1
|
Kolch W, Berta D, Rosta E. Dynamic regulation of RAS and RAS signaling. Biochem J 2023; 480:1-23. [PMID: 36607281 PMCID: PMC9988006 DOI: 10.1042/bcj20220234] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/16/2022] [Accepted: 12/23/2022] [Indexed: 01/07/2023]
Abstract
RAS proteins regulate most aspects of cellular physiology. They are mutated in 30% of human cancers and 4% of developmental disorders termed Rasopathies. They cycle between active GTP-bound and inactive GDP-bound states. When active, they can interact with a wide range of effectors that control fundamental biochemical and biological processes. Emerging evidence suggests that RAS proteins are not simple on/off switches but sophisticated information processing devices that compute cell fate decisions by integrating external and internal cues. A critical component of this compute function is the dynamic regulation of RAS activation and downstream signaling that allows RAS to produce a rich and nuanced spectrum of biological outputs. We discuss recent findings how the dynamics of RAS and its downstream signaling is regulated. Starting from the structural and biochemical properties of wild-type and mutant RAS proteins and their activation cycle, we examine higher molecular assemblies, effector interactions and downstream signaling outputs, all under the aspect of dynamic regulation. We also consider how computational and mathematical modeling approaches contribute to analyze and understand the pleiotropic functions of RAS in health and disease.
Collapse
Affiliation(s)
- Walter Kolch
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Dénes Berta
- Department of Physics and Astronomy, University College London, Gower Street, London WC1E 6BT, U.K
| | - Edina Rosta
- Department of Physics and Astronomy, University College London, Gower Street, London WC1E 6BT, U.K
| |
Collapse
|
2
|
Abuasaker B, Garrido E, Vilaplana M, Gómez-Zepeda JD, Brun S, Garcia-Cajide M, Mauvezin C, Jaumot M, Pujol MD, Rubio-Martínez J, Agell N. α4-α5 Helices on Surface of KRAS Can Accommodate Small Compounds That Increase KRAS Signaling While Inducing CRC Cell Death. Int J Mol Sci 2023; 24:ijms24010748. [PMID: 36614192 PMCID: PMC9821572 DOI: 10.3390/ijms24010748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 01/03/2023] Open
Abstract
KRAS is the most frequently mutated oncogene associated with the genesis and progress of pancreatic, lung and colorectal (CRC) tumors. KRAS has always been considered as a therapeutic target in cancer but until now only two compounds that inhibit one specific KRAS mutation have been approved for clinical use. In this work, by molecular dynamics and a docking process, we describe a new compound (P14B) that stably binds to a druggable pocket near the α4-α5 helices of the allosteric domain of KRAS. This region had previously been identified as the binding site for calmodulin (CaM). Using surface plasmon resonance and pulldown analyses, we prove that P14B binds directly to oncogenic KRAS thus competing with CaM. Interestingly, P14B favors oncogenic KRAS interaction with BRAF and phosphorylated C-RAF, and increases downstream Ras signaling in CRC cells expressing oncogenic KRAS. The viability of these cells, but not that of the normal cells, is impaired by P14B treatment. These data support the significance of the α4-α5 helices region of KRAS in the regulation of oncogenic KRAS signaling, and demonstrate that drugs interacting with this site may destine CRC cells to death by increasing oncogenic KRAS downstream signaling.
Collapse
Affiliation(s)
- Baraa Abuasaker
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Eduardo Garrido
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
- Departament de Ciència de Materials i Química Física, Facultat de Química, Universitat de Barcelona & Institut de Recerca en Química Teòrica i Computacional (IQTCUB), 08028 Barcelona, Spain
| | - Marta Vilaplana
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Jesús Daniel Gómez-Zepeda
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Sonia Brun
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Marta Garcia-Cajide
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Caroline Mauvezin
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Montserrat Jaumot
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Maria Dolors Pujol
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Jaime Rubio-Martínez
- Departament de Ciència de Materials i Química Física, Facultat de Química, Universitat de Barcelona & Institut de Recerca en Química Teòrica i Computacional (IQTCUB), 08028 Barcelona, Spain
- Correspondence: (J.R.-M.); (N.A.); Tel.: +34-934039263 (J.R.-M.); +34-934035267 (N.A.)
| | - Neus Agell
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
- Correspondence: (J.R.-M.); (N.A.); Tel.: +34-934039263 (J.R.-M.); +34-934035267 (N.A.)
| |
Collapse
|
3
|
Pallara C, Cabot D, Rivas J, Brun S, Seco J, Abuasaker B, Tarragó T, Jaumot M, Prades R, Agell N. Peptidomimetics designed to bind to RAS effector domain are promising cancer therapeutic compounds. Sci Rep 2022; 12:15810. [PMID: 36138080 PMCID: PMC9499927 DOI: 10.1038/s41598-022-19703-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Oncogenic RAS proteins are important for driving tumour formation, and for maintenance of the transformed phenotype, and thus their relevance as a cancer therapeutic target is undeniable. We focused here on obtaining peptidomimetics, which have good pharmacological properties, to block Ras–effector interaction. Computational analysis was used to identify hot spots of RAS relevant for these interactions and to screen a library of peptidomimetics. Nine compounds were synthesized and assayed for their activity as RAS inhibitors in cultured cells. Most of them induced a reduction in ERK and AKT activation by EGF, a marker of RAS activity. The most potent inhibitor disrupted Raf and PI3K interaction with oncogenic KRAS, corroborating its mechanism of action as an inhibitor of protein–protein interactions, and thus validating our computational methodology. Most interestingly, improvement of one of the compounds allowed us to obtain a peptidomimetic that decreased the survival of pancreatic cancer cell lines harbouring oncogenic KRAS.
Collapse
Affiliation(s)
- Chiara Pallara
- Iproteos S.L., Barcelona Science Park, Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Debora Cabot
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, C/Casanova 143, 08036, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Josep Rivas
- Iproteos S.L., Barcelona Science Park, Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Sonia Brun
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, C/Casanova 143, 08036, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jesús Seco
- Iproteos S.L., Barcelona Science Park, Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Baraa Abuasaker
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, C/Casanova 143, 08036, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Teresa Tarragó
- Iproteos S.L., Barcelona Science Park, Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Montserrat Jaumot
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, C/Casanova 143, 08036, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Roger Prades
- Iproteos S.L., Barcelona Science Park, Baldiri Reixac 10, 08028, Barcelona, Spain.
| | - Neus Agell
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, C/Casanova 143, 08036, Barcelona, Spain. .,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
4
|
Cabot D, Brun S, Paco N, Ginesta MM, Gendrau-Sanclemente N, Abuasaker B, Ruiz-Fariña T, Barceló C, Cuatrecasas M, Bosch M, Rentero C, Pons G, Estanyol JM, Capellà G, Jaumot M, Agell N. KRAS phosphorylation regulates cell polarization and tumorigenic properties in colorectal cancer. Oncogene 2021; 40:5730-5740. [PMID: 34333552 DOI: 10.1038/s41388-021-01967-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/07/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023]
Abstract
Oncogenic mutations of KRAS are found in the most aggressive human tumors, including colorectal cancer. It has been suggested that oncogenic KRAS phosphorylation at Ser181 modulates its activity and favors cell transformation. Using nonphosphorylatable (S181A), phosphomimetic (S181D), and phospho-/dephosphorylatable (S181) oncogenic KRAS mutants, we analyzed the role of this phosphorylation to the maintenance of tumorigenic properties of colorectal cancer cells. Our data show that the presence of phospho-/dephosphorylatable oncogenic KRAS is required for preserving the epithelial organization of colorectal cancer cells in 3D cultures, and for supporting subcutaneous tumor growth in mice. Interestingly, gene expression differed according to the phosphorylation status of KRAS. In DLD-1 cells, CTNNA1 was only expressed in phospho-/dephosphorylatable oncogenic KRAS-expressing cells, correlating with cell polarization. Moreover, lack of oncogenic KRAS phosphorylation leads to changes in expression of genes related to cell invasion, such as SERPINE1, PRSS1,2,3, and NEO1, and expression of phosphomimetic oncogenic KRAS resulted in diminished expression of genes involved in enterocyte differentiation, such as HNF4G. Finally, the analysis, in a public data set of human colorectal cancer, of the gene expression signatures associated with phosphomimetic and nonphosphorylatable oncogenic KRAS suggests that this post-translational modification regulates tumor progression in patients.
Collapse
Affiliation(s)
- Débora Cabot
- Department Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sònia Brun
- Department Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Noelia Paco
- Department Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Mireia M Ginesta
- Hereditary Cancer Program, Translational Research Laboratory, Catalan Institute of Oncology, ICO-IDIBELL, Hospitalet de Llobregat, Barcelona, Spain and Centro de Investigación Biomédica en Red en Cáncer (CIBERONC), Madrid, Spain
| | - Núria Gendrau-Sanclemente
- Department Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.,Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Hospital Duran i Reynals, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Baraa Abuasaker
- Department Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Triana Ruiz-Fariña
- Department Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Carles Barceló
- Department Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma de Mallorca, Spain
| | - Miriam Cuatrecasas
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Departament de Fonaments Clínics, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona; Pathology Department and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd) and Tumor Bank-Biobank, Hospital Clínic, Barcelona, Spain
| | - Marta Bosch
- Department Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Carles Rentero
- Department Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Gabriel Pons
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona and Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Josep M Estanyol
- Department Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain.,Proteomics Unit, CCiT-UB, Universitat de Barcelona, Barcelona, Spain
| | - Gabriel Capellà
- Hereditary Cancer Program, Translational Research Laboratory, Catalan Institute of Oncology, ICO-IDIBELL, Hospitalet de Llobregat, Barcelona, Spain and Centro de Investigación Biomédica en Red en Cáncer (CIBERONC), Madrid, Spain
| | - Montserrat Jaumot
- Department Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain. .,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Neus Agell
- Department Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain. .,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
5
|
Abdelkarim H, Leschinsky N, Jang H, Banerjee A, Nussinov R, Gaponenko V. The dynamic nature of the K-Ras/calmodulin complex can be altered by oncogenic mutations. Curr Opin Struct Biol 2021; 71:164-170. [PMID: 34311289 DOI: 10.1016/j.sbi.2021.06.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/03/2021] [Accepted: 06/17/2021] [Indexed: 12/13/2022]
Abstract
Oncogenic mutant K-Ras promotes cancer cell proliferation, migration, invasion, and survival by assembling signaling complexes. To date, the functional and structural roles of K-Ras mutations within these complexes are incompletely understood despite their mechanistic and therapeutic significance. Here, we review recent advances in understanding specific binding between K-Ras and the calcium sensor calmodulin. This interaction positively and negatively regulates diverse functions of K-Ras in cancer, suggesting flexibility in K-Ras/calmodulin complex formation. Also, structural data suggest that oncogenic K-Ras likely samples several conformational states, influencing its distinct assemblies with calmodulin and with other proteins. Understanding how K-Ras interacts with calmodulin and with other partners is essential to discovering novel inhibitors of K-Ras in cancer.
Collapse
Affiliation(s)
- Hazem Abdelkarim
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Nicholas Leschinsky
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD, 21702, USA
| | - Avik Banerjee
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
| | - Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD, 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Vadim Gaponenko
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, 60607, USA.
| |
Collapse
|
6
|
Pleiotropic Roles of Calmodulin in the Regulation of KRas and Rac1 GTPases: Functional Diversity in Health and Disease. Int J Mol Sci 2020; 21:ijms21103680. [PMID: 32456244 PMCID: PMC7279331 DOI: 10.3390/ijms21103680] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 12/21/2022] Open
Abstract
Calmodulin is a ubiquitous signalling protein that controls many biological processes due to its capacity to interact and/or regulate a large number of cellular proteins and pathways, mostly in a Ca2+-dependent manner. This complex interactome of calmodulin can have pleiotropic molecular consequences, which over the years has made it often difficult to clearly define the contribution of calmodulin in the signal output of specific pathways and overall biological response. Most relevant for this review, the ability of calmodulin to influence the spatiotemporal signalling of several small GTPases, in particular KRas and Rac1, can modulate fundamental biological outcomes such as proliferation and migration. First, direct interaction of calmodulin with these GTPases can alter their subcellular localization and activation state, induce post-translational modifications as well as their ability to interact with effectors. Second, through interaction with a set of calmodulin binding proteins (CaMBPs), calmodulin can control the capacity of several guanine nucleotide exchange factors (GEFs) to promote the switch of inactive KRas and Rac1 to an active conformation. Moreover, Rac1 is also an effector of KRas and both proteins are interconnected as highlighted by the requirement for Rac1 activation in KRas-driven tumourigenesis. In this review, we attempt to summarize the multiple layers how calmodulin can regulate KRas and Rac1 GTPases in a variety of cellular events, with biological consequences and potential for therapeutic opportunities in disease settings, such as cancer.
Collapse
|
7
|
Garrido E, Lázaro J, Jaumot M, Agell N, Rubio-Martinez J. Modeling and subtleties of K-Ras and Calmodulin interaction. PLoS Comput Biol 2018; 14:e1006552. [PMID: 30376570 PMCID: PMC6226203 DOI: 10.1371/journal.pcbi.1006552] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 11/09/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022] Open
Abstract
K-Ras, one of the most common small GTPases of the cell, still presents many riddles, despite the intense efforts to unveil its mysteries. Such is the case of its interaction with Calmodulin, a small acidic protein known for its role as a calcium ion sensor. Although the interaction between these two proteins and its biological implications have been widely studied, a model of their interaction has not been performed. In the present work we analyse this intriguing interaction by computational means. To do so, both conventional molecular dynamics and scaled molecular dynamics have been used. Our simulations suggest a model in which Calmodulin would interact with both the hypervariable region and the globular domain of K-Ras, using a lobe to interact with each of them. According to the presented model, the interface of helixes α4 and α5 of the globular domain of K-Ras would be relevant for the interaction with a lobe of Calmodulin. These results were also obtained when bringing the proteins together in a step wise manner with the umbrella sampling methodology. The computational results have been validated using SPR to determine the relevance of certain residues. Our results demonstrate that, when mutating residues of the α4-α5 interface described to be relevant for the interaction with Calmodulin, the interaction of the globular domain of K-Ras with Calmodulin diminishes. However, it is to be considered that our simulations indicate that the bulk of the interaction would fall on the hypervariable region of K-Ras, as many more interactions are identified in said region. All in all our simulations present a suitable model in which K-Ras could interact with Calmodulin at membrane level using both its globular domain and its hypervariable region to stablish an interaction that leads to an altered signalling. K-Ras is one of the most mutated oncogenes in human cancer. Although several studies validate K-Ras protein as good candidate for direct therapeutic targeting, pharmacologic targeting has not been successful. During the last years increasing evidences demonstrate that oncogenic K-Ras activity can be modulated in vivo by dimerization, nanoclustering at the plasma membrane or interaction with non-effector proteins, consequently opening new therapeutic strategies. We have previously demonstrated that Calmodulin, an ubiquitous Ca2+-binding protein, is one of this K-Ras interacting proteins and that it negatively modulates K-Ras signaling. Although experimental data were available showing the relevant regions for this interaction, a model of K-Ras and Calmodulin interaction was missing. In the present work by using different computational modeling techniques we obtained a model for this interaction that agrees with the experimental data. We believe the present model will help to better understand K-Ras regulation, and to design new inhibitors. For instance, base on our model, we can predict that the interaction can take place at the plasma membrane, and that since the surface of K-Ras that interact with Calmodulin is the same that it uses for dimerization, that Calmodulin could be inhibiting K-Ras dimerization.
Collapse
Affiliation(s)
- Eduardo Garrido
- Department of Materials Science and Physical Chemistry, Universitat de Barcelona, Institut de Recerca en Química Teòrica i Computacional (IQTCUB), Barcelona, Spain
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, IDIBAPS, Barcelona, Spain
| | - Juan Lázaro
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, IDIBAPS, Barcelona, Spain
| | - Montserrat Jaumot
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, IDIBAPS, Barcelona, Spain
| | - Neus Agell
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, IDIBAPS, Barcelona, Spain
- * E-mail: (NA); (JRM)
| | - Jaime Rubio-Martinez
- Department of Materials Science and Physical Chemistry, Universitat de Barcelona, Institut de Recerca en Química Teòrica i Computacional (IQTCUB), Barcelona, Spain
- * E-mail: (NA); (JRM)
| |
Collapse
|
8
|
Nussinov R, Tsai CJ, Jang H. Is Nanoclustering essential for all oncogenic KRas pathways? Can it explain why wild-type KRas can inhibit its oncogenic variant? Semin Cancer Biol 2018; 54:114-120. [PMID: 29307569 DOI: 10.1016/j.semcancer.2018.01.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/19/2017] [Accepted: 01/04/2018] [Indexed: 10/18/2022]
Abstract
Membrane-anchored oncogenic KRas can dimerize, form nanoclusters, and signal through the MAPK (Raf/MEK/ERK) and PI3Kα/Akt/mTOR. Both pathways are needed in KRAS-driven proliferation. Here we ask: Is oncogenic KRas nanoclustering (or dimerization) essential for all KRas signaling pathways? Raf kinase domain dimerization, thus MAPK activation, requires KRas nanoclusters. By contrast, the PI3Kα heterodimer acts as a monomeric unit; thus, does PI3Kα activation and PI3Kα/Akt/mTOR signaling require nanoclustering? Further, calmodulin binds only to oncogenic KRas4B. Here we ask: Does calmodulin downregulate KRas4B cancer development as suggested early on, or promote it? We also ask: Why is oncogenic KRas4B the most abundant isoform? Does wild-type Ras indeed inhibit its oncogenic variants as data appeared to suggest? And related to the last question, why is wild-type KRas a more potent inhibitor of its oncogenic form than wild-type NRas of its oncogenic form? Resolving these cardinal questions, and others, such as how exactly does RASSF5 (NORE1A) act as tumor suppressor, and why Ras isoforms tend to occur in distinct cancer types are crucial for effective pharmacology. In this review, we take a nanoclustering/dimerization-centric outlook and show that many questions can be explained by simply considering Ras nanoclustering.
Collapse
Affiliation(s)
- Ruth Nussinov
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Chung-Jung Tsai
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Hyunbum Jang
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| |
Collapse
|
9
|
Engin HB, Carlin D, Pratt D, Carter H. Modeling of RAS complexes supports roles in cancer for less studied partners. BMC BIOPHYSICS 2017; 10:5. [PMID: 28815022 PMCID: PMC5558186 DOI: 10.1186/s13628-017-0037-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background RAS protein interactions have predominantly been studied in the context of the RAF and PI3kinase oncogenic pathways. Structural modeling and X-ray crystallography have demonstrated that RAS isoforms bind to canonical downstream effector proteins in these pathways using the highly conserved switch I and II regions. Other non-canonical RAS protein interactions have been experimentally identified, however it is not clear whether these proteins also interact with RAS via the switch regions. Results To address this question we constructed a RAS isoform-specific protein-protein interaction network and predicted 3D complexes involving RAS isoforms and interaction partners to identify the most probable interaction interfaces. The resulting models correctly captured the binding interfaces for well-studied effectors, and additionally implicated residues in the allosteric and hyper-variable regions of RAS proteins as the predominant binding site for non-canonical effectors. Several partners binding to this new interface (SRC, LGALS1, RABGEF1, CALM and RARRES3) have been implicated as important regulators of oncogenic RAS signaling. We further used these models to investigate competitive binding and multi-protein complexes compatible with RAS surface occupancy and the putative effects of somatic mutations on RAS protein interactions. Conclusions We discuss our findings in the context of RAS localization to the plasma membrane versus within the cytoplasm and provide a list of RAS protein interactions with possible cancer-related consequences, which could help guide future therapeutic strategies to target RAS proteins. Electronic supplementary material The online version of this article (doi:10.1186/s13628-017-0037-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- H Billur Engin
- Division of Medical Genetics, Department of Medicine, Universsity of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093 USA
| | - Daniel Carlin
- Division of Medical Genetics, Department of Medicine, Universsity of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093 USA
| | - Dexter Pratt
- Division of Medical Genetics, Department of Medicine, Universsity of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093 USA
| | - Hannah Carter
- Division of Medical Genetics, Department of Medicine, Universsity of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093 USA
| |
Collapse
|
10
|
Jang H, Banerjee A, Chavan T, Gaponenko V, Nussinov R. Flexible-body motions of calmodulin and the farnesylated hypervariable region yield a high-affinity interaction enabling K-Ras4B membrane extraction. J Biol Chem 2017; 292:12544-12559. [PMID: 28623230 PMCID: PMC5535030 DOI: 10.1074/jbc.m117.785063] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 06/05/2017] [Indexed: 01/01/2023] Open
Abstract
In calmodulin (CaM)-rich environments, oncogenic KRAS plays a critical role in adenocarcinomas by promoting PI3K/Akt signaling. We previously proposed that at elevated calcium levels in cancer, CaM recruits PI3Kα to the membrane and extracts K-Ras4B from the membrane, organizing a K-Ras4B-CaM-PI3Kα ternary complex. CaM can thereby replace a missing receptor-tyrosine kinase signal to fully activate PI3Kα. Recent experimental data show that CaM selectively promotes K-Ras signaling but not of N-Ras or H-Ras. How CaM specifically targets K-Ras and how it extracts it from the membrane in KRAS-driven cancer is unclear. Obtaining detailed structural information for a CaM-K-Ras complex is still challenging. Here, using molecular dynamics simulations and fluorescence experiments, we observed that CaM preferentially binds unfolded K-Ras4B hypervariable regions (HVRs) and not α-helical HVRs. The interaction involved all three CaM domains including the central linker and both lobes. CaM specifically targeted the highly polybasic anchor region of the K-Ras4B HVR that stably wraps around CaM's acidic linker. The docking of the farnesyl group to the hydrophobic pockets located at both CaM lobes further enhanced CaM-HVR complex stability. Both CaM and K-Ras4B HVR are highly flexible molecules, suggesting that their interactions permit highly dynamic flexible-body motions. We, therefore, anticipate that the flexible-body interaction is required to extract K-Ras4B from the membrane, as conformational plasticity enables CaM to orient efficiently to the polybasic HVR anchor, which is partially diffused into the liquid-phase membrane. Our structural model of the CaM-K-Ras4B HVR association provides plausible clues to CaM's regulatory action in PI3Kα activation involving the ternary complex in cell proliferation signaling by oncogenic K-Ras.
Collapse
Affiliation(s)
- Hyunbum Jang
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, NCI-Frederick, Frederick, Maryland 21702
| | - Avik Banerjee
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois 60607
| | - Tanmay Chavan
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305
| | - Vadim Gaponenko
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois 60607.
| | - Ruth Nussinov
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, NCI-Frederick, Frederick, Maryland 21702; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
11
|
Abstract
K-Ras proteins are major drivers of human cancers, playing a direct causal role in about one million cancer cases/year. In cancers driven by mutant K-Ras, the protein is locked in the active, GTP-bound state constitutively, through a defect in the off-switch mechanism. As such, the mutant protein resembles the normal K-Ras protein from a structural perspective, making therapeutic attack extremely challenging. K-Ras is a member of a large family of related proteins, which share very similar GDP/GTP-binding domains, making specific therapies more difficult. Furthermore, Ras proteins lack pockets to which small molecules can bind with high affinity, with a few interesting exceptions. However, new insights into the structure and function of K-Ras proteins reveal opportunities for intervention that were not appreciated many years ago, when efforts were launched to develop K-Ras therapies. Furthermore, K-Ras undergoes post-translational modification and interactions with cellular signaling proteins that present additional therapeutic opportunities, such as specific binding to calmodulin and regulation of non-canonical Wnt signaling.
Collapse
|
12
|
Cox AD, Der CJ, Philips MR. Targeting RAS Membrane Association: Back to the Future for Anti-RAS Drug Discovery? Clin Cancer Res 2016; 21:1819-27. [PMID: 25878363 DOI: 10.1158/1078-0432.ccr-14-3214] [Citation(s) in RCA: 289] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
RAS proteins require membrane association for their biologic activity, making this association a logical target for anti-RAS therapeutics. Lipid modification of RAS proteins by a farnesyl isoprenoid is an obligate step in that association, and is an enzymatic process. Accordingly, farnesyltransferase inhibitors (FTI) were developed as potential anti-RAS drugs. The lack of efficacy of FTIs as anticancer drugs was widely seen as indicating that blocking RAS membrane association was a flawed approach to cancer treatment. However, a deeper understanding of RAS modification and trafficking has revealed that this was an erroneous conclusion. In the presence of FTIs, KRAS and NRAS, which are the RAS isoforms most frequently mutated in cancer, become substrates for alternative modification, can still associate with membranes, and can still function. Thus, FTIs failed not because blocking RAS membrane association is an ineffective approach, but because FTIs failed to accomplish that task. Recent findings regarding RAS isoform trafficking and the regulation of RAS subcellular localization have rekindled interest in efforts to target these processes. In particular, improved understanding of the palmitoylation/depalmitoylation cycle that regulates RAS interaction with the plasma membrane, endomembranes, and cytosol, and of the potential importance of RAS chaperones, have led to new approaches. Efforts to validate and target other enzymatically regulated posttranslational modifications are also ongoing. In this review, we revisit lessons learned, describe the current state of the art, and highlight challenging but promising directions to achieve the goal of disrupting RAS membrane association and subcellular localization for anti-RAS drug development. Clin Cancer Res; 21(8); 1819-27. ©2015 AACR. See all articles in this CCR Focus section, "Targeting RAS-Driven Cancers."
Collapse
Affiliation(s)
- Adrienne D Cox
- University of North Carolina at Chapel Hill, Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina.
| | - Channing J Der
- University of North Carolina at Chapel Hill, Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina.
| | - Mark R Philips
- Perlmutter Cancer Institute, New York University School of Medicine, New York, New York.
| |
Collapse
|
13
|
Alvarez-Moya B, Barceló C, Tebar F, Jaumot M, Agell N. CaM interaction and Ser181 phosphorylation as new K-Ras signaling modulators. Small GTPases 2014; 2:99-103. [PMID: 21776410 DOI: 10.4161/sgtp.2.2.15555] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Revised: 03/21/2011] [Accepted: 03/22/2011] [Indexed: 12/30/2022] Open
Abstract
The small G-protein Ras was the first oncogene to be identified and has a very important contribution to human cancer development (20-23% prevalence). K-RasB, one of the members of the Ras family, is the one that is most mutated and plays a prominent role in pancreatic, colon and lung cancer development. Ras proteins are membrane bound GTPases that cycle between inactive, GDP-bound and active, GTP-bound, states. Most of the research into K-RasB activity regulation has focused on the analysis of how GTP-exchange factors (GEFs) and GTPase activating proteins (GAPs) are regulated by external and internal signals. In contrast, oncogenic K-RasB has a very low GTPase activity and furthermore is not deactivated by GAPs. Consequently, the consensus was that activity of oncogenic K-RasB was not modulated. In this extra view we recapitulate some recent data showing that calmodulin binding to K-RasB inhibits phosphorylation of K-RasB at Ser181, near to the membrane anchoring domain, modulating signaling of both non-oncogenic and oncogenic K-RasB. This may be relevant to normal cell physiology, but also opens new therapeutic perspectives for the inhibition of oncogenic K-RasB signaling in tumors.
Collapse
Affiliation(s)
- Blanca Alvarez-Moya
- Departament de Biologia Cel·lular, Immunologia i Neurociències; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS); Facultat de Medicina; Universitat de Barcelona; Barcelona, Spain
| | | | | | | | | |
Collapse
|
14
|
Berchtold MW, Villalobo A. The many faces of calmodulin in cell proliferation, programmed cell death, autophagy, and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1843:398-435. [PMID: 24188867 DOI: 10.1016/j.bbamcr.2013.10.021] [Citation(s) in RCA: 236] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 10/24/2013] [Accepted: 10/26/2013] [Indexed: 12/21/2022]
Abstract
Calmodulin (CaM) is a ubiquitous Ca(2+) receptor protein mediating a large number of signaling processes in all eukaryotic cells. CaM plays a central role in regulating a myriad of cellular functions via interaction with multiple target proteins. This review focuses on the action of CaM and CaM-dependent signaling systems in the control of vertebrate cell proliferation, programmed cell death and autophagy. The significance of CaM and interconnected CaM-regulated systems for the physiology of cancer cells including tumor stem cells, and processes required for tumor progression such as growth, tumor-associated angiogenesis and metastasis are highlighted. Furthermore, the potential targeting of CaM-dependent signaling processes for therapeutic use is discussed.
Collapse
Key Words
- (4-[3,5-bis-[2-(4-hydroxy-3-methoxy-phenyl)-ethyl]-4,5-dihydro-pyrazol-1-yl]-benzoic acid
- (4-[3,5-bis-[2-(4-hydroxy-3-methoxy-phenyl)-vinyl]-4,5-dihydro-pyrazol-1-yl]-phenyl)-(4-methyl-piperazin-1-yl)-methanone
- (−) enantiomer of dihydropyrine 3-methyl-5-3-(4,4-diphenyl-1-piperidinyl)-propyl-1,4-dihydro-2,6-dimethyl-4-(3-nitrophenyl)-piridine-3,5-dicarboxylate-hydrochloride (niguldipine)
- 1-[N,O-bis(5-isoquinolinesulfonyl)-N-methyl-l-tyrosyl]-4-phenylpiperazine
- 12-O-tetradecanoyl-phorbol-13-acetate
- 2-chloro-(ε-amino-Lys(75))-[6-(4-(N,N′-diethylaminophenyl)-1,3,5-triazin-4-yl]-CaM adduct
- 3′-(β-chloroethyl)-2′,4′-dioxo-3,5′-spiro-oxazolidino-4-deacetoxy-vinblastine
- 7,12-dimethylbenz[a]anthracene
- Apoptosis
- Autophagy
- B859-35
- CAPP(1)-CaM
- Ca(2+) binding protein
- Calmodulin
- Cancer biology
- Cell proliferation
- DMBA
- EBB
- FL-CaM
- FPCE
- HBC
- HBCP
- J-8
- KAR-2
- KN-62
- KN-93
- N-(4-aminobutyl)-2-naphthalenesulfonamide
- N-(4-aminobutyl)-5-chloro-2-naphthalenesulfonamide
- N-(6-aminohexyl)-1-naphthalenesulfonamide
- N-(6-aminohexyl)-5-chloro-1-naphthalenesulfonamide
- N-8-aminooctyl-5-iodo-naphthalenesulfonamide
- N-[2-[N-(4-chlorocinnamyl)-N-methylaminomethyl]phenyl]-N-(2-hydroxyethyl)-4-methoxybenzenesulfonamide
- O-(4-ethoxyl-butyl)-berbamine
- RITC-CaM
- TA-CaM
- TFP
- TPA
- W-12
- W-13
- W-5
- W-7
- fluorescein-CaM adduct
- fluphenazine-N-2-chloroethane
- norchlorpromazine-CaM adduct
- rhodamine isothiocyanate-CaM adduct
- trifluoperazine
Collapse
Affiliation(s)
- Martin W Berchtold
- Department of Biology, University of Copenhagen, Copenhagen Biocenter 4-2-09 Ole Maaløes Vej 5, DK-2200 Copenhagen N, Denmark.
| | - Antonio Villalobo
- Instituto de Investigaciones Biomédicas, Department of Cancer Biology, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/Arturo Duperier 4, E-28029 Madrid, Spain.
| |
Collapse
|
15
|
Chavan TS, Abraham S, Gaponenko V. Application of reductive ¹³C-methylation of lysines to enhance the sensitivity of conventional NMR methods. Molecules 2013; 18:7103-19. [PMID: 23778120 PMCID: PMC6270119 DOI: 10.3390/molecules18067103] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 06/13/2013] [Accepted: 06/14/2013] [Indexed: 01/25/2023] Open
Abstract
NMR is commonly used to investigate macromolecular interactions. However, sensitivity problems hamper its use for studying such interactions at low physiologically relevant concentrations. At high concentrations, proteins or peptides tend to aggregate. In order to overcome this problem, we make use of reductive 13C-methylation to study protein interactions at low micromolar concentrations. Methyl groups in dimethyl lysines are degenerate with one 13CH3 signal arising from two carbons and six protons, as compared to one carbon and three protons in aliphatic amino acids. The improved sensitivity allows us to study protein-protein or protein-peptide interactions at very low micromolar concentrations. We demonstrate the utility of this method by studying the interaction between the post-translationally lipidated hypervariable region of a human proto-oncogenic GTPase K-Ras and a calcium sensor protein calmodulin. Calmodulin specifically binds K-Ras and modulates its downstream signaling. This binding specificity is attributed to the unique lipidated hypervariable region of K-Ras. At low micromolar concentrations, the post-translationally modified hypervariable region of K-Ras aggregates and binds calmodulin in a non-specific manner, hence conventional NMR techniques cannot be used for studying this interaction, however, upon reductively methylating the lysines of calmodulin, we detected signals of the lipidated hypervariable region of K-Ras at physiologically relevant nanomolar concentrations. Thus, we utilize 13C-reductive methylation of lysines to enhance the sensitivity of conventional NMR methods for studying protein interactions at low concentrations.
Collapse
Affiliation(s)
- Tanmay S. Chavan
- Department of Medicinal Chemistry, University of Illinois at Chicago, 900 S Ashland, Chicago, IL 60607, USA; E-Mail:
| | - Sherwin Abraham
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA 94305, USA; E-Mail:
| | - Vadim Gaponenko
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, 900 S Ashland, Chicago, IL 60607, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-312-355-4839; Fax: +1-312-413-0353
| |
Collapse
|
16
|
Zafra D, Nocito L, Domínguez J, Guinovart JJ. Sodium tungstate activates glycogen synthesis through a non-canonical mechanism involving G-proteins. FEBS Lett 2012; 587:291-6. [PMID: 23260418 DOI: 10.1016/j.febslet.2012.11.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 11/28/2012] [Accepted: 11/30/2012] [Indexed: 12/28/2022]
Abstract
Tungstate treatment ameliorates experimental diabetes by increasing liver glycogen deposition through an as yet unidentified mechanism. The signalling mechanism of tungstate was studied in CHOIR cells and primary cultured hepatocytes. This compound exerted its pro-glycogenic effects through a new G-protein-dependent and Tyr-Kinase Receptor-independent mechanism. Chemical or genetic disruption of G-protein signalling prevented the activation of the Ras/ERK cascade and the downstream induction of glycogen synthesis caused by tungstate. Thus, these findings unveil a novel non-canonical signalling pathway that leads to the activation of glycogen synthesis and that could be exploited as an approach to treat diabetes.
Collapse
Affiliation(s)
- Delia Zafra
- Institute for Research in Biomedicine (IRB Barcelona) and Department of Biochemistry and Molecular Biology, University of Barcelona, and CIBER de Diabetes y Enfermedades Metabólicas (CIBERDEM), Baldiri Reixac 10-12, E-08028 Barcelona, Spain
| | | | | | | |
Collapse
|
17
|
A stochastic signaling network mediates the probabilistic induction of cerebellar long-term depression. J Neurosci 2012; 32:9288-300. [PMID: 22764236 DOI: 10.1523/jneurosci.5976-11.2012] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Many cellular processes involve a small number of molecules and undergo stochastic fluctuations in their levels of activity. Cerebellar long-term depression (LTD) is a form of synaptic plasticity expressed as a reduction in the number of synaptic AMPA receptors (AMPARs) in Purkinje cells. We developed a stochastic model of the LTD signaling network, including a PKC-ERK-cPLA(2) positive feedback loop and mechanisms of AMPAR trafficking, and tuned the model to replicate calcium uncaging experiments. The signaling network activity in single synapses switches between two discrete stable states (LTD and non-LTD) in a probabilistic manner. The stochasticity of the signaling network causes threshold dithering and allows at the macroscopic level for many different and stable mean magnitudes of depression. The probability of LTD occurrence in a single spine is only modulated by the concentration and duration of the signal used to trigger it, and inputs with the same magnitude can give rise to two different responses; there is no threshold for the input signal. The stochasticity is intrinsic to the signaling network and not mostly dependent on noise in the calcium input signal, as has been suggested previously. The activities of the ultrasensitive ERK and of cPLA(2) undergo strong stochastic fluctuations. Conversely, PKC, which acts as a noise filter, is more constantly activated. Systematic variation of the biochemical population size demonstrates that threshold dithering and the absence of spontaneous LTD depend critically on the number of molecules in a spine, indicating constraints on spine size in Purkinje cells.
Collapse
|
18
|
Chen HP, He M, Mei ZJ, Huang QR, Huang M. Sasanquasaponin up-regulates anion exchanger 3 expression and elicits cardioprotection via NO/RAS/ERK1/2 pathway. Can J Physiol Pharmacol 2012; 90:873-80. [PMID: 22693949 DOI: 10.1139/y2012-072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have shown recently that sasanquasaponin (SQS) can inhibit ischemia/reperfusion-induced elevation of intracellular Cl(-) concentration ([Cl(-)](i)) and elicit cardioprotection by up-regulating anion exchanger 3 (AE(3)) expression. In the present study, we futher analysed the intracellular signal transduction pathways by which SQS up-regulates AE(3) expression and elicits cardioprotection. Cardiomyocytes were incubated for 24 h with or without 10 µmol/L SQS, followed by simulated ischemia/reperfusion (sI/R). NO formation, Ras activity, and extracellular-regulated kinase 1/2 (ERK1/2) phosphorylation were measured appropriately. We showed that SQS pretreatment efficiently attenuated viability loss and lactate dehydrogenase leakage induced by sI/R in cardiomyocytes. Moreover, SQS induced NO production and promoted Ras activation, which futher promoted extracellular-regulated kinase 1/2 (ERK1/2) phosphorylation. These effects were paralleled by an increase in AE(3) expression. However, when the cardiomyocytes were treated with 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-l-oxyl-3-oxide (c-PTIO; an NO scavenger), S-trans-trans-farnesylthiosalicylic acid (FTS) (a Ras inhibitor), U0126 (an ERK1/2 inhibitor), respectively, the increase in AE(3) expression occurring during SQS pretreatment was almost completely abolished and, as a result, SQS-induced cardioprotection was prevented. Our findings indicate that SQS might up-regulate AE(3) expression through NO/Ras/ERK1/2 signal pathway to elicit cardioprotection in cultured cardiomyocytes.
Collapse
Affiliation(s)
- He-Ping Chen
- Department of Pharmacology & Molecular Therapeutics, School of Pharmaceutical Science, Nanchang University, PR China
| | | | | | | | | |
Collapse
|
19
|
Abstract
Ever since their discovery as cellular counterparts of viral oncogenes more than 25 years ago, much progress has been made in understanding the complex networks of signal transduction pathways activated by oncogenic Ras mutations in human cancers. The activity of Ras is regulated by nucleotide exchange factors (GEFs) and GTPase activating proteins (GAPs), and much emphasis has been put into the biochemical and structural analysis of the Ras/GAP complex. The mechanisms by which GAPs catalyze Ras-GTP hydrolysis have been clarified and revealed that oncogenic Ras mutations confer resistance to GAPs and remain constitutively active. However, it is yet unclear how cells coordinate the large and divergent GAP protein family to promote Ras inactivation and ensure a certain biological response. Different domain arrangements in GAPs to create differential protein-protein and protein-lipid interactions are probably key factors determining the inactivation of the 3 Ras isoforms H-, K-, and N-Ras and their effector pathways. In recent years, in vitro as well as cell- and animal-based studies examining GAP activity, localization, interaction partners, and expression profiles have provided further insights into Ras inactivation and revealed characteristics of several GAPs to exert specific and distinct functions. This review aims to summarize knowledge on the cell biology of RasGAP proteins that potentially contributes to differential regulation of spatiotemporal Ras signaling.
Collapse
Affiliation(s)
- Thomas Grewal
- Faculty of Pharmacy, University of Sydney, Sydney, NSW, Australia
| | | | | | | |
Collapse
|
20
|
Arozarena I, Calvo F, Crespo P. Ras, an actor on many stages: posttranslational modifications, localization, and site-specified events. Genes Cancer 2011; 2:182-94. [PMID: 21779492 DOI: 10.1177/1947601911409213] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Among the wealth of information that we have gathered about Ras in the past decade, the introduction of the concept of space in the field has constituted a major revolution that has enabled many pieces of the Ras puzzle to fall into place. In the early days, it was believed that Ras functioned exclusively at the plasma membrane. Today, we know that within the plasma membrane, the 3 Ras isoforms-H-Ras, K-Ras, and N-Ras-occupy different microdomains and that these isoforms are also present and active in endomembranes. We have also discovered that Ras proteins are not statically associated with these localizations; instead, they traffic dynamically between compartments. And we have learned that at these localizations, Ras is under site-specific regulatory mechanisms, distinctively engaging effector pathways and switching on diverse genetic programs to generate different biological responses. All of these processes are possible in great part due to the posttranslational modifications whereby Ras proteins bind to membranes and to regulatory events such as phosphorylation and ubiquitination that Ras is subject to. As such, space and these control mechanisms act in conjunction to endow Ras signals with an enormous signal variability that makes possible its multiple biological roles. These data have established the concept that the Ras signal, instead of being one single, homogeneous entity, results from the integration of multiple, site-specified subsignals, and Ras has become a paradigm of how space can differentially shape signaling.
Collapse
Affiliation(s)
- Imanol Arozarena
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Consejo Superior de Investigaciones Científicas (CSIC)-IDICAN-Universidad de Cantabria, Departamento de Biología Molecular, Facultad de Medicina, Cantabria, Spain
| | | | | |
Collapse
|
21
|
Both the C-terminal polylysine region and the farnesylation of K-RasB are important for its specific interaction with calmodulin. PLoS One 2011; 6:e21929. [PMID: 21750741 PMCID: PMC3130059 DOI: 10.1371/journal.pone.0021929] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 06/09/2011] [Indexed: 02/03/2023] Open
Abstract
Background Ras protein, as one of intracellular signal switches, plays various roles in several cell activities such as differentiation and proliferation. There is considerable evidence showing that calmodulin (CaM) binds to K-RasB and dissociates K-RasB from membrane and that the inactivation of CaM is able to induce K-RasB activation. However, the mechanism for the interaction of CaM with K-RasB is not well understood. Methodology/Principal Findings Here, by applying fluorescence spectroscopy and isothermal titration calorimetry, we have obtained thermodynamic parameters for the interaction between these two proteins and identified the important elements of K-RasB for its interaction with Ca2+/CaM. One K-RasB molecule interacts with one CaM molecule in a GTP dependent manner with moderate, micromolar affinity at physiological pH and physiologic ionic strength. Mutation in the polybasic domain of K-Ras decreases the binding affinity. By using a chimera in which the C-terminal polylysine region of K-RasB has been replaced with that of H-Ras and vice versa, we find that at physiological pH, H-Ras-(KKKKKK) and Ca2+/CaM formed a 1∶1 complex with an equilibrium association constant around 105 M−1, whereas no binding reaction of K-RasB-(DESGPC) with Ca2+/CaM is detected. Furthermore, the interaction of K-RasB with Ca2+/CaM is found to be enhanced by the farnesylation of K-RasB. Conclusions/Significance We demonstrate that the polylysine region of K-RasB not only contributes importantly to the interaction of K-RasB with Ca2+/CaM, but also defines its isoform specific interaction with Ca2+/CaM. The farnesylation of K-RasB is also important for its specific interaction with Ca2+/CaM. Information obtained here can enhance our understanding of how CaM interacts with K-RasB in physiological environments.
Collapse
|
22
|
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
|
23
|
Lin MC, Lin SB, Lee SC, Lin CC, Hui CF, Chen JY. Antimicrobial peptide of an anti-lipopolysaccharide factor modulates of the inflammatory response in RAW264.7 cells. Peptides 2010; 31:1262-72. [PMID: 20385189 DOI: 10.1016/j.peptides.2010.04.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Revised: 04/02/2010] [Accepted: 04/05/2010] [Indexed: 12/23/2022]
Abstract
In this study, to clarify the protective mechanism of a peptide from shrimp anti-lipopolysaccharide (LPS) factor (SALF) against endotoxin shock, we evaluated the effects of the SALF and LPS on the production and release of tumor necrosis factor (TNF)-alphain vitro using the RAW264.7 murine macrophage cell line. Stimulation by LPS induced the production of inflammatory cytokines, and the SALF was able to modulate TNF-alpha production in LPS-stimulated RAW264.7 cells. Microarray studies revealed a transcriptional profile which was assessed in the presence or absence of the SALF by a quantitative real-time polymerase chain reaction. Pretreatment with the SALF significantly downregulated the expression of nuclear factor (NF)-kappaB in the presence of LPS. In contrast, pretreatment with the SALF significantly elevated the expressions of Anp32a, CLU, and SLPI, which are considered to be immune-related genes in the presence of LPS. Inhibitor studies suggested that the SALF's modulation of LPS-induced TNF-alpha production involved a complex mechanism with mitogen-activated protein kinase kinase, calcium, and protein kinase C. The data from this study, which imply that the SALF can suppress TNF-alpha production, suggest a role for the SALF in the defense mechanism which can potentially be applied to mammals for endotoxin treatment.
Collapse
Affiliation(s)
- Ming-Ching Lin
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dahuen Rd., Jiaushi, Ilan 262, Taiwan
| | | | | | | | | | | |
Collapse
|
24
|
The effects of silencing EDF-1 in human endothelial cells. Atherosclerosis 2010; 211:55-60. [PMID: 20185128 DOI: 10.1016/j.atherosclerosis.2010.01.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 01/18/2010] [Accepted: 01/19/2010] [Indexed: 11/21/2022]
Abstract
OBJECTIVE EDF-1, a 16 kDa highly conserved intracellular protein, serves as a calmodulin binding protein and, upon nuclear translocation, functions as a coactivator of several transcription factors. To understand whether EDF-1 is implicated in regulating endothelial function, we silenced EDF-1 expression using small hairpin (sh) RNA. METHODS Human umbilical vein endothelial cells (HUVEC) were utilized and EDF-1 levels were detected by western blot. Cell proliferation, cell organization in fibrin gel and nitric oxide release were evaluated in cells silencing EDF-1 after transfection with shRNA. RESULTS EDF-1 was downregulated in quiescent and senescent HUVEC, whereas it was upregulated in proliferating cells. Knocking down EDF-1 promoted the acquisition of a spindle phenotype, inhibited cell proliferation, accelerated the organization into capillary-like networks on fibrin gels and induced the production of nitric oxide (NO). While the total amounts and the degree of phosphorylation of endothelial NO synthase are not altered in cells silencing EDF-1, we found an increased interaction between calmodulin and endothelial NO synthase. Accordingly, the calmodulin inhibitor calmidazolium significantly decreased NO release in cells silencing EDF-1. These results suggest that knocking down EDF-1 might increase free calmodulin which ultimately activates endothelial NO synthase. CONCLUSIONS Since EDF-1 (i) is involved in the control of endothelial proliferation and organization, events which are crucial to repair damages to the vessel wall, and (ii) increases NO, which exerts anti-atherogenic and anti-thrombotic effects, we conclude that EDF-1 is implicated in molecular events that are pivotal to endothelial function and, therefore, to vascular integrity.
Collapse
|
25
|
Sánchez-González P, Jellali K, Villalobo A. Calmodulin-mediated regulation of the epidermal growth factor receptor. FEBS J 2009; 277:327-42. [PMID: 19951361 DOI: 10.1111/j.1742-4658.2009.07469.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In this review, we first describe the mechanisms by which the epidermal growth factor receptor generates a Ca(2+) signal and, subsequently, we compile the available experimental evidence regarding the role that the Ca(2+)/calmodulin complex, formed after the rise in cytosolic free Ca(2+) concentration, exerts on the receptor. We focus not only on the indirect action that Ca(2+)/calmodulin exerts on the epidermal growth factor receptor, as a result of the activation of distinct calmodulin-dependent kinases, but also, and more extensively, on the direct interaction of Ca(2+)/calmodulin with the receptor. We also describe several mechanistic models that could account for the Ca(2+)/calmodulin-mediated regulation of epidermal growth factor receptor activity. The control exerted by calmodulin on distinct epidermal growth factor receptor-mediated cellular functions is also discussed. Finally, the phosphorylation of this Ca(2+) sensor by the epidermal growth factor receptor is highlighted.
Collapse
Affiliation(s)
- Pablo Sánchez-González
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | | | | |
Collapse
|
26
|
Abraham SJ, Nolet RP, Calvert RJ, Anderson LM, Gaponenko V. The hypervariable region of K-Ras4B is responsible for its specific interactions with calmodulin. Biochemistry 2009; 48:7575-83. [PMID: 19583261 PMCID: PMC2729490 DOI: 10.1021/bi900769j] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
K-Ras4B belongs to the family of p21 Ras GTPases, which play an important role in cell proliferation, survival, and motility. The p21 Ras proteins, such as K-Ras4B, K-Ras4A, H-Ras, and N-Ras, share 85% sequence homology and activate very similar signaling pathways. Only the C-terminal hypervariable regions differ significantly. A growing body of literature demonstrates that each Ras isoform possesses unique functions in normal physiological processes as well as in pathogenesis. One of the central questions in the field of Ras biology is how these very similar proteins achieve such remarkable specificity in protein-protein interactions that regulate signal transduction pathways. Here we explore specific binding of K-Ras4B to calmodulin. Using NMR techniques and isothermal titration calorimetry, we demonstrate that the hypervariable region of K-Ras4B contributes in a major way to the interaction with calmodulin, while the catalytic domain of K-Ras4B provides a way to control the interaction by nucleotide binding. The hypervariable region of K-Ras4B binds specifically to the C-terminal domain of Ca(2+)-loaded calmodulin with micromolar affinity, while the GTP-gamma-S-loaded catalytic domain of K-Ras4B may interact with the N-terminal domain of calmodulin.
Collapse
Affiliation(s)
- Sherwin J. Abraham
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, IL 60607
| | - Ryan P. Nolet
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, IL 60607
| | - Richard J. Calvert
- Laboratory of Comparative Carcinogenesis, National Cancer Institute, Frederick, MD 21702
- Division of Bioanalytical Chemistry, U.S. Food and Drug Administration, College Park, MD 20740
| | - Lucy M. Anderson
- Laboratory of Comparative Carcinogenesis, National Cancer Institute, Frederick, MD 21702
| | - Vadim Gaponenko
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, IL 60607
| |
Collapse
|
27
|
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
|
28
|
Abstract
At the cell surface, activation of the epidermal growth factor (EGF) receptor triggers a complex network of signalling events that regulate a variety of cellular processes. For signal termination, the activated EGF receptor is internalised and targeted to lysosomes for degradation. Microdomain localization at the plasma membrane and endocytic transport of the EGFR is important for the formation of compartment-specific signalling complexes and is regulated by scaffolding and targeting proteins. This includes Ca2+-effector proteins, such as calmodulin and annexins (Anx), in particular AnxA1, AnxA2, AnxA6 and as shown recently,AnxA8. Given that these annexins show differences in their expression patterns, subcellular localization and mode of action, they are likely to differentially contribute and cooperate in the fine-tuning of EGFR activity. In support of this hypothesis, current literature suggests these annexins to be involved in different steps that control the endocytic transport and signalling of the EGF receptor. This review summarizes how the coordinated activity of AnxA1, AnxA2, AnxA6 and AnxA8 can contribute to regulate EGF receptor localization and activity.
Collapse
Affiliation(s)
- Thomas Grewal
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Sydney, Sydney, Australia.
| | | |
Collapse
|
29
|
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
|
30
|
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
|
31
|
Jasinski P, Welsh B, Galvez J, Land D, Zwolak P, Ghandi L, Terai K, Dudek AZ. A novel quinoline, MT477: suppresses cell signaling through Ras molecular pathway, inhibits PKC activity, and demonstrates in vivo anti-tumor activity against human carcinoma cell lines. Invest New Drugs 2007; 26:223-32. [PMID: 17957339 DOI: 10.1007/s10637-007-9096-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2007] [Accepted: 10/10/2007] [Indexed: 01/19/2023]
Abstract
MT477 is a novel thiopyrano[2,3-c]quinoline that has been identified using molecular topology screening as a potential anticancer drug with a high activity against protein kinase C (PKC) isoforms. The objective of the present study was to determine the mechanism of action of MT477 and its activity against human cancer cell lines. MT477 interfered with PKC activity as well as phosphorylation of Ras and ERK1/2 in H226 human lung carcinoma cells. It also induced poly-caspase-dependent apoptosis. MT477 had a dose-dependent (0.006 to 0.2 mM) inhibitory effect on cellular proliferation of H226, MCF-7, U87, LNCaP, A431 and A549 cancer cell lines as determined by in vitro proliferation assays. Two murine xenograft models of human A431 and H226 lung carcinoma were used to evaluate tumor response to intraperitoneal administration of MT477 (33 microg/kg, 100 microg/kg, and 1 mg/kg). Tumor growth was inhibited by 24.5% in A431 and 43.67% in H226 xenografts following MT477 treatment, compared to vehicle controls (p < 0.05). In conclusion, our empirical findings are consistent with molecular modeling of MT477's activity against PKC. We also found, however, that its mechanism of action occurs through suppressing Ras signaling, indicating that its effects on apoptosis and tumor growth in vivo may be mediated by Ras as well as PKC. We propose, therefore, that MT477 warrants further development as an anticancer drug.
Collapse
Affiliation(s)
- Piotr Jasinski
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Mayo Mail Code 480, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Cerezo-Guisado M, GarcíA-Román N, García-MaríN L, Álvarez-Barrientos A, Bragado M, Lorenzo M. Lovastatin inhibits the extracellular-signal-regulated kinase pathway in immortalized rat brain neuroblasts. Biochem J 2007; 401:175-83. [PMID: 16952276 PMCID: PMC1698684 DOI: 10.1042/bj20060731] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We have shown previously that lovastatin, a 3-hydroxy-3-methyl- glutaryl coenzyme A reductase inhibitor, induces apoptosis in spontaneously immortalized rat brain neuroblasts. In the present study, we analysed the intracellular signal transduction pathways by which lovastatin induces neuroblast apoptosis. We showed that lovastatin efficiently inhibited Ras activation, which was associated with a significant decrease in ERK1/2 (extracellular-signal-regulated kinase 1/2) phosphorylation. Lovastatin also decreased CREB phosphorylation and CREB-mediated gene expression. The effects of lovastatin on the Ras/ERK1/2/CREB pathway were time- and concentration-dependent and fully prevented by mevalonate. In addition, we showed that two MEK [MAPK (mitogen-activated protein kinase)/ERK kinase] inhibitors, PD98059 and PD184352, were poor inducers of apoptosis in serum-treated neuroblasts. However, these inhibitors significantly increased apoptosis induced by lovastatin treatment. Furthermore, we showed that pharmacological inhibition of both MEK and phosphoinositide 3-kinase activities was able to induce neuroblast apoptosis with similar efficacy as lovastatin. Our results suggest that lovastatin triggers neuroblast apoptosis by regulating several signalling pathways, including the Ras/ERK1/2 pathway. These findings might also contribute to elucidate the intracellular mechanisms involved in the central nervous system side effects associated with statin therapy.
Collapse
Affiliation(s)
- Maria Isabel Cerezo-Guisado
- *Departamento de Bioquímica, Biología Molecular y Genética Universidad de Extremadura, E-10071 Cáceres, Spain
| | - Natalia GarcíA-Román
- †Departamento de Bioquímica y Biología Molecular, Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain
| | | | | | - Maria Julia Bragado
- *Departamento de Bioquímica, Biología Molecular y Genética Universidad de Extremadura, E-10071 Cáceres, Spain
| | - Maria Jesús Lorenzo
- *Departamento de Bioquímica, Biología Molecular y Genética Universidad de Extremadura, E-10071 Cáceres, Spain
- To whom correspondence should be addressed (email )
| |
Collapse
|
33
|
Kim HS, Kim JW, Gang J, Wen J, Koh SS, Koh JS, Chung HH, Song SY. The farnesyltransferase inhibitor, LB42708, inhibits growth and induces apoptosis irreversibly in H-ras and K-ras-transformed rat intestinal epithelial cells. Toxicol Appl Pharmacol 2006; 215:317-29. [PMID: 16712893 DOI: 10.1016/j.taap.2006.03.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2005] [Revised: 03/20/2006] [Accepted: 03/22/2006] [Indexed: 02/04/2023]
Abstract
LB42708 (LB7) and LB42908 (LB9) are pyrrole-based orally active farnesyltransferase inhibitors (FTIs) that have similar structures. The in vitro potencies of these compounds against FTase and GGTase I are remarkably similar, and yet they display different activity in apoptosis induction and morphological reversion of ras-transformed rat intestinal epithelial (RIE) cells. Both FTIs induced cell death despite K-ras prenylation, implying the participation of Ras-independent mechanism(s). Growth inhibition by these two FTIs was accompanied by G1 and G2/M cell cycle arrests in H-ras and K-ras-transformed RIE cells, respectively. We identified three key markers, p21(CIP1/WAF1), RhoB and EGFR, that can explain the differences in the molecular mechanism of action between two FTIs. Only LB7 induced the upregulation of p21(CIP1/WAF1) and RhoB above the basal level that led to the cell cycle arrest and to distinct morphological alterations of ras-transformed RIE cells. Both FTIs successfully inhibited the ERK and activated JNK in RIE/K-ras cells. While the addition of conditioned medium from RIE/K-ras reversed the growth inhibition of ras-transformed RIE cells by LB9, it failed to overcome the growth inhibitory effect of LB7 in both H-ras- and K-ras-transformed RIE cells. We found that LB7, but not LB9, decreased the expression of EGFRs that confers the cellular unresponsiveness to EGFR ligands. These results suggest that LB7 causes the induction of p21(CIP1/WAF1) and RhoB and downregulation of EGFR that may serve as critical steps in the mechanism by which FTIs trigger irreversible inhibitions on the cell growth and apoptosis in ras-transformed cells.
Collapse
Affiliation(s)
- Han-Soo Kim
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Liao J, Planchon SM, Wolfman JC, Wolfman A. Growth factor-dependent AKT activation and cell migration requires the function of c-K(B)-Ras versus other cellular ras isoforms. J Biol Chem 2006; 281:29730-8. [PMID: 16908523 DOI: 10.1074/jbc.m600668200] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
K-Ras-negative fibroblasts are defective in their steady-state expression of MMP-2. This occurs through c-K(B)-Ras dependent regulation of basal levels of AKT activity. In this report, we have extended those studies to demonstrate that in the absence of K-Ras expression, PDGF-BB fails to induce significant AKT activation, although this was not the case in N-Ras-negative cells. This phenotype was directly linked to PDGF-dependent cell migration. All of the independently immortalized K-Ras-negative cells failed to migrate upon the addition of PDGF. Only ectopic expression of c-K(B)-Ras, not c-K(A)-Ras nor oncogenic N-Ras, could restore both PDGF-dependent AKT activation and cell migration. Since most Ras binding partners can interact with all Ras isoforms, the specificity of PDGF-dependent activation of AKT and enhanced cell migration suggests that these outcomes are likely to be regulated through a c-K(B)-Ras-specific binding partner. Others have published that of the four Ras isoforms, only K(B)-Ras can form a stable complex with calmodulin (CaM). Along those lines, we provide evidence that 1) PDGF addition results in increased levels of a complex between c-K(B)-Ras and CaM and 2) the biological outcomes that are strictly dependent on c-K(B)-Ras (AKT activation and cell migration) are blocked by CaM antagonists. The PDGF-dependent activation of ERK is unaffected by the absence of K(B)-Ras and presence of CaM antagonists. This is the first example of a linkage between a specific biological outcome, cell migration, and the activity of a single Ras isoform, c-K(B)-Ras.
Collapse
Affiliation(s)
- Jinhui Liao
- Department of Cell Biology, Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, Ohio 44195, USA
| | | | | | | |
Collapse
|
35
|
Cullen PJ. Decoding complex Ca2+ signals through the modulation of Ras signaling. Curr Opin Cell Biol 2006; 18:157-61. [PMID: 16488591 DOI: 10.1016/j.ceb.2006.02.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2005] [Accepted: 02/08/2006] [Indexed: 12/22/2022]
Abstract
Calcium (Ca(2+)) is a universal regulator of a wide variety of cellular processes. For such control to be achieved, information is encoded within spatial and temporal components of the underlying Ca(2+) signal. One pathway through which Ca(2+) signals are decoded is the Ras binary switch. Here I describe some recent advances that have shed light on how cells can decode the spatial and temporal aspects of Ca(2+) signals through the regulation of this important signalling switch.
Collapse
Affiliation(s)
- Peter J Cullen
- Department of Biochemistry, School of Medical Sciences, University of Bristol, Bristol BS8 1TD, UK.
| |
Collapse
|
36
|
Cook SJ, Lockyer PJ. Recent advances in Ca(2+)-dependent Ras regulation and cell proliferation. Cell Calcium 2006; 39:101-12. [PMID: 16343616 DOI: 10.1016/j.ceca.2005.10.014] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2005] [Revised: 10/17/2005] [Accepted: 10/27/2005] [Indexed: 12/20/2022]
Abstract
Our understanding of the mechanisms whereby growth factors stimulate cell proliferation through the Ras pathway stems largely from studies of the canonical pathway involving recruitment of Ras activators and inhibitors to the vicinity of receptor tyrosine kinases via phosphotyrosine-binding adaptor proteins. Ca(2+) has seldom joined the party, despite the identification of phospholipase Cgamma and Ca(2+) entry as receptor tyrosine kinase-dependent signals. Mechanisms by which Ca(2+) can directly influence Ras activity have remained relatively elusive. Similarly, the mechanisms whereby Ca(2+) modulates the cell cycle have been equally murky, and yet there are some interesting parallels in the role of Ras and Ca(2+) in cell cycle re-entry. This review focuses on a number of novel mechanisms that link Ca(2+) with the regulation of Ras activity and signaling output. Their collective discovery adds to the complexities of Ras regulation and raises further questions about the role of Ca(2+) signals in Ras-dependent cell proliferation.
Collapse
Affiliation(s)
- Simon J Cook
- Laboratory of Molecular Signaling, The Babraham Institute, Babraham Research Campus, Cambridge CB2 4AT, UK
| | | |
Collapse
|
37
|
Grewal T, Enrich C. Molecular mechanisms involved in Ras inactivation: the annexin A6–p120GAP complex. Bioessays 2006; 28:1211-20. [PMID: 17120209 DOI: 10.1002/bies.20503] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
In mammalian cells, a complex network of signaling pathways tightly regulates a variety of cellular processes, such as proliferation and differentiation. New insights from one of the most-important signaling cascades involved in oncogenesis, the Ras-Raf-MAPK pathway, suggest that the subcellular localisation and assembly of signaling modules of this pathway is crucial to control the biological response. This commonly requires membrane targeting events that are mediated by adaptor/scaffold proteins. Of particular interest is the translocation and complex formation of GTPase-activating proteins (GAPs), such as p120GAP, at the plasma membrane to inactivate Ras. Recent studies indicate that one member of the annexin family, annexin A6 acts as a targeting protein for p120GAP. This review discusses how annexin A6 modulates the involvement of negative regulators of the Ras-Raf-MAPK pathway contributing to Ras inactivation.
Collapse
Affiliation(s)
- Thomas Grewal
- Centre for Immunology, St. Vincent's Hospital, University of New South Wales, Sydney, Australia.
| | | |
Collapse
|
38
|
Abstract
The plasma membrane is a complex, dynamic structure that provides platforms for the assembly of many signal transduction pathways. These platforms have the capacity to impose an additional level of regulation on cell signalling networks. In this review, we will consider specifically how Ras proteins interact with the plasma membrane. The focus will be on recent studies that provide novel spatial and dynamic insights into the micro-environments that different Ras proteins utilize for signal transduction. We will correlate these recent studies suggesting Ras proteins might operate within a heterogeneous plasma membrane with earlier biochemical work on Ras signal transduction.
Collapse
Affiliation(s)
- John F Hancock
- Institute for Molecular Bioscience, University of Queensland, Brisbane, 4072, Australia.
| | | |
Collapse
|
39
|
Abstract
Ras GTPases are universal molecular switches that act as kinetic timers of signal transduction events. They are post-translationally modified by the addition of lipid groups to their hypervariable carboxyl termini, which plug the proteins to membranes and influence their dynamic sorting and trafficking. For the past twenty years, the plasma membrane has been considered to be the predominant platform from which Ras operates. Recent work using live-cell imaging and novel probes to visualize where and when Ras is active has supported this long-held belief. However, an equally fascinating aspect of these imaging studies has been the discovery of dynamic Ras activity, as well as distinct signal output, from intracellular organelles. Activation of Ras on the Golgi exhibits kinetics different from Ras activation on the plasma membrane, and compartmentalized Ras signalling seems particularly prominent in lymphocytes. However, data on the spatial and temporal regulation of Ras activity has frequently differed depending on the nature of the probe, the cell type and the stimulus. Nevertheless, because Ras traffics through endomembranes en route to the plasma membrane, it seems likely that Ras can signal from such compartments. The burning question in this field concerns the significance of this observation for endogenous Ras signalling output.
Collapse
Affiliation(s)
- Simon A Walker
- Laboratory of Molecular Signalling, The Babraham Institute, Babraham Research Campus, Babraham, Cambridge, CB2 4AT, UK
| | | |
Collapse
|
40
|
Rentero C, Evans R, Wood P, Tebar F, Vilà de Muga S, Cubells L, de Diego I, Hayes TE, Hughes WE, Pol A, Rye KA, Enrich C, Grewal T. Inhibition of H-Ras and MAPK is compensated by PKC-dependent pathways in annexin A6 expressing cells. Cell Signal 2005; 18:1006-16. [PMID: 16183252 DOI: 10.1016/j.cellsig.2005.08.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2005] [Revised: 08/22/2005] [Accepted: 08/22/2005] [Indexed: 01/06/2023]
Abstract
High-density lipoprotein (HDL)-induced activation of the Ras/MAPK pathway can be mediated by protein kinase C (PKC)-dependent and independent pathways. Although both pathways co-exist in cells, we showed that binding of HDL to scavenger receptor BI (SR-BI) in CHO cells activates Ras and MAPK in a PKC-independent manner. We have recently identified that HDL-induced activation of Ras and Raf-1 is reduced in annexin A6 expressing CHO cells (CHOanx6). In the present study we demonstrate that despite the loss of Ras and Raf-1 activity, HDL induces MAPK phosphorylation in CHOanx6 cells. Since annexin A6 is a PKCalpha-binding protein we therefore investigated the possible involvement of PKC in HDL-induced Ras and MAPK activation in CHOanx6 cells. Taken together our findings demonstrate that HDL-induced H-Ras and MAPK activation is PKC-dependent in cells expressing annexin A6 to compensate for the loss of PKC-independent activation of H-Ras and MAPK.
Collapse
Affiliation(s)
- Carles Rentero
- Departament de Biologia Cellular, Facultat de Medicina, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain, and Centre for Immunology, St. Vincent's Hospital, Sydney, NSW, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Plowman SJ, Hancock JF. Ras signaling from plasma membrane and endomembrane microdomains. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2005; 1746:274-83. [PMID: 16039730 DOI: 10.1016/j.bbamcr.2005.06.004] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2005] [Revised: 05/30/2005] [Accepted: 06/10/2005] [Indexed: 11/29/2022]
Abstract
Ras proteins are compartmentalized by dynamic interactions with both plasma membrane microdomains and intracellular membranes. The mechanisms underlying Ras compartmentalization involve a series of protein/lipid, lipid/lipid and cytoskeleton interactions, resulting in the generation of discrete microdomains from which Ras operates. Segregation of Ras proteins to these different platforms regulates the formation of Ras signaling complexes and the generation of discrete signal outputs. This temporal and spatial modulation of Ras signal transduction provides a mechanism for the generation of different biological outcomes from different Ras isoforms, as well as flexibility in the signal output from a single activated isoform.
Collapse
Affiliation(s)
- S J Plowman
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, QLD 4072, Australia
| | | |
Collapse
|
42
|
Roberson MS, Bliss SP, Xie J, Navratil AM, Farmerie TA, Wolfe MW, Clay CM. Gonadotropin-releasing hormone induction of extracellular-signal regulated kinase is blocked by inhibition of calmodulin. Mol Endocrinol 2005; 19:2412-23. [PMID: 15890671 DOI: 10.1210/me.2005-0094] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Our previous studies demonstrate that GnRH-induced ERK activation required influx of extracellular Ca2+ in alphaT3-1 and rat pituitary cells. In the present studies, we examined the hypothesis that calmodulin (Cam) plays a fundamental role in mediating the effects of Ca2+ on ERK activation. Cam inhibition using W7 was sufficient to block GnRH-induced reporter gene activity for the c-Fos, murine glycoprotein hormone alpha-subunit, and MAPK phosphatase (MKP)-2 promoters, all shown to require ERK activation. Inhibition of Cam (using a dominant negative) was sufficient to block GnRH-induced ERK but not c-Jun N-terminal kinase activity activation. The Cam-dependent protein kinase (CamK) II inhibitor KN62 did not recapitulate these findings. GnRH-induced phosphorylation of MAPK/ERK kinase 1 and c-Raf kinase was blocked by Cam inhibition, whereas activity of phospholipase C was unaffected, suggesting that Ca2+/Cam modulation of the ERK cascade potentially at the level of c-Raf kinase. Enrichment of Cam-interacting proteins using a Cam agarose column revealed that c-Raf kinase forms a complex with Cam. Reconstitution studies reveal that recombinant c-Raf kinase can associate directly with Cam in a Ca2+-dependent manner and this interaction is reduced in vitro by addition of W7. Cam was localized in lipid rafts consistent with the formation of a Ca2+-sensitive signaling platform including the GnRH receptor and c-Raf kinase. These data support the conclusion that Cam may have a critical role as a Ca2+ sensor in specifically linking Ca2+ flux with ERK activation within the GnRH signaling pathway.
Collapse
Affiliation(s)
- Mark S Roberson
- Department of Biomedical Sciences, Cornell University, T3-004d Veterinary Research Tower, Ithaca, New York 14853, usa.
| | | | | | | | | | | | | |
Collapse
|
43
|
Hofer MD, Fecko A, Shen R, Setlur SR, Pienta KG, Tomlins SA, Chinnaiyan AM, Rubin MA. Expression of the platelet-derived growth factor receptor in prostate cancer and treatment implications with tyrosine kinase inhibitors. Neoplasia 2005; 6:503-12. [PMID: 15548358 PMCID: PMC1531653 DOI: 10.1593/neo.04157] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The platelet-derived growth factor receptor (PDGFR) is a receptor tyrosine kinase overexpressed in a subset of solid tumors and therefore is the target of drugs inhibiting this function such as imatinib mesylate (Gleevec). Thus far, drug therapy has played a limited role in the treatment of localized prostate cancer (PCa). This study characterizes PDGFR-beta expression in a wide spectrum of PCa samples to provide empirical data as part of a rational treatment strategy. A survey of five published prostate expression array studies, including 100 clinically localized PCa, did not identify tumors with increased PDGFR-beta expression level. Protein expression of PDGFR-beta, as determined by immunohistochemistry, revealed 5% of clinically localized PCa and 16% of metastatic PCa cases to show moderate or strong expression. To develop a strategy to detect patients most likely to profit from Gleevec treatment, we analyzed cDNA expression array data from 10,000 transcripts for PDGFR-beta expression and divided tumors in groups based on PDGFR-beta expression level. Performing a supervised analysis to identify potential comarkers of PDGFR-beta in PCa, we identified a set of genes whose expression was associated with PDGFR-beta status including early growth response 1 (Egr1), an upstream effector of PDGF (4.2-fold upregulation), alpha-methylacyl-CoA racemase, as well as v-Maf and neuroblastoma suppressor of tumorigenicity (both with a 2.2-fold downregulation). Taken together, this study suggests that only a small subset of PCas may be amenable to tyrosine kinase inhibitors specific for PDGFR.
Collapse
Affiliation(s)
- Matthias D Hofer
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Aspenström P. Integration of signalling pathways regulated by small GTPases and calcium. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2005; 1742:51-8. [PMID: 15590055 DOI: 10.1016/j.bbamcr.2004.09.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2004] [Revised: 09/21/2004] [Accepted: 09/24/2004] [Indexed: 10/26/2022]
Abstract
The Ras superfamily of small GTPases constitutes a large group of structurally and functionally related proteins. They function as signalling switches in numerous signalling cascades in the cell. During the recent years, an increased awareness of a communication between signalling systems employing Ras-like GTPases and signalling systems employing calcium has emerged. For instance, the intensity of the activation of Ras-like GTPases is regulated by calcium-dependent mechanisms, acting on proteins that facilitate the activation or inactivation of the small GTPases. Other Ras-like GTPases have a direct influence on calcium signalling by regulating the activity of certain calcium channels. In addition, several small GTPases collaborate with calcium signalling in regulating cellular processes, such as cell adhesion, cell migration and exocytosis.
Collapse
Affiliation(s)
- Pontus Aspenström
- Biomedical Center, Ludwig Institute for Cancer Research, Box 595, S-751 24 Uppsala, Sweden.
| |
Collapse
|
45
|
Williams CL. The polybasic region of Ras and Rho family small GTPases: a regulator of protein interactions and membrane association and a site of nuclear localization signal sequences. Cell Signal 2003; 15:1071-80. [PMID: 14575862 DOI: 10.1016/s0898-6568(03)00098-6] [Citation(s) in RCA: 164] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Many small GTPases in the Ras and Rho families have a C-terminal polybasic region (PBR) comprised of multiple lysines or arginines. The PBR controls diverse functions of these small GTPases, including their ability to associate with membranes, interact with specific proteins, and localize in subcellular compartments. Different signaling pathways mediated by Ras and Rho family members may converge when the small GTPases are directed by their PBRs to shared binding sites in specific proteins or at cell membranes. The PBR promotes the interactions of small GTPases with SmgGDS, which is a nucleocytoplasmic shuttling protein that stimulates guanine nucleotide exchange by small GTPases. The PBR of Rac1 was recently found to have a functional nuclear localization signal (NLS) sequence, which enhances the nuclear accumulation of protein complexes containing SmgGDS and Rac1. Sequence analysis demonstrates that canonical NLS sequences (K-K/R-x-K/R) are present in the PBRs of additional Ras and Rho family members, and are evolutionarily conserved across several phyla. These findings suggest that the PBR regulates the nucleocytoplasmic shuttling of some Ras and Rho family members when they are in protein complexes that are too large to diffuse through nuclear pores. These diverse functions of the PBR indicate its critical role in signaling by Ras and Rho family GTPases.
Collapse
Affiliation(s)
- Carol L Williams
- Molecular Pharmacology Laboratory, Guthrie Research Institute, One Guthrie Square, Sayre, PA 18840, USA.
| |
Collapse
|
46
|
Illario M, Cavallo AL, Bayer KU, Di Matola T, Fenzi G, Rossi G, Vitale M. Calcium/calmodulin-dependent protein kinase II binds to Raf-1 and modulates integrin-stimulated ERK activation. J Biol Chem 2003; 278:45101-8. [PMID: 12954639 DOI: 10.1074/jbc.m305355200] [Citation(s) in RCA: 126] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Integrin activation generates different signalings in a cell type-dependent manner and stimulates cell proliferation through the Ras/Raf-1/Mek/Erk pathway. In this study, we demonstrate that integrin stimulation by fibronectin (FN), besides activating the Ras/Erk pathway, generates an auxiliary calcium signal that activates calmodulin and the Ca2+/calmodulin-dependent protein kinase II (CaMKII). This signal regulates Raf-1 activation by Ras and modulates the FN-stimulated extracellular signal-regulated kinase (Erk-1/2). The binding of soluble FN to integrins induced increase of intracellular calcium concentration associated with phosphorylation and activation of CaMKII. In two different cell lines, inhibition of CaMKII activity by specific inhibitors inhibited Erk-1/2 phosphorylation. Whereas CaMK inhibition affected neither integrin-stimulated Akt phosphorylation nor p21Ras or Mek-1 activity, it was necessary for Raf-1 activity. FN-induced Raf-1 activity was abrogated by the CaMKII specific inhibitory peptide ant-CaNtide. Integrin activation by FN induced the formation of a Raf-1/CaMKII complex, abrogated by inhibition of CaMKII. Active CaMKII phosphorylated Raf-1 in vitro. This is the first demonstration that CaMKII interplays with Raf-1 and regulates Erk activation induced by Ras-stimulated Raf-1. These findings also provide evidence supporting the possible existence of cross-talk between other intracellular pathways involving CaMKII and Raf-1.
Collapse
Affiliation(s)
- Maddalena Illario
- Dipartimento di Biologia e Patologia Cellulare e Molecolare, Università Federico II, Napoli, 80131 Italy
| | | | | | | | | | | | | |
Collapse
|
47
|
Uyama N, Shimahara Y, Okuyama H, Kawada N, Kamo S, Ikeda K, Yamaoka Y. Carbenoxolone inhibits DNA synthesis and collagen gene expression in rat hepatic stellate cells in culture. J Hepatol 2003; 39:749-55. [PMID: 14568257 DOI: 10.1016/s0168-8278(03)00375-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND/AIMS This study using primary-cultured rat hepatic stellate cells (HSCs) was aimed to reveal the effect of carbenoxolone and the other gap-junction blockers on the proliferation and activation of HSCs. METHODS HSC morphology was microscopically evaluated. DNA synthesis was determined by [3H]thymidine incorporation. Expression of HSC activation markers and cell cycle-related proteins was evaluated by Western blot. Collagen alpha1(I) mRNA expression was evaluated by quantitative reverse transcription polymerase chain reaction. RESULTS Carbenoxolone triggered the morphological change of activated HSCs without inducing apoptosis. Culture-induced DNA synthesis was suppressed to 22.6 and 8.51%, respectively, by 40 and 80 microM carbenoxolone. The other gap-junction blockers failed to affect the morphology and the DNA synthesis of activated HSCs. Carbenoxolone decreased the expression of cyclins D1/2 and cyclin-dependent kinases 4/6. Platelet-derived growth factor (PDGF)-BB-elicited DNA synthesis was reduced to 45.6 and 3.27%, respectively, by 40 and 80 microM carbenoxolone. Phosphorylation of c-Raf, MEK and mitogen-activated protein kinase, but not PDGF receptor beta, under PDGF-BB stimulation was attenuated by carbenoxolone. Collagen alpha1(I) mRNA expression was significantly reduced. In addition, carbenoxolone suppressed the activation process of quiescent HSCs. CONCLUSIONS Carbenoxolone reduced the DNA synthesis and the expression of collagen alpha1(I) mRNA in activated HSCs independently of its pharmacological action as gap-junction blocker.
Collapse
Affiliation(s)
- Naoki Uyama
- Department of Gastroenterological Surgery, Graduate School of Medicine, Kyoto University, 54-Kawaracho, Shogoin, Sakyoku, Kyoto 606-8507, Japan
| | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Ras GTPases are binary switches, cycling between an inactive GDP-bound form and an active GTP-bound form at the membrane. They transduce signals into the cytoplasm via effector pathways that regulate cell growth, differentiation and apoptosis. Ras activation is enhanced by guanine nucleotide exchange factors (GEFs); deactivation is accelerated by GTPase-activating proteins (GAPs). Recently, new roles for Ca(2+) and diacylglycerol (DAG) in the control of Ras cycling have emerged with the discovery of a series of novel GEFs and GAPs. These regulators of Ras cycling are likely to play a key role in the information processing of Ca(2+) and DAG signals.
Collapse
Affiliation(s)
- Simon A Walker
- Inositide Group, Henry Wellcome Laboratories for Integrated Signalling, Department of Biochemistry, School of Medical Sciences, University of Bristol, Bristol BS8 1TD, UK
| | | | | | | |
Collapse
|
49
|
Sidhu RS, Clough RR, Bhullar RP. Ca2+/calmodulin binds and dissociates K-RasB from membrane. Biochem Biophys Res Commun 2003; 304:655-60. [PMID: 12727204 DOI: 10.1016/s0006-291x(03)00635-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We have investigated the interaction of calmodulin (CaM) with Ras-p21 and the significance of this association. All Ras-p21 isoforms tested (H-, K-, and N-Ras) were detected in the particulate fraction of human platelets and MCF-7 cells, a human breast cancer cell line. In MCF-7 cells, H- and N-Ras were also detected in the cytosolic fraction. K-RasB from platelet and MCF-7 cell lysates was found to bind CaM in a Ca2+ -dependent but GTPgammaS-independent manner. The yeast two-hybrid analysis demonstrated that K-RasB binds to CaM in vivo. Incubation of isolated membranes from platelet and MCF-7 cells with CaM caused dissociation of only K-RasB from membranes in a Ca2+ -dependent manner. CaM antagonist, W7, inhibited dissociation of K-RasB. Addition of platelet or MCF-7 cytosol alone to isolated platelet membranes did not cause dissociation of K-RasB and only addition of exogenous CaM caused dissociation. The results suggest a potential role for Ca2+/CaM in the regulation of K-RasB function.
Collapse
Affiliation(s)
- Ranjinder S Sidhu
- Department of Oral Biology, University of Manitoba, 780 Bannatyne Avenue, Winnipeg, Manitoba, Canada R3E 0W2
| | | | | |
Collapse
|
50
|
Abstract
Ras signalling has classically been thought to occur exclusively at the inner surface of a relatively uniform plasma membrane. Recent studies have shown that Ras proteins interact dynamically with specific microdomains of the plasma membrane as well as with other internal cell membranes. These different membrane microenvironments modulate Ras signal output and highlight the complex interplay between Ras location and function.
Collapse
Affiliation(s)
- John F Hancock
- Institute for Molecular Bioscience and Department of Molecular and Cellular Pathology, University of Queensland, Brisbane, Australia 4072.
| |
Collapse
|