151
|
Abstract
Despite recent advances, the biology underlying nevogenesis remains unclear. Activating mutations in NRAS, HRAS, BRAF, and GNAQ have been identified in benign nevi. Their presence roughly correlates with congenital, Spitz, acquired, and blue nevi, respectively. These mutations are likely to play a critical role in driving nevogenesis. While each mutation is able to activate the MAP kinase pathway, they also interact with a host of different proteins in other pathways. The different melanocytic developmental pathways activated by each mutation cause the cells to migrate, proliferate, and differentiate to different extents within the skin. This causes each mutation to give rise to a characteristic growth pattern. The exact location and differentiation state of the cell of origin for benign moles remains to be discovered. Further research is necessary to fully understand nevus development given that most of the same developmental pathways are also present in melanoma.
Collapse
|
152
|
Ras Isoprenylation and pAkt Inhibition by Zoledronic Acid and Fluvastatin Enhances Paclitaxel Activity in T24 Bladder Cancer Cells. Cancers (Basel) 2011; 3:662-74. [PMID: 24212635 PMCID: PMC3756383 DOI: 10.3390/cancers3010662] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Revised: 01/30/2011] [Accepted: 02/09/2011] [Indexed: 11/16/2022] Open
Abstract
Background Bisphosphonates interfere with the mevalonate pathway and inhibit the prenylation of small GTP-binding proteins such as ras and rap. We hypothesized that zoledronic acid would synergistically inhibit T24 bladder cancer cell growth in combination with fluvastatin and paclitaxel. Methods Increasing doses of fluvastatin, zoledronic acid, and paclitaxel were investigated as single agents and in combination, and synergistic interactions were evaluated by the Chou-Talalay method. Western blots were used to assess effects on signal transduction pathways. Results Growth of T24 was significantly inhibited with IC50 values of 2.67 ± 0.61 μM for fluvastatin and 5.35 ± 1.35 μM for zoledronic acid after 72 hours treatment. Geranylgeranyl pyrophosphate and farnesyl pyrophosphate was able to block, in part, this inhibitory activity. The combinations of zoledronic acid and paclitaxel, zoledronic acid and fluvastatin, and fluvastatin and paclitaxel were all synergistic. Both fluvastatin and zoledronic acid inhibited Ras and Rap prenylation, and the phosphorylation of ERK1/2 and AKT. The degree of inhibition of phosphorylation of these key signaling transduction pathways appears to closely correlate with their synergistic interactions. Conclusions Zoledronic acid enhances fluvastatin and paclitaxel activity against T24 in a synergistic manner and this is mediated largely by inhibition of both the Ras/Raf/MEK/ERK and PI3K/AKT signaling pathways via isoprenylation inhibition.
Collapse
|
153
|
Mukhopadhyay A, Krishnaswami SR, Yu BDY. Activated Kras alters epidermal homeostasis of mouse skin, resulting in redundant skin and defective hair cycling. J Invest Dermatol 2011; 131:311-9. [PMID: 20944652 PMCID: PMC3335744 DOI: 10.1038/jid.2010.296] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Germline mutations in the RAS-mitogen-activated protein kinase (RAS/MAPK) pathway are associated with genodermatoses, characterized by cutaneous, cardiac, and craniofacial defects, and cancer predisposition. Whereas activating mutations in HRAS are associated with the vast majority of patients with Costello syndrome, mutations in its paralog, KRAS, are rare. To better understand the disparity among RAS paralogs in human syndromes, we generated mice that activate a gain-of-function Kras allele (Lox-Stop-Lox (LSL)-Kras(G12D)) in ectodermal tissue using two different Cre transgenic lines. Using Msx2-Cre or ligand-inducible keratin 15 (K15)-CrePR, the embryonic effects of activated Kras were bypassed and the effects of Kras(G12D) expression from its endogenous promoter were determined. We found that Kras(G12D) induced redundant skin, papillomas, shortened nails, and hair loss. Redundant skin was associated with basal keratinocyte hyperplasia and an increase in body surface area. Paradoxically, Kras(G12D) also prevented hair cycle activation. We find that Kras(G12D) blocks proliferation in the bulge region of the hair follicle, when activated through Msx2-Cre but not through K15-CrePR. These studies reveal that KRAS, although infrequently involved in RAS/MAPK syndromes, is capable of inducing multiple cutaneous features that grossly resemble human RAS/MAPK syndromes.
Collapse
Affiliation(s)
- Anandaroop Mukhopadhyay
- Division of Dermatology, Department of Medicine, UCSD Stem Cell Program, and Institute for Genomic Medicine, University of California, San Diego, San Diego, California, USA
| | - Suguna R. Krishnaswami
- Division of Dermatology, Department of Medicine, UCSD Stem Cell Program, and Institute for Genomic Medicine, University of California, San Diego, San Diego, California, USA
| | - Benjamin D.-Y. Yu
- Division of Dermatology, Department of Medicine, UCSD Stem Cell Program, and Institute for Genomic Medicine, University of California, San Diego, San Diego, California, USA
| |
Collapse
|
154
|
Abstract
Ras GTPases are best known for their ability to serve as molecular switches regulating cell growth, differentiation and survival. Gene mutations that result in expression of constitutively active forms of Ras have been linked to oncogenesis in animal models and humans. However, over the past two decades, evidence has gradually accumulated to support a paradoxical role for Ras proteins in the initiation of cell death pathways. In this review we survey the literature pointing to the ability of activated Ras to promote cell death under conditions where cancer cells encounter apoptotic stimuli or Ras is ectopically expressed. In some of these cases Ras acts through known effectors and well defined apoptotic death pathways. However, in other cases it appears that Ras operates by triggering novel non-apoptotic death mechanisms that are just beginning to be characterized. Understanding these mechanisms and the factors that go into changing the nature of Ras signaling from pro-survival to pro-death could set the stage for development of novel therapeutic approaches aimed at manipulating pro-death Ras signaling pathways in cancer.
Collapse
Affiliation(s)
- Jean H Overmeyer
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine, Toledo, Ohio 43614, USA
| | | |
Collapse
|
155
|
Kompier LC, Lurkin I, van der Aa MNM, van Rhijn BWG, van der Kwast TH, Zwarthoff EC. FGFR3, HRAS, KRAS, NRAS and PIK3CA mutations in bladder cancer and their potential as biomarkers for surveillance and therapy. PLoS One 2010; 5:e13821. [PMID: 21072204 PMCID: PMC2972209 DOI: 10.1371/journal.pone.0013821] [Citation(s) in RCA: 238] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2010] [Accepted: 10/13/2010] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Fifty percent of patients with muscle-invasive bladder cancer (MI-BC) die from their disease and current chemotherapy treatment only marginally increases survival. Novel therapies targeting receptor tyrosine kinases or activated oncogenes may improve outcome. Hence, it is necessary to stratify patients based on mutations in relevant oncogenes. Patients with non-muscle-invasive bladder cancer (NMI-BC) have excellent survival, however two-thirds develop recurrences. Tumor specific mutations can be used to detect recurrences in urine assays, presenting a more patient-friendly diagnostic procedure than cystoscopy. METHODOLOGY/PRINCIPAL FINDINGS To address these issues, we developed a mutation assay for the simultaneous detection of 19 possible mutations in the HRAS, KRAS, and NRAS genes. With this assay and mutation assays for the FGFR3 and PIK3CA oncogenes, we screened primary bladder tumors of 257 patients and 184 recurrences from 54 patients. Additionally, in primary tumors p53 expression was obtained by immunohistochemistry. Of primary tumors 64% were mutant for FGFR3, 11% for RAS, 24% for PIK3CA, and 26% for p53. FGFR3 mutations were mutually exclusive with RAS mutations (p = 0.001) and co-occurred with PIK3CA mutations (p = 0.016). P53 overexpression was mutually exclusive with PIK3CA and FGFR3 mutations (p≤0.029). Mutations in the RAS and PIK3CA genes were not predictors for recurrence-free, progression-free and disease-specific survival. In patients presenting with NMI-BC grade 3 and MI-BC, 33 and 36% of the primary tumors were mutant. In patients with low-grade NMI-BC, 88% of the primary tumors carried a mutation and 88% of the recurrences were mutant. CONCLUSIONS/SIGNIFICANCE The mutation assays present a companion diagnostic to define patients for targeted therapies. In addition, the assays are a potential biomarker to detect recurrences during surveillance. We showed that 88% of patients presenting with low-grade NMI-BC are eligible for such a follow-up. This may contribute to a reduction in the number of cystoscopical examinations.
Collapse
Affiliation(s)
| | - Irene Lurkin
- Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | | | - Bas W. G. van Rhijn
- Department of Urology, University Health Network, Toronto General Hospital, Toronto, Canada
| | - Theo H. van der Kwast
- Department of Laboratory Medicine and Pathobiology, University Health Network, Toronto General Hospital, Toronto, Canada
| | | |
Collapse
|
156
|
de Launay D, Vreijling J, Hartkamp LM, Karpus ON, Abreu JRF, van Maanen MA, Sanders ME, Grabiec AM, Hamann J, Ørum H, Vervoordeldonk MJ, Fluiter K, Tak PP, Reedquist KA. Silencing the expression of Ras family GTPase homologues decreases inflammation and joint destruction in experimental arthritis. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:3010-24. [PMID: 20971740 DOI: 10.2353/ajpath.2010.091053] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Changes in the expression and activation status of Ras proteins are thought to contribute to the pathological phenotype of stromal fibroblast-like synoviocytes (FLS) in rheumatoid arthritis, a prototypical immune-mediated inflammatory disease. Broad inhibition of Ras and related proteins has shown protective effects in animal models of arthritis, but each of the Ras family homologues (ie, H-, K-, and N-Ras) makes distinct contributions to cellular activation. We examined the expression of each Ras protein in synovial tissue and FLS obtained from patients with rheumatoid arthritis and other forms of inflammatory arthritis. Each Ras protein was expressed in synovial tissue and cultured FLS. Each homolog was also activated following FLS stimulation with tumor necrosis factor-α or interleukin (IL)-1β. Constitutively active mutants of each Ras protein enhanced IL-1β-induced FLS matrix metalloproteinase-3 production, while only active H-Ras enhanced IL-8 production. Gene silencing demonstrated that each Ras protein contributed to IL-1β-dependent IL-6 production, while H-Ras and N-Ras supported IL-1β-dependent matrix metalloproteinase-3 and IL-8 production, respectively. The overlap in contributions of Ras homologues to FLS activation suggests that broad targeting of Ras GTPases in vivo suppresses global inflammation and joint destruction in arthritis. Consistent with this, simultaneous silencing of H-Ras, K-Ras, and N-Ras expression significantly reduces inflammation and joint destruction in murine collagen-induced arthritis, while specific targeting of N-Ras alone is less effective in providing clinical benefits.
Collapse
Affiliation(s)
- Daphne de Launay
- Division of Clinical Immunology and Rheumatology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
157
|
Zhang B, Li S, Harbrecht BG. Akt-mediated signaling is induced by cytokines and cyclic adenosine monophosphate and suppresses hepatocyte inducible nitric oxide synthase expression independent of MAPK P44/42. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1813:73-9. [PMID: 20934465 DOI: 10.1016/j.bbamcr.2010.10.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Revised: 09/29/2010] [Accepted: 10/01/2010] [Indexed: 11/26/2022]
Abstract
Cyclic AMP inhibits the expression of nitric oxide synthase (Harbrecht et al., 1995 [1]) in hepatocytes but the mechanism for this effect is incompletely understood. Cyclic AMP can activate several intracellular signaling pathways in hepatocytes including Protein Kinase A (PKA), cAMP regulated guanine nucleotide exchange factors (cAMP-GEFs), and calcium-mediated Protein Kinases. There is considerable overlap and cross-talk between many of these signaling pathways, however, and how these cascades regulate hepatocyte iNOS is not known. We hypothesized that Akt mediates the effect of cAMP on hepatocyte iNOS expression. Hepatocytes cultured with cytokines and dbcAMP increased Akt phosphorylation up to 2h of culture. Akt phosphorylation was inhibited by the PI3K inhibitor LY294002 (10μM), farnyltranferase inhibitor FTI-276, or transfection with a dominant negative Akt. The cyclic AMP-induced suppression of cytokine-stimulated iNOS was partially reversed by LY294002 and FTI-276. LY294002 also increased NFκB nucleus translocation by Western blot analysis in nuclear extracts. Cyclic AMP increased phosphorylation of Raf1 at serine 259 which was blocked by LY294002 and associated with decreased MAPK P44/42 phosphorylation. However, inhibition of MAPK P44/42 signaling with PD98059 failed to suppress cytokine-induced hepatocyte iNOS expression and did not enhance the inhibitory effect of dbcAMP on iNOS production. A constitutively active MAPK P44/42 plasmid had no effect on cytokine-stimulated NO production. These data demonstrate that dbcAMP regulates hepatocyte iNOS expression through an Akt-mediated signaling mechanism that is independent of MAPK P44/42.
Collapse
Affiliation(s)
- Baochun Zhang
- University of Louisville, Department of Surgery, Louisville, KY 40292, USA.
| | | | | |
Collapse
|
158
|
Lukman S, Grant BJ, Gorfe AA, Grant GH, McCammon JA. The distinct conformational dynamics of K-Ras and H-Ras A59G. PLoS Comput Biol 2010; 6:e1000922. [PMID: 20838576 PMCID: PMC2936511 DOI: 10.1371/journal.pcbi.1000922] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2010] [Accepted: 08/06/2010] [Indexed: 02/07/2023] Open
Abstract
Ras proteins regulate signaling cascades crucial for cell proliferation and differentiation by switching between GTP- and GDP-bound conformations. Distinct Ras isoforms have unique physiological functions with individual isoforms associated with different cancers and developmental diseases. Given the small structural differences among isoforms and mutants, it is currently unclear how these functional differences and aberrant properties arise. Here we investigate whether the subtle differences among isoforms and mutants are associated with detectable dynamical differences. Extensive molecular dynamics simulations reveal that wild-type K-Ras and mutant H-Ras A59G are intrinsically more dynamic than wild-type H-Ras. The crucial switch 1 and switch 2 regions along with loop 3, helix 3, and loop 7 contribute to this enhanced flexibility. Removing the gamma-phosphate of the bound GTP from the structure of A59G led to a spontaneous GTP-to-GDP conformational transition in a 20-ns unbiased simulation. The switch 1 and 2 regions exhibit enhanced flexibility and correlated motion when compared to non-transitioning wild-type H-Ras over a similar timeframe. Correlated motions between loop 3 and helix 5 of wild-type H-Ras are absent in the mutant A59G reflecting the enhanced dynamics of the loop 3 region. Taken together with earlier findings, these results suggest the existence of a lower energetic barrier between GTP and GDP states of the mutant. Molecular dynamics simulations combined with principal component analysis of available Ras crystallographic structures can be used to discriminate ligand- and sequence-based dynamic perturbations with potential functional implications. Furthermore, the identification of specific conformations associated with distinct Ras isoforms and mutants provides useful information for efforts that attempt to selectively interfere with the aberrant functions of these species.
Collapse
Affiliation(s)
- Suryani Lukman
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
- Bioinformatics Institute, Agency for Science, Technology and Research, Singapore
| | - Barry J. Grant
- Department of Chemistry and Biochemistry, Center for Theoretical Biological Physics, University of California San Diego, La Jolla, California, United States of America
- Howard Hughes Medical Institute, University of California San Diego, La Jolla, California, United States of America
| | - Alemayehu A. Gorfe
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas, United States of America
| | - Guy H. Grant
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - J. Andrew McCammon
- Department of Chemistry and Biochemistry, Center for Theoretical Biological Physics, University of California San Diego, La Jolla, California, United States of America
- Department of Pharmacology, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
159
|
Hou DX, Kumamoto T. Flavonoids as protein kinase inhibitors for cancer chemoprevention: direct binding and molecular modeling. Antioxid Redox Signal 2010; 13:691-719. [PMID: 20070239 DOI: 10.1089/ars.2009.2816] [Citation(s) in RCA: 143] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Protein kinases play crucial roles in the regulation of multiple cell signaling pathways and cellular functions. Deregulation of protein kinase function has been implicated in carcinogenesis. The inhibition of protein kinases has emerged as an important target for cancer chemoprevention and therapy. Accumulated data revealed that flavonoids exert chemopreventive effects through acting at protein kinase signaling pathways, more than as conventional hydrogen-donating antioxidants. Recent studies show that flavonoids can bind directly to some protein kinases, including Akt/protein kinase B (Akt/PKB), Fyn, Janus kinase 1 (JAK1), mitogen-activated protein kinase kinase 1 (MEK1), phosphoinositide 3-kinase (PI3K), mitogen-activated protein (MAP) kinase kinase 4 (MKK4), Raf1, and zeta chain-associated 70-kDa protein (ZAP-70) kinase, and then alter their phosphorylation state to regulate multiple cell signaling pathways in carcinogenesis processes. In this review, we report recent results on the interactions of flavonoids and protein kinases, especially their direct binding and molecular modeling. The data suggest that flavonoids act as protein kinase inhibitors for cancer chemoprevention that were thought previously as conventional hydrogen-donating antioxidant. Moreover, the molecular modeling data show some hints for creating natural compound-based protein kinase inhibitors for cancer chemoprevention and therapy.
Collapse
Affiliation(s)
- De-Xing Hou
- The United Graduate School of Agricultural Sciences, Faculty of Agriculture, Kagoshima University, Kagoshima City, Japan
| | | |
Collapse
|
160
|
K-Ras4B phosphorylation at Ser181 is inhibited by calmodulin and modulates K-Ras activity and function. Oncogene 2010; 29:5911-22. [PMID: 20802526 DOI: 10.1038/onc.2010.298] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Fine tuning of Ras activity is widely known as a mechanism to induce different cellular responses. Recently, we have shown that calmodulin (CaM) binds to K-Ras and that K-Ras phosphorylation inhibits its interaction with CaM. In this study we report that CaM inhibits K-Ras phosphorylation at Ser181 by protein kinase C (PKC) in vivo, and this is a mechanism to modulate K-Ras activity and signaling. Although CaM inhibition increased the activation of endogenous K-Ras, PKC inhibition decreased its activation status. We demonstrate that K-Ras phosphorylation decreased susceptibility to p120GAP activity. Accordingly, we also observed that non-phosphorylable K-Ras mutant exhibits a less sustained activation profile and do not efficiently activate AKT at low growth factor doses compared with wild-type K-Ras. It is interesting that the physiological responses induced by K-Ras are affected by this phosphorylation; when K-Ras cannot be phosphorylated it exhibits a remarkably decreased ability to stimulate proliferation in non-saturated serum conditions. Finally, we demonstrate that phosphorylation also regulates oncogenic K-Ras functions, as focus formation capacity, mobility and apoptosis resistance upon adriamycin treatment of cells expressing oncogenic K-Ras that cannot be phosphorylated are highly compromised. Moreover, at low serum concentration proliferation and survival is practically inhibited when cells cannot phosphorylate oncogenic K-Ras. In this condition, K-Ras phosphorylation is essential to ensure a proper activation of mitogen-activated protein kinase and PI3K/AKT pathways. In summary, our findings suggest that the interplay between CaM interaction and PKC phosphorylation is essential to regulate non-oncogenic and oncogenic K-Ras activity and functionality.
Collapse
|
161
|
Ras membrane orientation and nanodomain localization generate isoform diversity. Proc Natl Acad Sci U S A 2010; 107:1130-5. [PMID: 20080631 DOI: 10.1073/pnas.0903907107] [Citation(s) in RCA: 184] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The structural elements encoding functional diversity among Ras GTPases are poorly defined. The orientation of the G domain of H-ras with respect to the plane of the plasma membrane is recognized by the Ras binding domain of C-Raf, coupling orientation to MAPK activation. We now show that two other proteins, phosphoinositide-3-kinase-alpha and the structurally unrelated galectin-1, also recognize G-domain orientation. These results rationalize the role of galectin-1 in generating active GTP-H-ras signaling nanoclusters. However, molecular dynamics simulations of K-ras membrane insertion and fluorescence lifetime imaging microscopy (FLIM)-Förster resonance energy transfer (FRET) imaging of the effector interactions of N-Ras, K-Ras, and M-ras suggest that there are two hyperactive, signaling-competent orientations of the Ras G domain. Mutational and functional analyses establish a clear relationship between effector binding and the amphilicities of helix alpha4 and the C-terminal hypervariable region, thus confirming that these structural elements critically tune the orientation of the Ras G domain. Finally, we show that G-domain orientation and nanoclustering synergize to generate Ras isoform specificity with respect to effector interactions.
Collapse
|
162
|
Castellano E, Downward J. Role of RAS in the regulation of PI 3-kinase. Curr Top Microbiol Immunol 2010; 346:143-69. [PMID: 20563706 DOI: 10.1007/82_2010_56] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Ras proteins are key regulators of signalling cascades, controlling many processes such as proliferation, differentiation and apoptosis. Mutations in these proteins or in their effectors, activators and regulators are associated with pathological conditions, particularly the development of various forms of human cancer. RAS proteins signal through direct interaction with a number of effector enzymes, one of the best characterized being type I phosphatidylinositol (PI) 3-kinases. Although the ability of RAS to control PI 3-kinase has long been well established in cultured cells, evidence for a role of the interaction of endogenous RAS with PI 3-kinase in normal and malignant cell growth in vivo has only been obtained recently. Mice with mutations in the PI 3-kinase catalytic p110a isoform that block its ability to interact with RAS are highly resistant to endogenous KRAS oncogene induced lung tumourigenesis and HRAS oncogene induced skin carcinogenesis. Cells from these mice show proliferative defects and selective disruption of signalling from certain growth factors to PI 3-kinase, while the mice also display delayed development of the lymphatic vasculature. The interaction of RAS with p110a is thus required in vivo for some normal growth factor signalling and also for RAS-driven tumour formation. RAS family members were among the first oncogenes identified over 40 years ago. In the late 1960s, the rat-derived Harvey and Kirsten murine sarcoma retroviruses were discovered and subsequently shown to promote cancer formation through related oncogenes, termed RAS (from rat sarcoma virus). The central role of RAS proteins in human cancer is highlighted by the large number of tumours in which they are activated by mutation: approximately 20% of human cancers carry a mutation in RAS proteins. Because of the complex signalling network in which RAS operates, with multiple activators and effectors, each with a different pattern of tissue-specific expression and a distinct set of intracellular functions, one of the critical issues concerns the specific role of each effector in RAS-driven oncogenesis. In this chapter, we summarize current knowledge about how RAS regulates one of its best-known effectors, phosphoinositide 3-kinase (PI3K).
Collapse
Affiliation(s)
- Esther Castellano
- Signal Transduction Laboratory, Cancer Research UK London Research Institute, London, WC2A 3PX, UK
| | | |
Collapse
|
163
|
Lau KS, Haigis KM. Non-redundancy within the RAS oncogene family: insights into mutational disparities in cancer. Mol Cells 2009; 28:315-20. [PMID: 19812895 PMCID: PMC3976423 DOI: 10.1007/s10059-009-0143-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Accepted: 09/11/2009] [Indexed: 12/11/2022] Open
Abstract
The RAS family of oncoproteins has been studied extensively for almost three decades. While we know that activation of RAS represents a key feature of malignant transformation for many cancers, we are only now beginning to understand the complex underpinnings of RAS biology. Here, we will discuss emerging cancer genome sequencing data in the context of what is currently known about RAS function. Taken together, retrospective studies of primary human tissues and prospective studies of experimental models support the notion that the variable mutation frequencies exhibited by the RAS oncogenes reflect unique functions of the RAS oncoproteins.
Collapse
Affiliation(s)
- Ken S. Lau
- Molecular Pathology Unit and Center for Cancer Research, Massachusetts General Hospital and Department of Pathology, Harvard Medical School, USA
| | - Kevin M. Haigis
- Molecular Pathology Unit and Center for Cancer Research, Massachusetts General Hospital and Department of Pathology, Harvard Medical School, USA
| |
Collapse
|
164
|
Sartore-Bianchi A, Di Nicolantonio F, Nichelatti M, Molinari F, De Dosso S, Saletti P, Martini M, Cipani T, Marrapese G, Mazzucchelli L, Lamba S, Veronese S, Frattini M, Bardelli A, Siena S. Multi-determinants analysis of molecular alterations for predicting clinical benefit to EGFR-targeted monoclonal antibodies in colorectal cancer. PLoS One 2009; 4:e7287. [PMID: 19806185 PMCID: PMC2750753 DOI: 10.1371/journal.pone.0007287] [Citation(s) in RCA: 209] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2009] [Accepted: 09/02/2009] [Indexed: 12/15/2022] Open
Abstract
Background KRAS mutations occur in 35–45% of metastatic colorectal cancers (mCRC) and preclude responsiveness to EGFR-targeted therapy with cetuximab or panitumumab. However, less than 20% patients displaying wild-type KRAS tumors achieve objective response. Alterations in other effectors downstream of the EGFR, such as BRAF, and deregulation of the PIK3CA/PTEN pathway have independently been found to give rise to resistance. We present a comprehensive analysis of KRAS, BRAF, PIK3CA mutations, and PTEN expression in mCRC patients treated with cetuximab or panitumumab, with the aim of clarifying the relative contribution of these molecular alterations to resistance. Methodology/Principal Findings We retrospectively analyzed objective tumor response, progression-free (PFS) and overall survival (OS) together with the mutational status of KRAS, BRAF, PIK3CA and expression of PTEN in 132 tumors from cetuximab or panitumumab treated mCRC patients. Among the 106 non-responsive patients, 74 (70%) had tumors with at least one molecular alteration in the four markers. The probability of response was 51% (22/43) among patients with no alterations, 4% (2/47) among patients with 1 alteration, and 0% (0/24) for patients with ≥2 alterations (p<0.0001). Accordingly, PFS and OS were increasingly worse for patients with tumors harboring none, 1, or ≥2 molecular alteration(s) (p<0.001). Conclusions/Significance When expression of PTEN and mutations of KRAS, BRAF and PIK3CA are concomitantly ascertained, up to 70% of mCRC patients unlikely to respond to anti-EGFR therapies can be identified. We propose to define as ‘quadruple negative’, the CRCs lacking alterations in KRAS, BRAF, PTEN and PIK3CA. Comprehensive molecular dissection of the EGFR signaling pathways should be considered to select mCRC patients for cetuximab- or panitumumab-based therapies.
Collapse
Affiliation(s)
| | - Federica Di Nicolantonio
- Laboratory of Molecular Genetics, Division of Genetics and Oncogenomics, Institute for Cancer Research and Treatment (IRCC), University of Torino Medical School, Candiolo, Turin, Italy
| | | | - Francesca Molinari
- Laboratory of Molecular Diagnostic, Istituto Cantonale di Patologia, Locarno, Switzerland
| | - Sara De Dosso
- Oncology Institute of Southern Switzerland, Ospedale San Giovanni, Bellinzona, Switzerland
| | - Piercarlo Saletti
- Oncology Institute of Southern Switzerland, Ospedale San Giovanni, Bellinzona, Switzerland
| | - Miriam Martini
- Laboratory of Molecular Genetics, Division of Genetics and Oncogenomics, Institute for Cancer Research and Treatment (IRCC), University of Torino Medical School, Candiolo, Turin, Italy
| | - Tiziana Cipani
- The Falck Division of Medical Oncology, Ospedale Niguarda Ca' Granda, Milan, Italy
| | - Giovanna Marrapese
- The Falck Division of Medical Oncology, Ospedale Niguarda Ca' Granda, Milan, Italy
| | - Luca Mazzucchelli
- Laboratory of Molecular Diagnostic, Istituto Cantonale di Patologia, Locarno, Switzerland
| | - Simona Lamba
- Laboratory of Molecular Genetics, Division of Genetics and Oncogenomics, Institute for Cancer Research and Treatment (IRCC), University of Torino Medical School, Candiolo, Turin, Italy
| | - Silvio Veronese
- Division of Pathology, Ospedale Niguarda Ca' Granda, Milan, Italy
| | - Milo Frattini
- Laboratory of Molecular Diagnostic, Istituto Cantonale di Patologia, Locarno, Switzerland
| | - Alberto Bardelli
- Laboratory of Molecular Genetics, Division of Genetics and Oncogenomics, Institute for Cancer Research and Treatment (IRCC), University of Torino Medical School, Candiolo, Turin, Italy
- FIRC Institute of Molecular Oncology, Milan, Italy
- * E-mail: (SS); (AB)
| | - Salvatore Siena
- The Falck Division of Medical Oncology, Ospedale Niguarda Ca' Granda, Milan, Italy
- * E-mail: (SS); (AB)
| |
Collapse
|
165
|
Li R, Pourpak A, Morris SW. Inhibition of the insulin-like growth factor-1 receptor (IGF1R) tyrosine kinase as a novel cancer therapy approach. J Med Chem 2009; 52:4981-5004. [PMID: 19610618 PMCID: PMC2888655 DOI: 10.1021/jm9002395] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Rongshi Li
- Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Oncologic Sciences, University of South Florida, 12902 Magnolia Drive, Tampa, FL 33612
| | - Alan Pourpak
- Departments of Pathology and Oncology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105-3678 USA
| | - Stephan W. Morris
- Departments of Pathology and Oncology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105-3678 USA
| |
Collapse
|
166
|
Krueger F, Madeja Z, Hemberger M, McMahon M, Cook SJ, Gaunt SJ. Down-regulation of Cdx2 in colorectal carcinoma cells by the Raf-MEK-ERK 1/2 pathway. Cell Signal 2009; 21:1846-56. [PMID: 19686845 DOI: 10.1016/j.cellsig.2009.07.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Revised: 07/06/2009] [Accepted: 07/29/2009] [Indexed: 12/30/2022]
Abstract
Cdx2 is a homeodomain transcription factor that regulates normal intestinal cell differentiation. Cdx2 is frequently lost during progression of colorectal cancer (CRC) and is widely viewed as a colorectal tumour suppressor. A previous study suggested that activation of protein kinase C (PKC) may be responsible for Cdx2 down-regulation in CRC cells. Here we show that activation of PKC does indeed promote down-regulation of Cdx2 at both the mRNA and protein levels. However, PKC-dependent loss of Cdx2 is dependent upon activation of the Raf-MEK-ERK1/2 pathway. Indeed, specific activation of the ERK1/2 pathway using the conditional kinase DeltaRaf-1:ER is sufficient to inhibit Cdx2 transcription. The Raf-MEK-ERK1/2 pathway is hyper-activated in a large fraction of colorectal cancers due to mutations in K-Ras and we show that treatment of CRC cell lines with MEK inhibitors causes an increase in Cdx2 expression. Furthermore, activation of the ERK1/2 pathway promotes the phosphorylation and proteasome-dependent degradation of the Cdx2 protein. The inhibitory effect of ERK1/2 upon Cdx2 in CRC cells is in sharp contrast to its stimulatory effect upon Cdx2 expression in trophectoderm and trophoblast stem cells. These results provide important new insights into the regulation of the Cdx2 tumour suppressor by linking it to ERK1/2, a pathway which is frequently activated in CRC.
Collapse
Affiliation(s)
- Felix Krueger
- Laboratory of Developmental Genetics & Imprinting, The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | | | | | | | | | | |
Collapse
|
167
|
Moretó J, Vidal-Quadras M, Pol A, Santos E, Grewal T, Enrich C, Tebar F. Differential involvement of H- and K-Ras in Raf-1 activation determines the role of calmodulin in MAPK signaling. Cell Signal 2009; 21:1827-36. [PMID: 19666110 DOI: 10.1016/j.cellsig.2009.07.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Revised: 07/27/2009] [Accepted: 07/29/2009] [Indexed: 01/08/2023]
Abstract
We have previously demonstrated that inhibition of calmodulin (CaM) and the concomitant reduction of PI3K interfere with H-Ras-mediated activation of Raf-1 [1]. In the present study, we show that CaM has completely opposite effects on K-Ras-mediated Raf-1 activation. The differential contribution of CaM in the regulation of Raf-1 kinase activity via K- or H-Ras correlates with the stimulatory or inhibitory effect of CaM on MAPK phosphorylation depending on the cell type analyzed. FRET microscopy and biochemical analysis show that inhibition of CaM increases K-Ras-GTP levels and consequently its association with Raf-1. Though inhibition of CaM, using the CaM antagonist W-13, significantly increased Raf-1 activation by K-Ras-GTP, MAPK activation downstream K-Ras/Raf-1 was strongly reduced in COS-1 and several other cell lines. In contrast, in other cell lines such as NIH3T3-wt8, W-13-mediated inhibition of CaM increased Raf-1 activity, but resulted in an increase in MAPK phosphorylation. These findings suggest that modulation of K-Ras activity via CaM regulates MAPK signaling only in certain cell types. In support of this hypothesis, the comparison of H- and K-Ras expression, GTP loading and Raf-1 interaction in COS-1 and NIH3T3-wt8 suggests that the overall role of CaM in MAPK signal output is determined by the ratio of activated H- and K-Ras and the cell-specific contribution of each isoform in Raf-1 activation.
Collapse
Affiliation(s)
- Jemina Moretó
- Departament de Biologia Cel.lular, Immunologia i Neurociències, Facultat de Medicina, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Casanova 143, 08036-Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
168
|
Lee J, Kanatsu-Shinohara M, Morimoto H, Kazuki Y, Takashima S, Oshimura M, Toyokuni S, Shinohara T. Genetic Reconstruction of Mouse Spermatogonial Stem Cell Self-Renewal In Vitro by Ras-Cyclin D2 Activation. Cell Stem Cell 2009; 5:76-86. [DOI: 10.1016/j.stem.2009.04.020] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Revised: 04/05/2009] [Accepted: 04/27/2009] [Indexed: 12/24/2022]
|
169
|
Kim SH, Miller FR, Tait L, Zheng J, Novak RF. Proteomic and phosphoproteomic alterations in benign, premalignant and tumor human breast epithelial cells and xenograft lesions: biomarkers of progression. Int J Cancer 2009; 124:2813-28. [PMID: 19291795 PMCID: PMC4123863 DOI: 10.1002/ijc.24278] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The MCF10A human breast epithelial cell lineage includes the benign MCF10A cells, premalignant cells (MCF10AT, MCF10ATG3B) and malignant MCF10CA1a tumor cells. The premalignant and tumor cells recapitulate the progressive alterations associated with the temporal development of PBD and carcinoma. Ras protein levels were elevated by 6.9-, 22.4- and 32.2-fold in 10AT, 10ATG3B and 10CA1a cells, respectively, relative to 10A cells. K-Ras was not detected, N-Ras levels were unchanged; Rac and Rho levels increased in 10CA1a tumor cells. Phospho-phosphatidylinositol 3-kinase, phosphoinositide-dependent protein kinase 1 (PDK1), phospho-PDK1, phospho-eukaryotic translation initiation factor 4E (eIF4E) and phospho-eukaryotic initiation factor 4E binding protein 1 (4E-BP1) levels progressively increased in the cell lineage, with the greatest increase monitored in 10CA1a tumor cells. Phospho Ser 473 and Thr 408 Akt levels increased 10.2- and 136-fold in 10CA1a cells, respectively, relative to 10A cells. Phospho-p70S6 kinase (p70S6K) increased >2-fold in 10CA1a cells, relative to 10A cells. Immunohistochemistry confirmed Ras, phospho-Akt and phospho-p70S6K (Thr 421/ Ser 424) expression in lesions arising from premalignant and tumor cells. FOXO 1, phospho-FOXO 1 and phospho-FOXO 4 were significantly elevated in 10ATG3B premalignant and 10CA1a tumor cells. Phospho-FOXO 3a was progressively elevated, with the greatest levels detected in 10CA1a tumor cells. Immunohistochemistry revealed that phospho-FOXO 1, 3a and 4 staining was less in benign lesions, but elevated in advanced 10ATG3B and malignant 10CA1a lesions, showing a correspondence between the cells and lesions. Hence, phospho-Akt and phospho-FOXO 1, 3a and 4 merit consideration as biomarkers of tumorigenic risk from hyperplastic breast tissue.
Collapse
Affiliation(s)
- So Hee Kim
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA
- College of Dentistry and Institute of Oral Science, Kangnung National University, Kangnung, Republic of Korea
| | - Fred R. Miller
- The Environmental Health Science Center in Molecular and Cellular Toxicology, Wayne State University, Detroit, MI, USA
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Larry Tait
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Jie Zheng
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA
| | - Raymond F. Novak
- Institute of Environmental Health Sciences, Wayne State University, Detroit, MI, USA
- The Environmental Health Science Center in Molecular and Cellular Toxicology, Wayne State University, Detroit, MI, USA
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| |
Collapse
|
170
|
Rosenberger G, Meien S, Kutsche K. Oncogenic HRAS mutations cause prolonged PI3K signaling in response to epidermal growth factor in fibroblasts of patients with Costello syndrome. Hum Mutat 2009; 30:352-62. [PMID: 19035362 DOI: 10.1002/humu.20855] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Costello syndrome (CS) is a rare congenital disorder characterized by failure to thrive, craniofacial dysmorphisms, cardiac and skin abnormalities, mental retardation, and predisposition to malignancies. CS is caused by heterozygous gain-of-function mutations in HRAS that also occur as somatic alterations in human tumors. HRAS is one of the three classical RAS proteins and cycles between an active, GTP- and an inactive, GDP-bound conformation. We used primary human skin fibroblasts from patients with CS as a model system to study the functional consequences of HRAS mutations on endogenous signaling pathways. The GTP-bound form of HRAS was significantly enriched in CS compared with normal fibroblasts. Active HRAS is known to stimulate both the RAF-MEK-ERK and the PI3K-AKT signaling cascade. Phosphorylation of MEK and ERK was normal in CS fibroblasts under basal conditions and slightly prolonged after epidermal growth factor (EGF) stimulation. Interestingly, basal phosphorylation of AKT was increased yet more in CS fibroblasts. Moreover, AKT phosphorylation was diminished in the early and enhanced in the late phase of EGF stimulation. Taken together, these results document that CS-associated HRAS mutations result in prolonged signal flux in a ligand-dependent manner. Our data suggest that altered cellular response to growth factors rather than constitutive activation of HRAS downstream signaling molecules may contribute to some of the clinical features in patients with CS.
Collapse
Affiliation(s)
- Georg Rosenberger
- Institut für Humangenetik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | | | | |
Collapse
|
171
|
Sheng Z, Wang SZ, Green MR. Transcription and signalling pathways involved in BCR-ABL-mediated misregulation of 24p3 and 24p3R. EMBO J 2009; 28:866-76. [PMID: 19229297 PMCID: PMC2670863 DOI: 10.1038/emboj.2009.35] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Accepted: 01/23/2009] [Indexed: 11/08/2022] Open
Abstract
Lipocalin 24p3 is a secreted protein that can induce apoptosis in cells containing the 24p3 cell surface receptor, 24p3R. The oncoprotein BCR-ABL activates 24p3 and represses 24p3R expression. Thus, BCR-ABL(+) cells synthesise and secrete 24p3, which induces apoptosis in normal 24p3R-containing cells but not in BCR-ABL(+) cells. The cell signalling and transcription factor pathways by which BCR-ABL misregulates expression of 24p3 and 24p3R remain to be elucidated. Here we show that BCR-ABL upregulates 24p3 expression through activation of the JAK/STAT pathway, which culminates in binding of Stat5 to the 24p3 promoter. We find that 24p3R expression is regulated by Runx transcription factors, and that BCR-ABL induces a switch in binding from Runx3, an activator of 24p3R expression, to Runx1, a repressor of 24p3R expression, through a Ras signalling pathway. Finally, we show that repression of 24p3R by BCR-ABL is a critical feature of the mechanism by which imatinib kills BCR-ABL(+) cells. Our results reveal diverse signalling/transcription pathways that regulate 24p3 and 24p3R expression in response to BCR-ABL and are directly relevant to the treatment of BCR-ABL(+) disease.
Collapse
Affiliation(s)
- Zhi Sheng
- Howard Hughes Medical Institute and Programs in Gene Function and Expression and Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Shu-Zong Wang
- Howard Hughes Medical Institute and Programs in Gene Function and Expression and Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Michael R Green
- Howard Hughes Medical Institute and Programs in Gene Function and Expression and Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
172
|
Rotshenker S, Reichert F, Gitik M, Haklai R, Elad-Sfadia G, Kloog Y. Galectin-3/MAC-2, Ras and PI3K activate complement receptor-3 and scavenger receptor-AI/II mediated myelin phagocytosis in microglia. Glia 2009; 56:1607-13. [PMID: 18615637 DOI: 10.1002/glia.20713] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The removal of degenerated myelin is essential for repair in Wallerian degeneration that follows traumatic injury to axons and in autoimmune demyelinating diseases (e.g., multiple sclerosis). Microglia can remove degenerated myelin through phosphatidylinositol-3-kinase (PI3K)-dependent phagocytosis mediated by complement receptor-3 (CR3/MAC-1) and scavenger receptor-AI/II (SRAI/II). Paradoxically, these receptors are expressed in microglia after injury but myelin is not phagocytosed. Additionally, Galectin-3/MAC-2 is expressed in microglia that phagocytose but not in microglia that do not phagocytose, suggesting that Galectin-3/MAC-2 is instrumental in activating phagocytosis. S-trans, trans-farnesylthiosalicylic (FTS), which inhibits Galectin-3/MAC-2 dependent activation of PI3K through Ras, inhibited phagocytosis. K-Ras-GTP levels and PI3K activity increased during normal phagocytosis and decreased during FTS-inhibited phagocytosis. Galectin-3/MAC-2, which binds and stabilizes active Ras, coimmunoprecipitated with Ras and levels of the coimmunoprecipitate increased during normal phagocytosis. A role for Galectin-3/MAC-2 dependent activation of PI3K through Ras, mostly K-Ras, is thus suggested. An explanation may thus be offered for deficient phagocytosis by microglia that express CR3/MAC-1 and SRAI/II without Galectin-3/MAC-2 and efficient phagocytosis when CR3/MAC-1 and SRAI/II are co-expressed with Galectin-3/MAC-2.
Collapse
Affiliation(s)
- Shlomo Rotshenker
- Department of Anatomy and Cell Biology, Hebrew University Faculty of Medicine, and the Eric Roland Center for Neurodegenerative Diseases, Jerusalem, Israel.
| | | | | | | | | | | |
Collapse
|
173
|
Abstract
Differential subcellular compartmentalization of the three main Ras isoforms (H-Ras, N-Ras and K-Ras) is believed to underlie their biological differences. Modulatable interactions between cellular membranes and Ras C-terminal hypervariable region motifs determine differences in trafficking and the relative proportions of each isoform in cell-surface signalling nanoclusters and intracellular endoplasmic reticulum/Golgi, endosomal and mitochondrial compartments. Ras regulators, effectors and scaffolds are also differentially distributed, potentially enabling preferential coupling to specific signalling pathways in each subcellular location. Here we summarize the mechanisms underlying compartment-specific Ras signalling and the outputs generated.
Collapse
|
174
|
Functional differences between two classes of oncogenic mutation in the PIK3CA gene. Biochem Biophys Res Commun 2009; 381:577-81. [PMID: 19233141 DOI: 10.1016/j.bbrc.2009.02.081] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Accepted: 02/18/2009] [Indexed: 02/07/2023]
Abstract
PIK3CA codes for the p110alpha isoform of class-IA PI 3-kinase and oncogenic mutations in the helical domain and kinase domain are common in several cancers. We studied the biochemical properties of a common helical domain mutant (E545K) and a common kinase domain mutant (H1047R). Both retain the ability to autophosphorylate Ser608 of p85alpha and are also inhibited by a range of PI 3-kinase inhibitors (Wortmannin, LY294002, PI-103 and PIK-75) to a similar extent as WT p110alpha. Both mutants display an increased V(max) but while a PDGF derived diphosphotyrosylpeptide caused an increase in V(max) for WT p85alpha/p110alpha it did not for the E545K variant and actually decreased V(max) for the H1047R variant. Further, the E545K mutant was activated by H-Ras whereas the H1047R mutant was not. Together these results suggest helical domain mutants are in a state mimicking activation by growth factors whereas kinase domain mutants mimic the state activated by H-Ras.
Collapse
|
175
|
Di Nicolantonio F, Martini M, Molinari F, Sartore-Bianchi A, Arena S, Saletti P, De Dosso S, Mazzucchelli L, Frattini M, Siena S, Bardelli A. Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer. J Clin Oncol 2008; 26:5705-12. [PMID: 19001320 DOI: 10.1200/jco.2008.18.0786] [Citation(s) in RCA: 1239] [Impact Index Per Article: 72.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Cetuximab or panitumumab are effective in 10% to 20% unselected metastatic colorectal cancer (CRC) patients. KRAS mutations account for approximately 30% to 40% patients who are not responsive. The serine-threonine kinase BRAF is the principal effector of KRAS. We hypothesized that, in KRAS wild-type patients, BRAF mutations could have a predictive/prognostic value. PATIENTS AND METHODS We retrospectively analyzed objective tumor responses, time to progression, overall survival (OS), and the mutational status of KRAS and BRAF in 113 tumors from cetuximab- or panitumumab-treated metastatic CRC patients. The effect of the BRAF V600E mutation on cetuximab or panitumumab response was also assessed using cellular models of CRC. Results KRAS mutations were present in 30% of the patients and were associated with resistance to cetuximab or panitumumab (P = .011). The BRAF V600E mutation was detected in 11 of 79 patients who had wild-type KRAS. None of the BRAF-mutated patients responded to treatment, whereas none of the responders carried BRAF mutations (P = .029). BRAF-mutated patients had significantly shorter progression-free survival (P = .011) and OS (P < .0001) than wild-type patients. In CRC cells, the introduction of BRAF V600E allele impaired the therapeutic effect of cetuximab or panitumumab. Treatment with the BRAF inhibitor sorafenib restored sensitivity to panitumumab or cetuximab of CRC cells carrying the V600E allele. CONCLUSION BRAF wild-type is required for response to panitumumab or cetuximab and could be used to select patients who are eligible for the treatment. Double-hit therapies aimed at simultaneous inhibition of epidermal growth factor receptor and BRAF warrant exploration in CRC patients carrying the V600E oncogenic mutation.
Collapse
Affiliation(s)
- Federica Di Nicolantonio
- Laboratory of Molecular Genetics, The Oncogenomics Center, Institute for Cancer Research and Treatment, University of Torino, Medical School, Candiolo, Torino, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
176
|
Cho MC, Choi HS, Lee S, Kim BY, Jung M, Park SN, Yoon DY. Epiregulin expression by Ets-1 and ERK signaling pathway in Ki-ras-transformed cells. Biochem Biophys Res Commun 2008; 377:832-7. [PMID: 18948081 DOI: 10.1016/j.bbrc.2008.10.053] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2008] [Accepted: 10/14/2008] [Indexed: 11/18/2022]
Abstract
Epiregulin belongs to the epidermal growth factor family, binds to the epidermal growth factor receptor, and its expression is upregulated in various cancer cells, but the regulatory mechanism is unclear. We investigated the regulatory mechanism of epiregulin expression in Ki-ras-transformed cancer cells. In 267B1/Ki-ras cells, the RAF/MEK/ERK pathway was constitutively activated, epiregulin was up-regulated, and the expression and phosphorylation of Ets-1 were augmented. The inhibition of ERK by PD98059 decreased epiregulin and Ets-1 expression and suppressed the growth of 267B1/Ki-ras cells. A chromatin immunoprecipitation assay demonstrated that Ets-1 was bound to human epiregulin promoter, and this binding was abolished by PD98059. Silencing of Ets-1 by RNA interference decreased cellular epiregulin transcript expression. We suggest that the Ki-ras mutation in 267B1 prostate cells constitutively activates the RAF/MEK/ERK pathway and induces the activation of the Ets-1 transcription factor, ultimately leading to the increased expression of epiregulin.
Collapse
Affiliation(s)
- Min-Chul Cho
- Department of Bioscience and Biotechnology, Konkuk University, 1 Hwayng-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
177
|
Verveer PJ, Bastiaens PIH. Quantitative microscopy and systems biology: seeing the whole picture. Histochem Cell Biol 2008; 130:833-43. [DOI: 10.1007/s00418-008-0517-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2008] [Indexed: 12/22/2022]
|
178
|
Abstract
Understanding the signalling function of Ras GTPases has been the focus of much research for over 20 years. Both the catalytic domain and the membrane anchoring C terminal hypervariable region (HVR) of Ras are necessary for its cellular function. However, while the highly conserved catalytic domain has been characterized in atomic detail, the structure of the full-length membrane-bound Ras has remained elusive. Lack of structural knowledge on the full-length protein limited our understanding of Ras signalling. For example, structures of the Ras catalytic domain solved in complex with effectors do not provide a basis for the functional specificity of different Ras isoforms. Recent molecular dynamics simulations in combination with biophysical and cell biological experiments have shown that the HVR and parts of the G domain cofunction with the lipid tails to anchor H-ras to the plasma membrane. In the GTP-bound state, H-ras adopts an orientation that allows read out by Ras effectors and translation into corresponding MAPK signalling. Here we discuss details of an analysis that suggests a novel balance model for Ras functioning. The balance model rationalizes Ras membrane orientation and may help explain isoform specific interactions of Ras with its effectors and modulators.
Collapse
Affiliation(s)
- Daniel Abankwa
- Institute for Molecular Bioscience; University of Queensland; Brisbane, Australia
| | - Alemayehu A. Gorfe
- Department of Chemistry and Biochemistry and Center for Theoretical Biological Physics; University of California at San Diego; La Jolla, California USA
| | - John F. Hancock
- Institute for Molecular Bioscience; University of Queensland; Brisbane, Australia
| |
Collapse
|
179
|
Martin AP, Miller A, Emad L, Rahmani M, Walker T, Mitchell C, Hagan MP, Park MA, Yacoub A, Fisher PB, Grant S, Dent P. Lapatinib resistance in HCT116 cells is mediated by elevated MCL-1 expression and decreased BAK activation and not by ERBB receptor kinase mutation. Mol Pharmacol 2008; 74:807-22. [PMID: 18544666 PMCID: PMC2574656 DOI: 10.1124/mol.108.047365] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
We have defined some of the mechanisms by which the kinase inhibitor lapatinib kills HCT116 cells. Lapatinib inhibited radiation-induced activation of ERBB1/2, extracellular signal-regulated kinases 1/2, and AKT, and radiosensitized HCT116 cells. Prolonged incubation of HCT116 cells with lapatinib caused cell killing followed by outgrowth of lapatinib-adapted cells. Adapted cells were resistant to serum starvation-induced cell killing and were cross-resistant to multiple therapeutic drugs. Lapatinib was competent to inhibit basal and epidermal growth factor (EGF)-stimulated ERBB1 phosphorylation in adapted cells. Coexpression of dominant-negative ERBB1 and dominant-negative ERBB2 inhibited basal and EGF-stimulated ERBB1 and ERBB2 phosphorylation in parental and adapted cells. However, in neither parental nor adapted cells did expression of dominant-negative ERBB1 and dominant-negative ERBB2 recapitulate the cell death-promoting effects of lapatinib. Adapted cells had increased expression of MCL-1, decreased expression of BAX, and decreased activation of BAX and BAK. Overexpression of BCL-XL protected parental cells from lapatinib toxicity. Knockdown of MCL-1 expression enhanced lapatinib toxicity in adapted cells that was reverted by knockdown of BAK expression. Inhibition of caspase function modestly reduced lapatinib toxicity in parental cells, whereas knockdown of apoptosis-inducing factor expression suppressed lapatinib toxicity. Thus, in HCT116 cells, lapatinib adaptation can be mediated by altered expression of pro- and antiapoptotic proteins that maintain mitochondrial function.
Collapse
Affiliation(s)
- Aditi Pandya Martin
- Departments of Biochemistry (PD, AM, MAP, CM), Medicine (MR, SG), Pharmacology and Toxicology (APM, TW), Radiation Oncology (AY, MPH), Human Genetics (LE, PBF), Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Anna Miller
- Departments of Biochemistry (PD, AM, MAP, CM), Medicine (MR, SG), Pharmacology and Toxicology (APM, TW), Radiation Oncology (AY, MPH), Human Genetics (LE, PBF), Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Luni Emad
- Departments of Biochemistry (PD, AM, MAP, CM), Medicine (MR, SG), Pharmacology and Toxicology (APM, TW), Radiation Oncology (AY, MPH), Human Genetics (LE, PBF), Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Mohammed Rahmani
- Departments of Biochemistry (PD, AM, MAP, CM), Medicine (MR, SG), Pharmacology and Toxicology (APM, TW), Radiation Oncology (AY, MPH), Human Genetics (LE, PBF), Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Teneille Walker
- Departments of Biochemistry (PD, AM, MAP, CM), Medicine (MR, SG), Pharmacology and Toxicology (APM, TW), Radiation Oncology (AY, MPH), Human Genetics (LE, PBF), Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Clint Mitchell
- Departments of Biochemistry (PD, AM, MAP, CM), Medicine (MR, SG), Pharmacology and Toxicology (APM, TW), Radiation Oncology (AY, MPH), Human Genetics (LE, PBF), Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Michael P. Hagan
- Departments of Biochemistry (PD, AM, MAP, CM), Medicine (MR, SG), Pharmacology and Toxicology (APM, TW), Radiation Oncology (AY, MPH), Human Genetics (LE, PBF), Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Margaret A. Park
- Departments of Biochemistry (PD, AM, MAP, CM), Medicine (MR, SG), Pharmacology and Toxicology (APM, TW), Radiation Oncology (AY, MPH), Human Genetics (LE, PBF), Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Adly Yacoub
- Departments of Biochemistry (PD, AM, MAP, CM), Medicine (MR, SG), Pharmacology and Toxicology (APM, TW), Radiation Oncology (AY, MPH), Human Genetics (LE, PBF), Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Paul B. Fisher
- Departments of Biochemistry (PD, AM, MAP, CM), Medicine (MR, SG), Pharmacology and Toxicology (APM, TW), Radiation Oncology (AY, MPH), Human Genetics (LE, PBF), Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Steven Grant
- Departments of Biochemistry (PD, AM, MAP, CM), Medicine (MR, SG), Pharmacology and Toxicology (APM, TW), Radiation Oncology (AY, MPH), Human Genetics (LE, PBF), Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Paul Dent
- Departments of Biochemistry (PD, AM, MAP, CM), Medicine (MR, SG), Pharmacology and Toxicology (APM, TW), Radiation Oncology (AY, MPH), Human Genetics (LE, PBF), Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| |
Collapse
|
180
|
Kim S, Lee YZ, Kim YS, Bahk YY. A Proteomic approach for protein-profiling the oncogenic ras induced transformation (H-, K-, and N-Ras) in NIH/3T3 mouse embryonic fibroblasts. Proteomics 2008; 8:3082-93. [DOI: 10.1002/pmic.200800106] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
181
|
Kolokoltsova OA, Domina AM, Kolokoltsov AA, Davey RA, Weaver SC, Watowich SJ. Alphavirus production is inhibited in neurofibromin 1-deficient cells through activated RAS signalling. Virology 2008; 377:133-42. [PMID: 18485440 PMCID: PMC3280685 DOI: 10.1016/j.virol.2008.03.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2008] [Revised: 03/14/2008] [Accepted: 03/21/2008] [Indexed: 11/24/2022]
Abstract
Virus-host interactions essential for alphavirus pathogenesis are poorly understood. To address this shortcoming, we coupled retrovirus insertional mutagenesis and a cell survival selection strategy to generate clonal cell lines broadly resistant to Sindbis virus (SINV) and other alphaviruses. Resistant cells had significantly impaired SINV production relative to wild-type (WT) cells, although virus binding and fusion events were similar in both sets of cells. Analysis of the retroviral integration sites identified the neurofibromin 1 (NF1) gene as disrupted in alphavirus-resistant cell lines. Subsequent analysis indicated that expression of NF1 was significantly reduced in alphavirus-resistant cells. Importantly, independent down-regulation of NF1 expression in WT HEK 293 cells decreased virus production and increased cell viability during SINV infection, relative to infected WT cells. Additionally, we observed hyperactive RAS signalling in the resistant HEK 293 cells, which was anticipated because NF1 is a negative regulator of RAS. Expression of constitutively active RAS (HRAS-G12V) in a WT HEK 293 cell line resulted in a marked delay in virus production, compared with infected cells transfected with parental plasmid or dominant-negative RAS (HRAS-S17N). This work highlights novel host cell determinants required for alphavirus pathogenesis and suggests that RAS signalling may play an important role in neuronal susceptibility to SINV infection.
Collapse
Affiliation(s)
- Olga A Kolokoltsova
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA.
| | | | | | | | | | | |
Collapse
|
182
|
Vögler O, Barceló JM, Ribas C, Escribá PV. Membrane interactions of G proteins and other related proteins. BIOCHIMICA ET BIOPHYSICA ACTA 2008; 1778:1640-52. [PMID: 18402765 DOI: 10.1016/j.bbamem.2008.03.008] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2007] [Revised: 03/01/2008] [Accepted: 03/12/2008] [Indexed: 01/25/2023]
Abstract
Guanine nucleotide-binding proteins, G proteins, propagate incoming messages from receptors to effector proteins. They switch from an inactive to active state by exchanging a GDP molecule for GTP, and they return to the inactive form by hydrolyzing GTP to GDP. Small monomeric G proteins, such as Ras, are involved in controlling cell proliferation, differentiation and apoptosis, and they interact with membranes through isoprenyl moieties, fatty acyl moieties, and electrostatic interactions. This protein-lipid binding facilitates productive encounters of Ras and Raf proteins in defined membrane regions, so that signals can subsequently proceed through MEK and ERK kinases, which constitute the canonical MAP kinase signaling cassette. On the other hand, heterotrimeric G proteins undergo co/post-translational modifications in the alpha (myristic and/or palmitic acid) and the gamma (farnesol or geranylgeraniol) subunits. These modifications not only assist the G protein to localize to the membrane but they also help distribute the heterotrimer (Galphabetagamma) and the subunits generated upon activation (Galpha and Gbetagamma) to appropriate membrane microdomains. These proteins transduce messages from ubiquitous serpentine receptors, which control important functions such as taste, vision, blood pressure, body weight, cell proliferation, mood, etc. Moreover, the exchange of GDP by GTP is triggered by nucleotide exchange factors. Membrane receptors that activate G proteins can be considered as such, but other cytosolic, membranal or amphitropic proteins can accelerate the rate of G protein exchange or even activate this process in the absence of receptor-mediated activation. These and other protein-protein interactions of G proteins with other signaling proteins are regulated by their lipid preferences. Thus, G protein-lipid interactions control the features of messages and cell physiology.
Collapse
Affiliation(s)
- Oliver Vögler
- Molecular Cell Biomedicine, Department of Biology-IUNICS, Universitat de les Illes Balears, Palma de Mallorca, Spain
| | | | | | | |
Collapse
|
183
|
Plowman SJ, Ariotti N, Goodall A, Parton RG, Hancock JF. Electrostatic interactions positively regulate K-Ras nanocluster formation and function. Mol Cell Biol 2008; 28:4377-85. [PMID: 18458061 PMCID: PMC2447143 DOI: 10.1128/mcb.00050-08] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 02/18/2008] [Accepted: 04/18/2008] [Indexed: 11/20/2022] Open
Abstract
The organization of Ras proteins into plasma membrane nanoclusters is essential for high-fidelity signal transmission, but whether the nanoscale environments of different Ras nanoclusters regulate effector interactions is unknown. We show using high-resolution spatial mapping that Raf-1 is recruited to and retained in K-Ras-GTP nanoclusters. In contrast, Raf-1 recruited to the plasma membrane by H-Ras is not retained in H-Ras-GTP nanoclusters. Similarly, upon epidermal growth factor receptor activation, Raf-1 is preferentially recruited to K-Ras-GTP and not H-Ras-GTP nanoclusters. The formation of K-Ras-GTP nanoclusters is inhibited by phosphorylation of S181 in the C-terminal polybasic domain or enhanced by blocking S181 phosphorylation, with a concomitant reduction or increase in Raf-1 plasma membrane recruitment, respectively. Phosphorylation of S181 does not, however, regulate in vivo interactions with the nanocluster scaffold galectin-3 (Gal3), indicating separate roles for the polybasic domain and Gal3 in driving K-Ras nanocluster formation. Together, these data illustrate that Ras nanocluster composition regulates effector recruitment and highlight the importance of lipid/protein nanoscale environments to the activation of signaling cascades.
Collapse
Affiliation(s)
- Sarah J Plowman
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, Brisbane 4072, Australia
| | | | | | | | | |
Collapse
|
184
|
Rosseland CM, Wierød L, Flinder LI, Oksvold MP, Skarpen E, Huitfeldt HS. Distinct functions of H-Ras and K-Ras in proliferation and survival of primary hepatocytes due to selective activation of ERK and PI3K. J Cell Physiol 2008; 215:818-26. [PMID: 18163378 DOI: 10.1002/jcp.21367] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Ras proteins mediate signals both via extracellular signal-regulated kinase 1 and 2 (ERK), and phosphoinositide 3-kinase (PI3K). These signals are key events in cell protection and compensatory cell growth after exposure to cell damaging and pro-apoptotic stimuli, thus maintaining homeostasis. By transfection techniques, we found that both H-Ras and K-Ras were expressed and appeared functionally active in primary hepatocytes. We compared the ability of H-Ras and K-Ras homologues to preferentially activate one of the two pathways, thereby differentially controlling cell survival and growth. We found that ectopic expression of dominant negative (DN) H-RasN17, but not DN K-RasN17, efficiently inhibited both phosphorylation and translocation of ERK to the nuclear compartment, which are prerequisites for cell cycle progression. Furthermore, ectopic expression of constitutive active (CA) H-RasV12, but not CA K-RasV12, potentiated EGF-induced proliferation. We also found that expression of CA mutants of either H-Ras or K-Ras protected hepatocytes from transforming growth factor-beta1 (TGF-beta1)-induced apoptosis. However, H-Ras-induced survival was mediated by ERK/RSK as well as by PI3K, whereas K-Ras-induced survival was mediated by PI3K only. In conclusion, H-Ras and K-Ras had differential functions in proliferation and survival of primary hepatocytes. H-Ras was the major mediator of ERK-induced proliferation and survival, whereas H-Ras and K-Ras both mediated PI3K-induced survival.
Collapse
Affiliation(s)
- Carola M Rosseland
- Laboratory for Toxicopathology, Institute of Pathology, Rikshospitalet-Radiumhospitalet Medical Centre, University of Oslo, Norway.
| | | | | | | | | | | |
Collapse
|
185
|
Omerovic J, Hammond DE, Clague MJ, Prior IA. Ras isoform abundance and signalling in human cancer cell lines. Oncogene 2008; 27:2754-62. [PMID: 17998936 PMCID: PMC2557550 DOI: 10.1038/sj.onc.1210925] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2007] [Revised: 10/16/2007] [Accepted: 10/17/2007] [Indexed: 11/09/2022]
Abstract
The ubiquitously expressed major Ras isoforms: H-, K- and N-Ras, are highly conserved, yet exhibit different biological outputs. We have compared the relative efficiencies with which epidermal or hepatocyte growth factor activates Ras isoforms and the requirement for specific isoforms in the activation of downstream pathways. We find that the relative coupling efficiencies to each Ras isoform are conserved between stimuli. Furthermore, in both cases, inhibition of receptor endocytosis led to reduced N- and H-Ras activation, but K-Ras was unaffected. Acute knockdown of each isoform with siRNA allows endogenous Ras isoform function and abundance to be probed. This revealed that there is significant variation in the contribution of individual isoforms to total Ras across a panel of cancer cell lines although typically K> or =N>>H. Intriguingly, cancer cell lines where a significant fraction of endogenous Ras is oncogenically mutated showed attenuated activation of canonical Ras effector pathways. We profiled the contribution of each Ras isoform to the total Ras pool allowing interpretation of the effect of isoform-specific knockdown on signalling outcomes. In contrast to previous studies indicating preferential coupling of isoforms to Raf and PtdIns-3-kinase pathways, we find that endogenous Ras isoforms show no specific coupling to these major Ras pathways.
Collapse
Affiliation(s)
| | | | - Michael J. Clague
- The Physiological Laboratory, University of Liverpool, Crown St., Liverpool, L69 3BX, UK
| | - Ian A. Prior
- The Physiological Laboratory, University of Liverpool, Crown St., Liverpool, L69 3BX, UK
| |
Collapse
|
186
|
Lopez-Alcalá C, Alvarez-Moya B, Villalonga P, Calvo M, Bachs O, Agell N. Identification of Essential Interacting Elements in K-Ras/Calmodulin Binding and Its Role in K-Ras Localization. J Biol Chem 2008; 283:10621-31. [DOI: 10.1074/jbc.m706238200] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
187
|
Activated Kras, but not Hras or Nras, may initiate tumors of endodermal origin via stem cell expansion. Mol Cell Biol 2008; 28:2659-74. [PMID: 18268007 DOI: 10.1128/mcb.01661-07] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The three closely related human Ras genes, Hras, Nras, and Kras, are all widely expressed, engage a common set of downstream effectors, and can each exhibit oncogenic activity. However, the vast majority of activating Ras mutations in human tumors involve Kras. Moreover, Kras mutations are most frequently seen in tumors of endodermally derived tissues (lung, pancreas, and colon), suggesting that activated Kras may affect an endodermal progenitor to initiate oncogenesis. Using a culture model of retinoic acid (RA)-induced stem cell differentiation to endoderm, we determined that while activated HrasV12 promotes differentiation and growth arrest in these endodermal progenitors, KrasV12 promotes their proliferation. Furthermore, KrasV12-expressing endodermal progenitors fail to differentiate upon RA treatment and continue to proliferate and maintain stem cell characteristics. NrasV12 neither promotes nor prevents differentiation. A structure-function analysis demonstrated that these distinct effects of the Ras isoforms involve their variable C-terminal domains, implicating compartmentalized signaling, and revealed a requirement for several established Ras effectors. These findings indicate that activated Ras isoforms exert profoundly different effects on endodermal progenitors and that mutant Kras may initiate tumorigenesis by expanding a susceptible stem/progenitor cell population. These results potentially explain the high frequency of Kras mutations in tumors of endodermal origin.
Collapse
|
188
|
Moretó J, Lladó A, Vidal-Quadras M, Calvo M, Pol A, Enrich C, Tebar F. Calmodulin modulates H-Ras mediated Raf-1 activation. Cell Signal 2008; 20:1092-103. [PMID: 18356021 DOI: 10.1016/j.cellsig.2008.01.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2007] [Revised: 01/22/2008] [Accepted: 01/23/2008] [Indexed: 01/08/2023]
Abstract
We have previously demonstrated that, in COS-1 cells, inhibition of calmodulin increases Ras-GTP levels although it decreases Raf-1 activity and consequently MAPK. The present study analyzes the role of calmodulin in the regulation of Raf-1. First we show, using FRET microscopy, that inhibition of Raf-1 was not a consequence of a decreased interaction between H-Ras and Raf-1. Besides, the analysis of the phosphorylation state of Raf-1 showed that calmodulin, through downstream PI3K, is essential to ensure the Ser338-Raf-1 phosphorylation, critical for Raf-1 activation. We also show that the expression of a dominant negative mutant of PI3K impairs the calmodulin-mediated Raf-1 activation; in addition, both calmodulin and PI3K inhibitors decrease phospho-Ser338 and Raf-1 activity from upstream active H-Ras (H-RasG12V) and this effect is dependent on endocytosis. Importantly, in H-Ras depleted COS-1 cells, calmodulin does not modulate MAPK activation. Altogether, the results suggest that calmodulin regulation of MAPK in COS-1 cells relies upon H-Ras control of Raf-1 activity and involves PI3K.
Collapse
Affiliation(s)
- Jemina Moretó
- Departament de Biologia Cel.lular, Facultat de Medicina, Universitat de Barcelona, Casanova 143, 08036-Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
189
|
Laude AJ, Prior IA. Palmitoylation and localisation of RAS isoforms are modulated by the hypervariable linker domain. J Cell Sci 2008; 121:421-7. [PMID: 18211960 DOI: 10.1242/jcs.020107] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
RAS isoforms have been proposed to exhibit differing biological outputs due to differences in their relative occupancy of cellular organelles and signalling microdomains. The membrane binding and targeting motifs of RAS are encoded by the C-terminal hypervariable region (HVR), and the precise localisation depends upon interactions between the HVR and the host membrane. Classic studies revealed that all RAS proteins rely on farnesylation and either palmitoylation or a polybasic stretch for stable binding to membranes. We now show that, for N-RAS and Ki-RAS4A, mono-palmitoylation and farnesylation are not sufficient for specifying stable cell-surface localisation. A third motif that is present within the linker domain of all palmitoylated RAS HVRs is necessary for stabilising localisation to the plasma membrane. This motif comprises acidic residues that stabilise palmitoylation and basic amino acids that are likely to interact electrostatically with acidic phospholipids enriched at the cell surface. Importantly, altered localisation is achieved without changes in palmitoylation status. Our data provide a mechanism for distinct HVR membrane interactions controlling subcellular distribution. In the context of the full-length RAS proteins, this is likely to be of crucial importance for controlling signalling output and engagement with different pools of effectors.
Collapse
Affiliation(s)
- Alex J Laude
- Physiological Laboratory, University of Liverpool, Liverpool, UK
| | | |
Collapse
|
190
|
Scuto A, Zhang H, Zhao H, Rivera M, Yeatman TJ, Jove R, Torres-Roca JF. RbAp48 Regulates Cytoskeletal Organization and Morphology by Increasing K-Ras Activity and Signaling through Mitogen-Activated Protein Kinase. Cancer Res 2007; 67:10317-24. [DOI: 10.1158/0008-5472.can-06-3313] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
191
|
Abstract
Ras GTPases mediate a wide variety of cellular processes by converting a multitude of extracellular stimuli into specific biological responses including proliferation, differentiation and survival. In mammalian cells, three ras genes encode four Ras isoforms (H-Ras, K-Ras4A, K-Ras4B and N-Ras) that are highly homologous but functionally distinct. Differences between the isoforms, including their post-translational modifications and intracellular sorting, mean that Ras has emerged as an important model system of compartmentalised signalling and membrane biology. Ras isoforms in different subcellular locations are proposed to recruit distinct upstream and downstream accessory proteins and activate multiple signalling pathways. Here, we summarise data relating to isoform-specific signalling, its role in disease and the mechanisms promoting compartmentalised signalling. Further understanding of this field will reveal the role of Ras signalling in development, cellular homeostasis and cancer and may suggest new therapeutic approaches.
Collapse
Affiliation(s)
- J. Omerovic
- Physiological Laboratory, University of Liverpool, Crown St., Liverpool, L69 3BX UK
| | - A. J. Laude
- Physiological Laboratory, University of Liverpool, Crown St., Liverpool, L69 3BX UK
| | - I. A. Prior
- Physiological Laboratory, University of Liverpool, Crown St., Liverpool, L69 3BX UK
| |
Collapse
|
192
|
Abankwa D, Gorfe AA, Hancock JF. Ras nanoclusters: molecular structure and assembly. Semin Cell Dev Biol 2007; 18:599-607. [PMID: 17897845 PMCID: PMC2761225 DOI: 10.1016/j.semcdb.2007.08.003] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2007] [Revised: 08/10/2007] [Accepted: 08/16/2007] [Indexed: 01/02/2023]
Abstract
H-, N- and K-ras4B are lipid-anchored, peripheral membrane guanine nucleotide binding proteins. Recent work has shown that Ras proteins are laterally segregated into non-overlapping, dynamic domains of the plasma membrane called nanoclusters. This lateral segregation is important to specify Ras interactions with membrane-associated proteins, effectors and scaffolding proteins and is critical for Ras signal transduction. Here we review biological, in vitro and structural data that provide insight into the molecular basis of how palmitoylated Ras proteins are anchored to the plasma membrane. We explore possible mechanisms for how the interactions of H-ras with a lipid bilayer may drive nanocluster formation.
Collapse
Affiliation(s)
- Daniel Abankwa
- Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, Australia
| | | | | |
Collapse
|
193
|
Smith SC, Oxford G, Baras AS, Owens C, Havaleshko D, Brautigan DL, Safo MK, Theodorescu D. Expression of ral GTPases, their effectors, and activators in human bladder cancer. Clin Cancer Res 2007; 13:3803-13. [PMID: 17606711 DOI: 10.1158/1078-0432.ccr-06-2419] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The Ral family of small G proteins has been implicated in tumorigenesis, invasion, and metastasis in in vitro and animal model systems; however, a systematic evaluation of the state of activation, mutation, or expression of these GTPases has not been reported in any tumor type. EXPERIMENTAL DESIGN We determined the activation state of the RalA and RalB paralogs in 10 bladder cancer cell lines with varying Ras mutation status. We sequenced RalA and RalB cDNAs from 20 bladder cancer cell lines and functionally evaluated the mutations found. We determined the expression of Ral, Ral activators, and Ral effectors on the level of mRNA or protein in human bladder cancer cell lines and tissues. RESULTS We uncovered one E97Q substitution mutation of RalA in 1 of 20 cell lines tested and higher Ral activation in cells harboring mutant HRAS. We found overexpression of mRNAs for RalA and Aurora-A, a mitotic kinase that activates RalA, in bladder cancer (both P < 0.001), and in association with tumors of higher stage and grade. RalBP1, a canonical Ral effector, mRNA and protein was overexpressed in bladder cancer (P < 0.001), whereas Filamin A was underexpressed (P = 0.004). We determined that RalA mRNA levels correlated significantly with protein levels (P < 0.001) and found protein overexpression of both GTPases in homogenized invasive cancers. Available data sets suggest that RalA mRNA is also overexpressed in seminoma, glioblastoma, and carcinomas of the liver, pancreas, and prostate. CONCLUSION These findings of activation and differential expression of RalA and RalB anchor prior work in model systems to human disease and suggest therapeutic strategies targeting both GTPases in this pathway may be beneficial.
Collapse
Affiliation(s)
- Steven Christopher Smith
- Department of Molecular Physiology, University of Virginia, Charlottesville, Virginia 22908, USA
| | | | | | | | | | | | | | | |
Collapse
|
194
|
Eisenberg S, Henis YI. Interactions of Ras proteins with the plasma membrane and their roles in signaling. Cell Signal 2007; 20:31-9. [PMID: 17888630 DOI: 10.1016/j.cellsig.2007.07.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2007] [Accepted: 07/18/2007] [Indexed: 12/21/2022]
Abstract
The complex dynamic structure of the plasma membrane plays critical roles in cellular signaling; interactions with the membrane lipid milieu, spatial segregation within and between cellular membranes and/or targeting to specific membrane-associated scaffolds are intimately involved in many signal transduction pathways. In this review, we focus on the membrane interactions of Ras proteins. These small GTPases play central roles in the regulation of cell growth and proliferation, and their excessive activation is commonly encountered in human tumors. Ras proteins associate with the membrane continuously via C-terminal lipidation and additional interactions in both their inactive and active forms; this association, as well as the targeting of specific Ras isoforms to plasma membrane microdomains and to intracellular organelles, have recently been implicated in Ras signaling and oncogenic potential. We discuss biochemical and biophysical evidence for the roles of specific domains of Ras proteins in mediating their association with the plasma membrane, and consider the potential effects of lateral segregation and interactions with membrane-associated protein assemblies on the signaling outcomes.
Collapse
Affiliation(s)
- Sharon Eisenberg
- Department of Neurobiochemistry, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | | |
Collapse
|
195
|
Abankwa D, Vogel H. A FRET map of membrane anchors suggests distinct microdomains of heterotrimeric G proteins. J Cell Sci 2007; 120:2953-62. [PMID: 17690305 DOI: 10.1242/jcs.001404] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The standard model of heterotrimeric G protein signaling postulates a dissociation of Gα and Gβγ subunits after activation. We hypothesized that the different combination of lipid-modifications on Gα and Gαβγ subunits directs them into different microdomains. By characterizing rapidly and at high sensitivity 38 fluorescence resonance energy transfer (FRET) pairs of heterotrimeric-G-protein constructs, we defined their microdomains in relation to each other, free from the constraints of the raft/non-raft dualism. We estimated that in a cell ∼30% of these membrane-anchored proteins are mostly clustered in 3400-16,200 copies of 30-nm microdomains. We found that the membrane anchors of Gα and Gαβγ subunits of both the Gi/o and Gq family co-cluster differently with microdomain markers. Moreover, anchors of the Gαi/o and Gαq subunits co-clustered only weakly, whereas constructs that contained the anchors of the corresponding heterotrimers co-clustered considerably, suggesting the existence of at least three types of microdomain. Finally, FRET experiments with full-length heterotrimeric G proteins confirmed that the inactive, heterotrimerized Gα subunit is in microdomains shared by heterotrimers from different subclasses, from where it displaces upon activation into a membrane-anchor- and subclass-specific microdomain.
Collapse
Affiliation(s)
- Daniel Abankwa
- Ecole Polytechnique Fédérale de Lausanne (EPFL), Institut des Sciences et Ingénierie Chimiques, CH-1015 Lausanne, Switzerland.
| | | |
Collapse
|
196
|
McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Wong EWT, Chang F, Lehmann B, Terrian DM, Milella M, Tafuri A, Stivala F, Libra M, Basecke J, Evangelisti C, Martelli AM, Franklin RA. Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance. BIOCHIMICA ET BIOPHYSICA ACTA 2007; 1773:1263-84. [PMID: 17126425 PMCID: PMC2696318 DOI: 10.1016/j.bbamcr.2006.10.001] [Citation(s) in RCA: 1752] [Impact Index Per Article: 97.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/05/2006] [Revised: 10/02/2006] [Accepted: 10/03/2006] [Indexed: 02/07/2023]
Abstract
Growth factors and mitogens use the Ras/Raf/MEK/ERK signaling cascade to transmit signals from their receptors to regulate gene expression and prevent apoptosis. Some components of these pathways are mutated or aberrantly expressed in human cancer (e.g., Ras, B-Raf). Mutations also occur at genes encoding upstream receptors (e.g., EGFR and Flt-3) and chimeric chromosomal translocations (e.g., BCR-ABL) which transmit their signals through these cascades. Even in the absence of obvious genetic mutations, this pathway has been reported to be activated in over 50% of acute myelogenous leukemia and acute lymphocytic leukemia and is also frequently activated in other cancer types (e.g., breast and prostate cancers). Importantly, this increased expression is associated with a poor prognosis. The Ras/Raf/MEK/ERK and Ras/PI3K/PTEN/Akt pathways interact with each other to regulate growth and in some cases tumorigenesis. For example, in some cells, PTEN mutation may contribute to suppression of the Raf/MEK/ERK cascade due to the ability of activated Akt to phosphorylate and inactivate different Rafs. Although both of these pathways are commonly thought to have anti-apoptotic and drug resistance effects on cells, they display different cell lineage specific effects. For example, Raf/MEK/ERK is usually associated with proliferation and drug resistance of hematopoietic cells, while activation of the Raf/MEK/ERK cascade is suppressed in some prostate cancer cell lines which have mutations at PTEN and express high levels of activated Akt. Furthermore the Ras/Raf/MEK/ERK and Ras/PI3K/PTEN/Akt pathways also interact with the p53 pathway. Some of these interactions can result in controlling the activity and subcellular localization of Bim, Bak, Bax, Puma and Noxa. Raf/MEK/ERK may promote cell cycle arrest in prostate cells and this may be regulated by p53 as restoration of wild-type p53 in p53 deficient prostate cancer cells results in their enhanced sensitivity to chemotherapeutic drugs and increased expression of Raf/MEK/ERK pathway. Thus in advanced prostate cancer, it may be advantageous to induce Raf/MEK/ERK expression to promote cell cycle arrest, while in hematopoietic cancers it may be beneficial to inhibit Raf/MEK/ERK induced proliferation and drug resistance. Thus the Raf/MEK/ERK pathway has different effects on growth, prevention of apoptosis, cell cycle arrest and induction of drug resistance in cells of various lineages which may be due to the presence of functional p53 and PTEN and the expression of lineage specific factors.
Collapse
Affiliation(s)
- James A McCubrey
- Department of Microbiology and Immunology, Leo Jenkins Cancer Center, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
197
|
Abstract
RAS proteins are small GTPases that play a central role in transducing signals that regulate cell proliferation, survival, and differentiation. The RAS proteins interact with a common set of activators and effectors; however, they associate with different microdomains of the plasma membrane as well as other endomembranes and are capable of generating distinct signal outputs. Mutations that result in constitutive activation of RAS proteins are associated with approximately 30% of all human cancers; however, different RAS oncogenes are preferentially associated with different types of human cancer. In myeloid malignancies, NRAS mutations are more frequent than KRAS mutations, whereas HRAS mutations are rare. The mechanism underlying the different frequencies of RAS isoforms mutated in myeloid leukemia is not known. In this study, we compared the leukemogenic potential of activated NRAS, KRAS, and HRAS in the same bone marrow transduction/transplantation model system. We found that all three RAS oncogenes have the ability to induce myeloid leukemias, yet have distinct leukemogenic strengths and phenotypes. The models established here provide a system for further studying the molecular mechanisms in the pathogenesis of myeloid malignancies and for testing targeted therapies.
Collapse
Affiliation(s)
- Chaitali Parikh
- Rosenstiel Basic Medical Sciences Research Center, Department of Biology, Brandeis University, Waltham, Massachusetts
| | - Ramesh Subrahmanyam
- Rosenstiel Basic Medical Sciences Research Center, Department of Biology, Brandeis University, Waltham, Massachusetts
- Graduate Program in Biophysics and Structural Biology, Department of Biochemistry, Brandeis University, Waltham, Massachusetts
| | - Ruibao Ren
- Rosenstiel Basic Medical Sciences Research Center, Department of Biology, Brandeis University, Waltham, Massachusetts
| |
Collapse
|
198
|
Tian T, Harding A, Inder K, Plowman S, Parton RG, Hancock JF. Plasma membrane nanoswitches generate high-fidelity Ras signal transduction. Nat Cell Biol 2007; 9:905-14. [PMID: 17618274 DOI: 10.1038/ncb1615] [Citation(s) in RCA: 320] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Accepted: 05/22/2007] [Indexed: 11/09/2022]
Abstract
Ras proteins occupy dynamic plasma membrane nanodomains called nanoclusters. The significance of this spatial organization is unknown. Here we show, using in silico and in vivo analyses of mitogen-activated protein (MAP) kinase signalling, that Ras nanoclusters operate as sensitive switches, converting graded ligand inputs into fixed outputs of activated extracellular signal-regulated kinase (ERK). By generating Ras nanoclusters in direct proportion to ligand input, cells build an analogue-digital-analogue circuit relay that transmits a signal across the plasma membrane with high fidelity. Signal transmission is completely dependent on Ras spatial organization and fails if nanoclustering is abrogated. A requirement for high-fidelity signalling may explain the non-random distribution of other plasma membrane signalling complexes.
Collapse
Affiliation(s)
- Tianhai Tian
- Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, Australia
| | | | | | | | | | | |
Collapse
|
199
|
Zhu F, Zykova TA, Kang BS, Wang Z, Ebeling MC, Abe Y, Ma WY, Bode AM, Dong Z. Bidirectional signals transduced by TOPK-ERK interaction increase tumorigenesis of HCT116 colorectal cancer cells. Gastroenterology 2007; 133:219-31. [PMID: 17631144 DOI: 10.1053/j.gastro.2007.04.048] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2006] [Accepted: 04/12/2007] [Indexed: 12/31/2022]
Abstract
BACKGROUND & AIMS Aberrant activation of Ras and Raf in mitogen-activated protein kinase (MAPK) signaling has been linked with cancer. However, the role of MAPK kinases (MAPKKs or MEKs) in cancer is unclear, although constitutively activated MEK1, which does not exist in nature, is "oncogenic." Herein, we found that T-cell-originated protein kinase (TOPK), a member of the MAPKK protein family, is highly expressed in human colorectal cancer tissues and cell lines and plays an important role in the transformation of colorectal cancer. METHODS The biologic consequences of overexpression or knockdown of TOPK in JB6 Cl41 and HCT116 colorectal cancer cells were studied in vitro and in vivo, respectively. Kinase assay or transient transfection experiments were performed to study the bidirectional signaling pathway between TOPK and extracellular signal-regulated kinase (ERK). RESULTS TOPK was shown to promote transformation in vitro and in vivo, and knockdown of TOPK in HCT116 colorectal cancer cells reduced this cell lines' tumorigenic properties in vitro and in vivo. Furthermore, a positive feedback loop between TOPK and ERK2 was identified. With epidermal growth factor treatment, knockdown of either TOPK or ERK2 in HCT116 cells resulted in a decreased phosphorylation of ERK2 or TOPK, respectively, and knockdown of TOPK in HCT116 colorectal cancer cells blocked the phosphorylation of downstream substrates of ERK2. CONCLUSIONS The positive feedback loop between TOPK and ERK2 increases tumorigenesis properties of HCT116 colorectal cancer cells, and TOPK-regulated signaling may serve as a potential therapeutic target in colorectal cancer.
Collapse
Affiliation(s)
- Feng Zhu
- Hormel Institute, University of Minnesota, Austin, Minnesota 55912, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
200
|
Krzysiak AJ, Rawat DS, Scott SA, Pais JE, Handley M, Harrison ML, Fierke CA, Gibbs RA. Combinatorial modulation of protein prenylation. ACS Chem Biol 2007; 2:385-9. [PMID: 17530735 PMCID: PMC2922964 DOI: 10.1021/cb700062b] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The cell has >60 different farnesylated proteins. Many critically important signal transduction proteins are post-translationally modified with attachment of a farnesyl isoprenoid catalyzed by protein farnesyltransferase (FTase). Recently, it has been shown that farnesyl diphosphate (FPP) analogues can alter the peptide substrate specificity of FTase. We have used combinatorial screening of FPP analogues and peptide substrates to identify patterns in FTase substrate selectivity. Each FPP analogue displays a unique pattern of substrate reactivity with the tested peptides; FTase efficiently catalyzes the transfer of an FPP analogue selectively to one peptide and not another. Furthermore, we have demonstrated that these analogues can enter cells and be incorporated into proteins. These FPP analogues could serve as selective tools to examine the role prenylation plays in individual protein function.
Collapse
Affiliation(s)
- Amanda J. Krzysiak
- Department of Medicinal Chemistry and Molecular Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Purdue University, West Lafayette, Indiana 47907
| | | | - Sarah A. Scott
- Department of Medicinal Chemistry and Molecular Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Purdue University, West Lafayette, Indiana 47907
| | - June E. Pais
- Departments of Chemistry and Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Misty Handley
- Department of Medicinal Chemistry and Molecular Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Purdue University, West Lafayette, Indiana 47907
| | - Marietta L. Harrison
- Department of Medicinal Chemistry and Molecular Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Purdue University, West Lafayette, Indiana 47907
| | - Carol A. Fierke
- Departments of Chemistry and Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Richard A. Gibbs
- Department of Medicinal Chemistry and Molecular Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Purdue University, West Lafayette, Indiana 47907
| |
Collapse
|