazanietz1 and Maria J. Caloca2 Abstract Rho family GTPases are critical regulators of cellular func- mislocalization of Rac signaling components. The unexpected tions that play important roles in cancer progression. Aberrant pro-oncogenic functions of Rac GTPase-activating proteins also activity of Rho small G-proteins, particularly Rac1 and their challenged the dogma that these negative Rac regulators solely regulators, is a hallmark of cancer and contributes to the act as tumor suppressors. The potential contribution of Rac tumorigenic and metastatic phenotypes of cancer cells. This hyperactivation to resistance to anticancer agents, including review examines the multiple mechanisms leading to Rac1 targeted therapies, as well as to the suppression of antitumor hyperactivation, particularly focusing on emerging paradigms immune response, highlights the critical need to develop ther- that involve gain-of-function mutations in Rac and guanine apeutic strategies to target the Rac pathway in a clinical setting. nucleotide exchange factors, defects in Rac1 degradation, and Cancer Res; 77(20); 5445–51. Ó2017 AACR. Introduction directed toward targeting Rho-regulated pathways for battling cancer. Exactly 25 years ago, two seminal papers by Alan Hall and Nearly all Rho GTPases act as molecular switches that cycle colleagues illuminated us with one of the most influential dis- between GDP-bound (inactive) and GTP-bound (active) forms. coveries in cancer signaling: the association of Ras-related small Activation is promoted by guanine nucleotide exchange factors GTPases of the Rho family with actin cytoskeleton reorganization (GEF) responsible for GDP dissociation, a process that normally (1, 2).
    [Show full text]
  • Regulation of the Mammalian Target of Rapamycin Complex 2 (Mtorc2)
    Regulation of the Mammalian Target Of Rapamycin Complex 2 (mTORC2) Inauguraldissertation Zur Erlangung der Würde eines Doktors der Philosophie vorgelegt der Philosophisch-Naturwissenschaftlichen Fakultät der Universität Basel von Klaus-Dieter Molle aus Heilbronn, Deutschland Basel, 2006 Genehmigt von der Philosophisch-Naturwissenschaftlichen Fakultät Auf Antrag von Prof. Michael N. Hall und Prof. Markus Affolter. Basel, den 21.11.2006 Prof. Hans-Peter Hauri Dekan Summary The growth controlling mammalian Target of Rapamycin (mTOR) is a conserved Ser/Thr kinase found in two structurally and functionally distinct complexes, mTORC1 and mTORC2. The tumor suppressor TSC1-TSC2 complex inhibits mTORC1 by acting on the small GTPase Rheb, but the role of TSC1-TSC2 and Rheb in the regulation of mTORC2 is unclear. Here we examined the role of TSC1-TSC2 in the regulation of mTORC2 in human embryonic kidney 293 cells. Induced knockdown of TSC1 and TSC2 (TSC1/2) stimulated mTORC2-dependent actin cytoskeleton organization and Paxillin phosphorylation. Furthermore, TSC1/2 siRNA increased mTORC2-dependent Ser473 phosphorylation of plasma membrane bound, myristoylated Akt/PKB. This suggests that loss of Akt/PKB Ser473 phosphorylation in TSC mutant cells, as reported previously, is due to inhibition of Akt/PKB localization rather than inhibition of mTORC2 activity. Amino acids and overexpression of Rheb failed to stimulate mTORC2 signaling. Thus, TSC1-TSC2 also inhibits mTORC2, but possibly independently of Rheb. Our results suggest that mTORC2 hyperactivation may contribute to the pathophysiology of diseases such as cancer and Tuberous Sclerosis Complex. i Acknowledgement During my PhD studies in the Biozentrum I received a lot of support from many people around me who I mention here to express my gratefulness.
    [Show full text]
  • Lymphocyte/WBC-Based Comprehensive Testing Via Next-Gen Sequencing NF1/SPRED1 and Other Rasopathy Related Conditions on Blood/Saliva
    Last Updated October 2019 Institutional Price TAT Genetic Test CPT Codes Z codes Specimen Requirements (USD$) (working days) Lymphocyte/WBC-based Comprehensive Testing via Next-Gen Sequencing NF1/SPRED1 and Other RASopathy Related Conditions on Blood/Saliva NF1- only NGS testing and copy number analysis for the NF1 gene (NF1-NG) This testing includes an average coverage of >1600x to allow for the identification of (1) 3-6ml whole blood in EDTA (purple topped) tubes mosaicism as low as 3-5% of the alleles. In addition, novel variants identified in the $1,000 81408 25 (2) Oragene 575 saliva kit (provided by the MGL) ZB6A9 NF1 gene will be confirmed via RNA-based analysis at no additional charge. RNA- $1,600 (RUSH) 81479 15 (RUSH) (3) DNA sample (25ul volume at 3ug, O.D. value at based testing will also be provided to non-founder, multigenerational families with 260:280 ≥1.8) “classic” NF1 at no additional charge if next-generation sequencing is found negative. (1) 3-6ml whole blood in EDTA (purple topped) tubes SPRED1-only NGS testing and copy number analysis for SPRED1 (SPD1-NG) $800 81405 25 (2) Oragene 575 saliva kit (provided by the MGL) This testing includes an average coverage of >1600x to allow for the identification of ZB6AC $1,400 (RUSH) 81479 15 (RUSH) (3) DNA sample (25ul volume at 3ug, O.D. value at mosaicism as low as 3-5% of the alleles after comprehensive NF1 analysis. 260:280 ≥1.8) NF1/SPRED1 NGS testing and copy number analysis for NF1 and SPRED1 (NFSP-NG) (1) 3-6ml whole blood in EDTA (purple topped) tubes 81408 This testing includes an average coverage of >1600x to allow for the identification of $1,100 25 (2) Oragene 575 saliva kit (provided by the MGL) 81405 ZB6A8 mosaicism as low as 3-5% of the alleles.
    [Show full text]
  • A GTP-State Specific Cyclic Peptide Inhibitor of the Gtpase Gαs
    bioRxiv preprint doi: https://doi.org/10.1101/2020.04.25.054080; this version posted April 27, 2020. 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. A GTP-state specific cyclic peptide inhibitor of the GTPase Gαs Shizhong A. Dai1,2†, Qi Hu1,2†, Rong Gao3†, Andre Lazar1,4†, Ziyang Zhang1,2, Mark von Zastrow1,4, Hiroaki Suga3*, Kevan M. Shokat1,2* 5 1Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, 94158, USA 2Howard Hughes Medical Institute 3Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan 10 4Department of Psychiatry, University of California, San Francisco, San Francisco, CA, 94158, USA *Correspondence to: [email protected], [email protected] †These authors contributed equally. 15 20 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.04.25.054080; this version posted April 27, 2020. 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: The G protein-coupled receptor (GPCR) cascade leading to production of the second messenger cAMP is replete with pharmacologically targetable receptors and enzymes with the exception of the stimulatory G protein α subunit, Gαs.
    [Show full text]
  • Clinical Utility of Recently Identified Diagnostic, Prognostic, And
    Modern Pathology (2017) 30, 1338–1366 1338 © 2017 USCAP, Inc All rights reserved 0893-3952/17 $32.00 Clinical utility of recently identified diagnostic, prognostic, and predictive molecular biomarkers in mature B-cell neoplasms Arantza Onaindia1, L Jeffrey Medeiros2 and Keyur P Patel2 1Instituto de Investigacion Marques de Valdecilla (IDIVAL)/Hospital Universitario Marques de Valdecilla, Santander, Spain and 2Department of Hematopathology, MD Anderson Cancer Center, Houston, TX, USA Genomic profiling studies have provided new insights into the pathogenesis of mature B-cell neoplasms and have identified markers with prognostic impact. Recurrent mutations in tumor-suppressor genes (TP53, BIRC3, ATM), and common signaling pathways, such as the B-cell receptor (CD79A, CD79B, CARD11, TCF3, ID3), Toll- like receptor (MYD88), NOTCH (NOTCH1/2), nuclear factor-κB, and mitogen activated kinase signaling, have been identified in B-cell neoplasms. Chronic lymphocytic leukemia/small lymphocytic lymphoma, diffuse large B-cell lymphoma, follicular lymphoma, mantle cell lymphoma, Burkitt lymphoma, Waldenström macroglobulinemia, hairy cell leukemia, and marginal zone lymphomas of splenic, nodal, and extranodal types represent examples of B-cell neoplasms in which novel molecular biomarkers have been discovered in recent years. In addition, ongoing retrospective correlative and prospective outcome studies have resulted in an enhanced understanding of the clinical utility of novel biomarkers. This progress is reflected in the 2016 update of the World Health Organization classification of lymphoid neoplasms, which lists as many as 41 mature B-cell neoplasms (including provisional categories). Consequently, molecular genetic studies are increasingly being applied for the clinical workup of many of these neoplasms. In this review, we focus on the diagnostic, prognostic, and/or therapeutic utility of molecular biomarkers in mature B-cell neoplasms.
    [Show full text]
  • Tumor Markers
    Tumor Markers Alan H.B. Wu, Ph.D. Professor, Laboratory Medicine, UCSF Section Chief, Clinical Chemistry, Toxicology, Pharmacogenomics Laboratory, SFGH Learning objectives • Know the ideal characteristics of a tumor marker • Understand the role of tumor markers for diagnosis and management of patients with cancer. • Know the emerging technologies for tumor markers • Understand the role of tumor markers for therapeutic selection How do we diagnose cancer today? Physical Examination Blood tests CT scans Biopsy Human Prostate Cancer Normal Blood Smear Chronic Myeloid Leukemia Death rates for cancer vs. heart disease New cancer cases per year Cancer Site or Type New Cases Prostate 218,000 Lung 222,500 Breast 207,500 Colorectal 149,000 Urinary system 131,500 Skin 68,770 Pancreas 43,100 Ovarian 22,000 Myeloma 20,200 Thyroid 44,700 Germ Cell 9,000 Types of Tumor Markers • Hormones (hCG; calcitonin; gastrin; prolactin;) • Enzymes (acid phosphatase; alkaline phosphatase; PSA) • Cancer antigen proteins & glycoproteins (CA125; CA 15.3; CA19.9) • Metabolites (norepinephrine, epinephrine) • Normal proteins (thyroglobulin) • Oncofetal antigens (CEA, AFP) • Receptors (ER, PR, EGFR) • Genetic changes (mutations/translocations, etc.) Characteristics of an ideal tumor marker • Specificity for a single type of cancer • High sensitivity and specificity for cancerous growth • Correlation of marker level with tumor size • Homogeneous (i.e., minimal post-translational modifications) • Short half-life in circulation Roles for tumor markers • Determine risk (PSA)
    [Show full text]
  • Mutation Analysis of Son of Sevenless in Juvenile Myelomonocytic Leukemia
    Letters to the Editor 1108 3 Licht JD. Reconstructing a disease: what essential features of the the development of acute promyelocytic leukemia in transgenic retinoic acid receptor fusion oncoproteins generate acute promye- mice. Proc Natl Acad Sci USA 2000; 97: 13306–13311. locytic leukemia? Cancer Cell 2006; 9: 73–74. 10 Sternsdorf T, Phan VT, Maunakea ML, Ocampo CB, Sohal J, 4 Cools J, DeAngelo DJ, Gotlib J, Stover EH, Legare RD, Cortes J et al. Silletto A et al. Forced retinoic acid receptor alpha homodimers A tyrosine kinase created by fusion of the PDGFRA and FIP1L1 prime mice for APL-like leukemia. Cancer Cell 2006; 9: 81–94. genes as a therapeutic target of imatinib in idiopathic hypereosino- 11 Kwok C, Zeisig BB, Dong S, So CW. Forced homo-oligomerization philic syndrome. N Engl J Med 2003; 348: 1201–1214. of RARalpha leads to transformation of primary hematopoietic 5 Kaufmann I, Martin G, Friedlein A, Langen H, Keller W. Human cells. Cancer Cell 2006; 9: 95–108. Fip1 is a subunit of CPSF that binds to U-rich RNA elements and 12 Palaniswamy V, Moraes KC, Wilusz CJ, Wilusz J. Nucleophosmin stimulates poly(A) polymerase. EMBO J 2004; 23: 616–626. is selectively deposited on mRNA during polyadenylation. Nat 6 Sainty D, Liso V, Cantu-Rajnoldi A, Head D, Mozziconacci MJ, Struct Mol Biol 2006; 13: 429–435. Arnoulet C et al. A new morphologic classification system for acute 13 Rego EM, Ruggero D, Tribioli C, Cattoretti G, Kogan S, Redner RL promyelocytic leukemia distinguishes cases with underlying PLZF/ et al.
    [Show full text]
  • Screening for Tumor Suppressors: Loss of Ephrin PNAS PLUS Receptor A2 Cooperates with Oncogenic Kras in Promoting Lung Adenocarcinoma
    Screening for tumor suppressors: Loss of ephrin PNAS PLUS receptor A2 cooperates with oncogenic KRas in promoting lung adenocarcinoma Narayana Yeddulaa, Yifeng Xiaa, Eugene Kea, Joep Beumera,b, and Inder M. Vermaa,1 aLaboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037; and bHubrecht Institute, Utrecht, The Netherlands Contributed by Inder M. Verma, October 12, 2015 (sent for review July 28, 2015; reviewed by Anton Berns, Tyler Jacks, and Frank McCormick) Lung adenocarcinoma, a major form of non-small cell lung cancer, injections in embryonic skin cells identified several potential tu- is the leading cause of cancer deaths. The Cancer Genome Atlas morigenic factors (14–16). None of the reported studies have analysis of lung adenocarcinoma has identified a large number of performed direct shRNA-mediated high-throughput approaches previously unknown copy number alterations and mutations, re- in adult mice recapitulating the mode of tumorigenesis in humans. quiring experimental validation before use in therapeutics. Here, we Activating mutations at positions 12, 13, and 61 amino acids in describe an shRNA-mediated high-throughput approach to test a set Kirsten rat sarcoma viral oncogene homolog (KRas) contributes of genes for their ability to function as tumor suppressors in the to tumorigenesis in 32% of lung adenocarcinoma patients (2) by background of mutant KRas and WT Tp53. We identified several activating downstream signaling cascades. Mice with the KRasG12D candidate genes from tumors originated from lentiviral delivery of allele develop benign adenomatous lesions with long latency to shRNAs along with Cre recombinase into lungs of Loxp-stop-Loxp- develop adenocarcinoma (17, 18).
    [Show full text]
  • Supplementary Material DNA Methylation in Inflammatory Pathways Modifies the Association Between BMI and Adult-Onset Non- Atopic
    Supplementary Material DNA Methylation in Inflammatory Pathways Modifies the Association between BMI and Adult-Onset Non- Atopic Asthma Ayoung Jeong 1,2, Medea Imboden 1,2, Akram Ghantous 3, Alexei Novoloaca 3, Anne-Elie Carsin 4,5,6, Manolis Kogevinas 4,5,6, Christian Schindler 1,2, Gianfranco Lovison 7, Zdenko Herceg 3, Cyrille Cuenin 3, Roel Vermeulen 8, Deborah Jarvis 9, André F. S. Amaral 9, Florian Kronenberg 10, Paolo Vineis 11,12 and Nicole Probst-Hensch 1,2,* 1 Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland; [email protected] (A.J.); [email protected] (M.I.); [email protected] (C.S.) 2 Department of Public Health, University of Basel, 4001 Basel, Switzerland 3 International Agency for Research on Cancer, 69372 Lyon, France; [email protected] (A.G.); [email protected] (A.N.); [email protected] (Z.H.); [email protected] (C.C.) 4 ISGlobal, Barcelona Institute for Global Health, 08003 Barcelona, Spain; [email protected] (A.-E.C.); [email protected] (M.K.) 5 Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain 6 CIBER Epidemiología y Salud Pública (CIBERESP), 08005 Barcelona, Spain 7 Department of Economics, Business and Statistics, University of Palermo, 90128 Palermo, Italy; [email protected] 8 Environmental Epidemiology Division, Utrecht University, Institute for Risk Assessment Sciences, 3584CM Utrecht, Netherlands; [email protected] 9 Population Health and Occupational Disease, National Heart and Lung Institute, Imperial College, SW3 6LR London, UK; [email protected] (D.J.); [email protected] (A.F.S.A.) 10 Division of Genetic Epidemiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; [email protected] 11 MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, W2 1PG London, UK; [email protected] 12 Italian Institute for Genomic Medicine (IIGM), 10126 Turin, Italy * Correspondence: [email protected]; Tel.: +41-61-284-8378 Int.
    [Show full text]
  • Tuberous Sclerosis Complex: Molecular Pathogenesis and Animal Models
    Neurosurg Focus 20 (1):E4, 2006 Tuberous sclerosis complex: molecular pathogenesis and animal models LEANDRO R. PIEDIMONTE, M.D., IAN K. WAILES, M.D., AND HOWARD L. WEINER, M.D. Division of Pediatric Neurosurgery, Department of Neurosurgery, New York University School of Medicine, New York, New York Mutations in one of two genes, TSC1 and TSC2, result in a similar disease phenotype by disrupting the normal inter- action of their protein products, hamartin and tuberin, which form a functional signaling complex. Disruption of these genes in the brain results in abnormal cellular differentiation, migration, and proliferation, giving rise to the charac- teristic brain lesions of tuberous sclerosis complex (TSC) called cortical tubers. The most devastating complications of TSC affect the central nervous system and include epilepsy, mental retardation, autism, and glial tumors. Relevant animal models, including conventional and conditional knockout mice, are valuable tools for studying the normal func- tions of tuberin and hamartin and the way in which disruption of their expression gives rise to the variety of clinical features that characterize TSC. In the future, these animals will be invaluable preclinical models for the development of highly specific and efficacious treatments for children affected with TSC. KEY WORDS • tuberous sclerosis • pathogenesis • animal model Tuberous sclerosis complex is an autosomal-dominant zures, mental retardation, and autism.1,51 The number and tumor predisposition syndrome that affects approximately location of cortical tubers and the patient’s age at seizure 1 in 7500 individuals worldwide.19 The TSC is character- onset are highly correlated with neurological outcomes.1,30 ized by benign hamartomatous growths in multiple organs, Epilepsy occurs in approximately 80% of affected indi- including the kidney, skin, retina, lung, and brain.
    [Show full text]
  • The Effects of Oncogenic G12d Mutation on K-Ras Structure, Conformation and Dynamics
    bioRxiv preprint doi: https://doi.org/10.1101/178483; this version posted August 19, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. THE EFFECTS OF ONCOGENIC G12D MUTATION ON K-RAS STRUCTURE, CONFORMATION AND DYNAMICS Sezen Vatansever1, 2, 3, Zeynep H. Gümüş2, 3*, Burak Erman1* 1Department of Chemical and Biological Engineering, College of Engineering, Koç University, Rumelifeneri Yolu, 34450, Sarıyer, Istanbul, Turkey 2Department of Genetics and Genomics, 3Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 *Correspondence: [email protected] (B.E.) and [email protected] (Z.H.G.) ABSTRACT K-Ras is the most frequently mutated protein in human tumors. Activating K-Ras mutations drive cancer initiation, progression and drug resistance, directly leading to nearly a million deaths per year. To understand the mechanisms by which mutations alter K-Ras function, we need to understand their effects on protein dynamics. However, despite decades of research, how oncogenic mutations in K-Ras alter its conformation and dynamics remain to be understood. Here, we present how the most recurrent K-Ras oncogenic mutation, G12D, leads to structural, conformational and dynamical changes that lead to constitutively active K- Ras. We have developed a new integrated MD simulation data analysis approach to quantify such changes in a protein and applied it to K-Ras. Our results show that G12D mutation induces strong negative correlations between the fluctuations of SII and those of the P-loop, Switch I (SI) and α3 regions in K-RasG12D.
    [Show full text]