Calmodulin and PI3K Signaling in KRAS Cancers.

[1]  J. McDonald,et al.  The insulin receptor and calmodulin. Calmodulin enhances insulin-mediated receptor kinase activity and insulin stimulates phosphorylation of calmodulin. , 1986, The Journal of biological chemistry.

[2]  D. Sacks,et al.  Characteristics of calmodulin phosphorylation by the insulin receptor kinase. , 1988, Endocrinology.

[3]  D. Sacks,et al.  Insulin-stimulated phosphorylation of calmodulin by rat liver insulin receptor preparations. , 1988, The Journal of biological chemistry.

[4]  H. Nakano,et al.  In vitro tyrosine phosphorylation studies on RAS proteins and calmodulin suggest that polylysine-like basic peptides or domains may be involved in interactions between insulin receptor kinase and its substrate. , 1989, Proceedings of the National Academy of Sciences of the United States of America.

[5]  D. Sacks,et al.  Tyrosine-specific phosphorylation of calmodulin by the insulin receptor kinase purified from human placenta. , 1989, The Biochemical journal.

[6]  K. Glenn,et al.  The carboxyl terminal segment of the c-Ki-ras 2 gene product mediates insulin-stimulated phosphorylation of calmodulin and stimulates insulin-independent autophosphorylation of the insulin receptor. , 1989, Biochemical and biophysical research communications.

[7]  D. Sacks,et al.  Calmodulin as Substrate for Insulin-Receptor Kinase: Phosphorylation by Receptors From Rat Skeletal Muscle , 1989, Diabetes.

[8]  C. Kahn,et al.  Expression and function of IRS-1 in insulin signal transmission. , 1992, The Journal of biological chemistry.

[9]  B. Margolis,et al.  Phosphatidylinositol 3′‐kinase is activated by association with IRS‐1 during insulin stimulation. , 1992, The EMBO journal.

[10]  L. Cantley,et al.  Phosphoinositide 3-kinase is activated by phosphopeptides that bind to the SH2 domains of the 85-kDa subunit. , 1993, The Journal of biological chemistry.

[11]  J. Schlessinger,et al.  Specific phosphopeptide binding regulates a conformational change in the PI 3‐kinase SH2 domain associated with enzyme activation. , 1993, The EMBO journal.

[12]  R. Sharma,et al.  Tyrosine-phosphorylated calmodulin has reduced biological activity. , 1994, Archives of biochemistry and biophysics.

[13]  D. Sacks Alteration of calmodulin-protein interactions by a monoclonal antibody to calmodulin. , 1994, Biochimica et biophysica acta.

[14]  A. Benguría,et al.  Phosphorylation of calmodulin by the epidermal-growth-factor-receptor tyrosine kinase. , 1994, European journal of biochemistry.

[15]  Peter J. Parker,et al.  The activation of phosphatidylinositol 3-kinase by Ras , 1994, Current Biology.

[16]  R. Thoma,et al.  Identification of insulin-stimulated phosphorylation sites on calmodulin. , 1996, Biochemistry.

[17]  M. White,et al.  Calmodulin Activates Phosphatidylinositol 3-Kinase* , 1997, The Journal of Biological Chemistry.

[18]  G. Benaim,et al.  Comparative phosphorylation of calmodulin from trypanosomatids and bovine brain by calmodulin-binding protein kinases. , 1998, Comparative biochemistry and physiology. Part C, Pharmacology, toxicology & endocrinology.

[19]  A. Villalobo,et al.  A method for the purification of phospho(Tyr)calmodulin free of nonphosphorylated calmodulin. , 1999, Protein expression and purification.

[20]  A. Means,et al.  Calmodulin: a prototypical calcium sensor. , 2000, Trends in cell biology.

[21]  C. Marshall,et al.  Calmodulin Binds to K-Ras, but Not to H- or N-Ras, and Modulates Its Downstream Signaling , 2001, Molecular and Cellular Biology.

[22]  M. Waterfield,et al.  Synthesis and function of 3-phosphorylated inositol lipids. , 2001, Annual review of biochemistry.

[23]  G. Benaim,et al.  Phosphorylation of calmodulin. Functional implications. , 2002, European journal of biochemistry.

[24]  Vojo Deretic,et al.  Tuberculosis Toxin Blocking Phagosome Maturation Inhibits a Novel Ca2+/Calmodulin-PI3K hVPS34 Cascade , 2003, The Journal of experimental medicine.

[25]  Valéria Szukacsov,et al.  Lysophosphatidylcholine is a regulator of tyrosine kinase activity and intracellular Ca(2+) level in Jurkat T cell line. , 2004, Immunology letters.

[26]  J. Comella,et al.  Glial Cell Line-derived Neurotrophic Factor Increases Intracellular Calcium Concentration , 2004, Journal of Biological Chemistry.

[27]  C. Thompson,et al.  PKC regulates a farnesyl-electrostatic switch on K-Ras that promotes its association with Bcl-XL on mitochondria and induces apoptosis. , 2006, Molecular cell.

[28]  S. Planchon,et al.  Growth Factor-dependent AKT Activation and Cell Migration Requires the Function of c-K(B)-Ras Versus Other Cellular Ras Isoforms* , 2006, Journal of Biological Chemistry.

[29]  B. Vanhaesebroeck,et al.  Class IA phosphoinositide 3-kinases are obligate p85-p110 heterodimers , 2007, Proceedings of the National Academy of Sciences.

[30]  K. Kinzler,et al.  The Structure of a Human p110a/p85a Complex Elucidates the Effects of Oncogenic PI3Ka Mutations , 2007 .

[31]  Bert Vogelstein,et al.  The Structure of a Human p110α/p85α Complex Elucidates the Effects of Oncogenic PI3Kα Mutations , 2007, Science.

[32]  G. Stamp,et al.  Binding of Ras to Phosphoinositide 3-Kinase p110α Is Required for Ras- Driven Tumorigenesis in Mice , 2007, Cell.

[33]  Yuval Inbar,et al.  Mechanism of Two Classes of Cancer Mutations in the Phosphoinositide 3-Kinase Catalytic Subunit , 2007, Science.

[34]  Jing Xu,et al.  Subtoxic N‐methyl‐D‐aspartate delayed neuronal death in ischemic brain injury through TrkB receptor‐ and calmodulin‐mediated PI‐3K/Akt pathway activation , 2007, Hippocampus.

[35]  R. Copeland,et al.  Effects of oncogenic p110alpha subunit mutations on the lipid kinase activity of phosphoinositide 3-kinase. , 2008, The Biochemical journal.

[36]  Li Zhao,et al.  Helical domain and kinase domain mutations in p110α of phosphatidylinositol 3-kinase induce gain of function by different mechanisms , 2008, Proceedings of the National Academy of Sciences.

[37]  L. Zhao,et al.  Class I PI3K in oncogenic cellular transformation , 2008, Oncogene.

[38]  R. Copeland,et al.  Effects of oncogenic p110α subunit mutations on the lipid kinase activity of phosphoinositide 3-kinase , 2008 .

[39]  O. Bachs,et al.  Identification of Essential Interacting Elements in K-Ras/Calmodulin Binding and Its Role in K-Ras Localization* , 2008, Journal of Biological Chemistry.

[40]  L. Anderson,et al.  The hypervariable region of K-Ras4B is responsible for its specific interactions with calmodulin. , 2009, Biochemistry.

[41]  K. Kinzler,et al.  A frequent kinase domain mutation that changes the interaction between PI3Kα and the membrane , 2009, Proceedings of the National Academy of Sciences.

[42]  Karim Jellali,et al.  Calmodulin‐mediated regulation of the epidermal growth factor receptor , 2010, The FEBS journal.

[43]  M. Drosten,et al.  K-Ras4B phosphorylation at Ser181 is inhibited by calmodulin and modulates K-Ras activity and function , 2010, Oncogene.

[44]  Zoologie Glial Cell Line-Derived Neurotrophic Factor , 2011 .

[45]  John E. Burke,et al.  Structural Basis for Activation and Inhibition of Class I Phosphoinositide 3-Kinases , 2011, Science Signaling.

[46]  Jie Chen,et al.  Both the C-Terminal Polylysine Region and the Farnesylation of K-RasB Are Important for Its Specific Interaction with Calmodulin , 2011, PloS one.

[47]  N. Agell,et al.  CaM interaction and Ser181 phosphorylation as new K-Ras signaling modulators , 2011, Small GTPases.

[48]  Yong Zhou,et al.  Nonsteroidal Anti-inflammatory Drugs Alter the Spatiotemporal Organization of Ras Proteins on the Plasma Membrane* , 2012, The Journal of Biological Chemistry.

[49]  Carla Mattos,et al.  A comprehensive survey of Ras mutations in cancer. , 2012, Cancer research.

[50]  Glenn R Masson,et al.  Oncogenic mutations mimic and enhance dynamic events in the natural activation of phosphoinositide 3-kinase p110α (PIK3CA) , 2012, Proceedings of the National Academy of Sciences.

[51]  P. Bastiaens,et al.  The GDI-like solubilizing factor PDEδ sustains the spatial organization and signalling of Ras family proteins , 2011, Nature Cell Biology.

[52]  P. Bastiaens,et al.  The GDI-like solubilizing factor PDEδ sustains the spatial organization and signalling of Ras family proteins , 2012, Nature Cell Biology.

[53]  F. Bermejo-Pareja,et al.  Altered calmodulin degradation and signaling in non-neuronal cells from Alzheimer's disease patients. , 2012, Current Alzheimer research.

[54]  Activation of PI3Kα by physiological effectors and by oncogenic mutations: structural and dynamic effects , 2014, Biophysical Reviews.

[55]  V. Gaponenko,et al.  Application of Reductive 13C-Methylation of Lysines to Enhance the Sensitivity of Conventional NMR Methods , 2013, Molecules.

[56]  I. Prior,et al.  Oncogenic K-ras segregates at spatially distinct plasma membrane signaling platforms according to its phosphorylation status , 2013, Journal of Cell Science.

[57]  M. Berchtold,et al.  The many faces of calmodulin in cell proliferation, programmed cell death, autophagy, and cancer. , 2014, Biochimica et biophysica acta.

[58]  R. Xiang,et al.  Emerging roles of the p38 MAPK and PI3K/AKT/mTOR pathways in oncogene-induced senescence. , 2014, Trends in biochemical sciences.

[59]  Ozlem Keskin,et al.  Plasma membrane regulates Ras signaling networks , 2015, Cellular logistics.

[60]  Ozlem Keskin,et al.  Principles of K-Ras effector organization and the role of oncogenic K-Ras in cancer initiation through G1 cell cycle deregulation , 2015, Expert review of proteomics.

[61]  Ozlem Keskin,et al.  The Key Role of Calmodulin in KRAS-Driven Adenocarcinomas , 2015, Molecular Cancer Research.

[62]  A. Cox,et al.  K-Ras4A splice variant is widely expressed in cancer and uses a hybrid membrane-targeting motif , 2015, Proceedings of the National Academy of Sciences.

[63]  A. Balmain,et al.  K-Ras Promotes Tumorigenicity through Suppression of Non-canonical Wnt Signaling , 2015, Cell.

[64]  M. Menéndez,et al.  Characterization of Phospho-(Tyrosine)-Mimetic Calmodulin Mutants , 2015, PloS one.

[65]  A. Benguría,et al.  The activating role of phospho-(Tyr)-calmodulin on the epidermal growth factor receptor. , 2015, The Biochemical journal.

[66]  R. Nussinov,et al.  Mechanisms of Membrane Binding of Small GTPase K-Ras4B Farnesylated Hypervariable Region* , 2015, The Journal of Biological Chemistry.

[67]  J. Griffiths,et al.  Absolute Quantification of Endogenous Ras Isoform Abundance , 2015, PloS one.

[68]  G. Benaim,et al.  Ca2+/Calmodulin and Apo-Calmodulin Both Bind to and Enhance the Tyrosine Kinase Activity of c-Src , 2015, PloS one.

[69]  Ruth Nussinov,et al.  High-Affinity Interaction of the K-Ras4B Hypervariable Region with the Ras Active Site , 2015, Biophysical journal.

[70]  M. Lavin,et al.  β-Adducin siRNA disruption of the spectrin-based cytoskeleton in differentiating keratinocytes prevented by calcium acting through calmodulin/epidermal growth factor receptor/cadherin pathway. , 2015, Cellular signalling.

[71]  C. Der,et al.  Targeting RAS-mutant cancers: is ERK the key? , 2015, Trends in cancer.

[72]  C. Der,et al.  Targeting RAS Membrane Association: Back to the Future for Anti-RAS Drug Discovery? , 2015, Clinical Cancer Research.

[73]  Kendra Marcus,et al.  Direct Attack on RAS: Intramolecular Communication and Mutation-Specific Effects , 2015, Clinical Cancer Research.

[74]  Ruth Nussinov,et al.  A New View of Ras Isoforms in Cancers. , 2016, Cancer research.

[75]  T Aittokallio,et al.  Cancer stem cell drugs target K-ras signaling in a stemness context , 2016, Oncogene.

[76]  Suyong Choi,et al.  The Hidden Conundrum of Phosphoinositide Signaling in Cancer. , 2016, Trends in cancer.

[77]  Ruth Nussinov,et al.  Oncogenic KRAS signaling and YAP1/β-catenin: Similar cell cycle control in tumor initiation. , 2016, Seminars in cell & developmental biology.

[78]  L. Birnbaumer,et al.  Membrane translocation of TRPC6 channels and endothelial migration are regulated by calmodulin and PI3 kinase activation , 2016, Proceedings of the National Academy of Sciences.

[79]  Frank McCormick,et al.  K-Ras protein as a drug target , 2016, Journal of Molecular Medicine.

[80]  Herbert Waldmann,et al.  Regulation of K-Ras4B Membrane Binding by Calmodulin. , 2016, Biophysical journal.

[81]  R. Ghirlando,et al.  Structural basis of recognition of farnesylated and methylated KRAS4b by PDEδ , 2016, Proceedings of the National Academy of Sciences.

[82]  D. Calvisi,et al.  Comparison of liver oncogenic potential among human RAS isoforms , 2016, Oncotarget.

[83]  Wei Zhang,et al.  Identification of initial leads directed at the calmodulin-binding region on the Src-SH2 domain that exhibit anti-proliferation activity against pancreatic cancer. , 2016, Bioorganic & medicinal chemistry letters.

[84]  Ozlem Keskin,et al.  Ras Conformational Ensembles, Allostery, and Signaling. , 2016, Chemical reviews.

[85]  M. Hall,et al.  mTOR Signaling Confers Resistance to Targeted Cancer Drugs. , 2016, Trends in cancer.

[86]  Ozlem Keskin,et al.  K-Ras4B/calmodulin/PI3Kα: A promising new adenocarcinoma-specific drug target? , 2016, Expert opinion on therapeutic targets.

[87]  Ruth Nussinov,et al.  Independent and core pathways in oncogenic KRAS signaling , 2016, Expert review of proteomics.

[88]  Ruth Nussinov,et al.  The higher level of complexity of K‐Ras4B activation at the membrane , 2016, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[89]  Ruth Nussinov,et al.  The disordered hypervariable region and the folded catalytic domain of oncogenic K-Ras4B partner in phospholipid binding. , 2016, Current opinion in structural biology.

[90]  R. Nussinov,et al.  Comparison of the Conformations of KRAS Isoforms, K-Ras4A and K-Ras4B, Points to Similarities and Significant Differences. , 2016, The journal of physical chemistry. B.

[91]  Ruth Nussinov,et al.  Drugging Ras GTPase: a comprehensive mechanistic and signaling structural view. , 2016, Chemical Society reviews.

[92]  Xu Shen,et al.  SP6616 as a new Kv2.1 channel inhibitor efficiently promotes β-cell survival involving both PKC/Erk1/2 and CaM/PI3K/Akt signaling pathways , 2016, Cell Death and Disease.