Journal articles on the topic 'Ras-interacting protein'

To see the other types of publications on this topic, follow the link: Ras-interacting protein.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'Ras-interacting protein.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Kikuchi, A., S. D. Demo, Z. H. Ye, Y. W. Chen, and L. T. Williams. "ralGDS family members interact with the effector loop of ras p21." Molecular and Cellular Biology 14, no. 11 (November 1994): 7483–91. http://dx.doi.org/10.1128/mcb.14.11.7483-7491.1994.

Full text
Abstract:
Using a yeast two-hybrid system, we identified a novel protein which interacts with ras p21. This protein shares 69% amino acid homology with ral guanine nucleotide dissociation stimulator (ralGDS), a GDP/GTP exchange protein for ral p24. We designated this protein RGL, for ralGDS-like. Using the yeast two-hybrid system, we found that an effector loop mutant of ras p21 was defective in interacting with the ras p21-interacting domain of RGL, suggesting that this domain binds to ras p21 through the effector loop of ras p21. Since ralGDS contained a region highly homologous with the ras p21-interacting domain of RGL, we examined whether ralGDS could interact with ras p21. In the yeast two-hybrid system, ralGDS failed to interact with an effector loop mutant of ras p21. In insect cells, ralGDS made a complex with v-ras p21 but not with a dominant negative mutant of ras p21. ralGDS interacted with the GTP-bound form of ras p21 but not with the GDP-bound form in vitro. ralGDS inhibited both the GTPase-activating activity of the neurofibromatosis gene product (NF1) for ras p21 and the interaction of Raf with ras p21 in vitro. These results demonstrate that ralGDS specifically interacts with the active form of ras p21 and that ralGDS can compete with NF1 and Raf for binding to the effector loop of ras p21. Therefore, ralGDS family members may be effector proteins of ras p21 or may inhibit interactions between ras p21 and its effectors.
APA, Harvard, Vancouver, ISO, and other styles
2

Kikuchi, A., S. D. Demo, Z. H. Ye, Y. W. Chen, and L. T. Williams. "ralGDS family members interact with the effector loop of ras p21." Molecular and Cellular Biology 14, no. 11 (November 1994): 7483–91. http://dx.doi.org/10.1128/mcb.14.11.7483.

Full text
Abstract:
Using a yeast two-hybrid system, we identified a novel protein which interacts with ras p21. This protein shares 69% amino acid homology with ral guanine nucleotide dissociation stimulator (ralGDS), a GDP/GTP exchange protein for ral p24. We designated this protein RGL, for ralGDS-like. Using the yeast two-hybrid system, we found that an effector loop mutant of ras p21 was defective in interacting with the ras p21-interacting domain of RGL, suggesting that this domain binds to ras p21 through the effector loop of ras p21. Since ralGDS contained a region highly homologous with the ras p21-interacting domain of RGL, we examined whether ralGDS could interact with ras p21. In the yeast two-hybrid system, ralGDS failed to interact with an effector loop mutant of ras p21. In insect cells, ralGDS made a complex with v-ras p21 but not with a dominant negative mutant of ras p21. ralGDS interacted with the GTP-bound form of ras p21 but not with the GDP-bound form in vitro. ralGDS inhibited both the GTPase-activating activity of the neurofibromatosis gene product (NF1) for ras p21 and the interaction of Raf with ras p21 in vitro. These results demonstrate that ralGDS specifically interacts with the active form of ras p21 and that ralGDS can compete with NF1 and Raf for binding to the effector loop of ras p21. Therefore, ralGDS family members may be effector proteins of ras p21 or may inhibit interactions between ras p21 and its effectors.
APA, Harvard, Vancouver, ISO, and other styles
3

Marty, Caroline, Darren D. Browning, and Richard D. Ye. "Identification of Tetratricopeptide Repeat 1 as an Adaptor Protein That Interacts with Heterotrimeric G Proteins and the Small GTPase Ras." Molecular and Cellular Biology 23, no. 11 (June 1, 2003): 3847–58. http://dx.doi.org/10.1128/mcb.23.11.3847-3858.2003.

Full text
Abstract:
ABSTRACT The biological functions of heterotrimeric G proteins and small GTPases are modulated by both extracellular stimuli and intracellular regulatory proteins. Using Saccharomyces cerevisiae two-hybrid screening, we identified tetratricopeptide repeat 1 (TPR1), a 292-amino-acid protein with three TPR motifs, as a Gα16-binding protein. The interaction was confirmed both in vitro and in transfected mammalian cells, where TPR1 also binds to several other Gα proteins. TPR1 was found to interact with Ha-Ras preferentially in its active form. Overexpression of TPR1 promotes accumulation of active Ras. TPR1 was found to compete with the Ras-binding domain (RBD) of Raf-1 for binding to the active Ras, suggesting that it may also compete with Ras GTPase-activating protein, thus contributing to the accumulation of GTP-bound Ras. Expression of Gα16 strongly enhances the interaction between TPR1 and Ras. Removal of the TPR1 N-terminal 112 residues abolishes potentiation by Gα16 while maintaining the interaction with Gα16 and the ability to discriminate active Ras from wild-type Ras. We have also observed that LGN, a Gαi-interacting protein with seven TPR motifs, binds Ha-Ras. Thus, TPR1 is a novel adaptor protein for Ras and selected Gα proteins that may be involved in protein-protein interaction relating to G-protein signaling.
APA, Harvard, Vancouver, ISO, and other styles
4

Eccleston, Alex, Maria Jose Fernandez-Sarabia, and Frank McCormick. "CHARACTERISATION OF THE BCL-2 INTERACTING PROTEIN, R-RAS." Biochemical Society Transactions 24, no. 4 (November 1, 1996): 602S. http://dx.doi.org/10.1042/bst024602s.

Full text
APA, Harvard, Vancouver, ISO, and other styles
5

Pamonsinlapatham, Perayot, Réda Hadj-Slimane, Yves Lepelletier, Barbara Allain, Mirco Toccafondi, Christiane Garbay, and Françoise Raynaud. "P120-Ras GTPase activating protein (RasGAP): A multi-interacting protein in downstream signaling." Biochimie 91, no. 3 (March 2009): 320–28. http://dx.doi.org/10.1016/j.biochi.2008.10.010.

Full text
APA, Harvard, Vancouver, ISO, and other styles
6

Guo, Xiao-Xi, Su An, Yang Yang, Ying Liu, Qian Hao, and Tian-Rui Xu. "Rap-Interacting Proteins are Key Players in the Rap Symphony Orchestra." Cellular Physiology and Biochemistry 39, no. 1 (2016): 137–56. http://dx.doi.org/10.1159/000445612.

Full text
Abstract:
Rap, a member of the Ras-like small G-protein family, is a key node among G-protein coupled receptors (GPCR), receptor tyrosine kinases (RTKs), ion channels and many other downstream pathways. Rap plays a unique role in cell morphogenesis, adhesion, migration, exocytosis, proliferation, apoptosis and carcinogenesis. The complexity and diversity of Rap functions are tightly regulated by Rap-interacting proteins such as GEFs, GAPs, Rap effectors and scaffold proteins. These interacting proteins decide the subcellular localization of Rap, the interaction modes with downstream Rap effectors and tune Rap as an atypical molecular conductor, coupling extra- and intracellular signals to various pathways. In this review, we summarize four groups of Rap-interacting proteins, highlight their distinctions in Rap-binding properties and interactive modes and discuss their contribution to the spatiotemporal regulation of Rap as well as the implications of targeting Rap-interacting proteins in human cancer therapy.
APA, Harvard, Vancouver, ISO, and other styles
7

Ecker, Karin, Andreas Lorenz, Frank Wolf, Christian Ploner, Günther Böck, Tod Duncan, Stephan Geley, and Arno Helmberg. "A RAS recruitment screen identifies ZKSCAN4 as a glucocorticoid receptor-interacting protein." Journal of Molecular Endocrinology 42, no. 2 (November 13, 2008): 105–17. http://dx.doi.org/10.1677/jme-08-0087.

Full text
Abstract:
To search for proteins interacting with the glucocorticoid receptor, we adapted Aronheim's reverse RAS recruitment system relying on the Saccharomyces cerevisiae mutant cdc25-2, which has a temperature-dependent defect in its RAS signaling pathway driving proliferation. The full-length human glucocorticoid receptor (NR3C1, isoform-α) was attached to the yeast plasma membrane in either of two orientations and used as bait to screen a HeLa cell cDNA library. Library proteins were fused to constitutively active, soluble human RAS, complementing the defective yeast pathway in case of bait–prey interaction. Screening of 800 000 clones resulted in the isolation of 21 proteins, 8 of which were followed up to evaluate interaction with the receptor in human cell lines. One of these candidates, the SCAN- and KRAB-domain-containing zinc finger protein 307 (ZKSCAN4) was co-precipitated with the receptor when both proteins were overexpressed in HEK293 cells. Rabbit antisera against ZKSCAN4 were raised, affinity purified, and used to immunoprecipitate endogenous ZKSCAN4 from Hct116 cells, resulting in co-precipitation of endogenous glucocorticoid receptor. Overexpressed ZKSCAN4 was found to co-localize in granular nuclear structures with the activated glucocorticoid receptor and partially with chromatin regions characterized by histone H3 mono-methylated on lysine 4 (H3K4me1). Overexpressed ZKSCAN4 had no effect on an episomal glucocorticoid receptor-driven reporter plasmid. By contrast, ZKSCAN4 markedly reduced glucocorticoid induction of the mouse mammary tumor virus-promoter-driven reporter gene when this was chromosomally integrated, arguing for a chromatin-dependent inhibition of glucocorticoid receptor-mediated transactivation.
APA, Harvard, Vancouver, ISO, and other styles
8

Goldfinger, Lawrence E., Celeste Ptak, Erin D. Jeffery, Jeffrey Shabanowitz, Donald F. Hunt, and Mark H. Ginsberg. "RLIP76 (RalBP1) is an R-Ras effector that mediates adhesion-dependent Rac activation and cell migration." Journal of Cell Biology 174, no. 6 (September 11, 2006): 877–88. http://dx.doi.org/10.1083/jcb.200603111.

Full text
Abstract:
The Ras family of small GTPases regulates cell proliferation, spreading, migration and apoptosis, and malignant transformation by binding to several protein effectors. One such GTPase, R-Ras, plays distinct roles in each of these processes, but to date, identified R-Ras effectors were shared with other Ras family members (e.g., H-Ras). We utilized a new database of Ras-interacting proteins to identify RLIP76 (RalBP1) as a novel R-Ras effector. RLIP76 binds directly to R-Ras in a GTP-dependent manner, but does not physically associate with the closely related paralogues H-Ras and Rap1A. RLIP76 is required for adhesion-induced Rac activation and the resulting cell spreading and migration, as well as for the ability of R-Ras to enhance these functions. RLIP76 regulates Rac activity through the adhesion-induced activation of Arf6 GTPase and activation of Arf6 bypasses the requirement for RLIP76 in Rac activation and cell spreading. Thus, we identify a novel R-Ras effector, RLIP76, which links R-Ras to adhesion-induced Rac activation through a GTPase cascade that mediates cell spreading and migration.
APA, Harvard, Vancouver, ISO, and other styles
9

Miyasaka, Kyoko, Takahiro Fujimoto, Takako Kawanami, Soichi Takiguchi, Atsuo Jimi, Akihiro Funakoshi, and Senji Shirasawa. "Pancreatic Hypertrophy in Ki-ras-Induced Actin-Interacting Protein Gene Knockout Mice." Pancreas 40, no. 1 (January 2011): 79–83. http://dx.doi.org/10.1097/mpa.0b013e3181f66c22.

Full text
APA, Harvard, Vancouver, ISO, and other styles
10

Wang, Dakun, Zaibo Li, Edward M. Messing, and Guan Wu. "Activation of Ras/Erk Pathway by a Novel MET-interacting Protein RanBPM." Journal of Biological Chemistry 277, no. 39 (July 29, 2002): 36216–22. http://dx.doi.org/10.1074/jbc.m205111200.

Full text
APA, Harvard, Vancouver, ISO, and other styles
11

Horiuchi, Masatsugu, Jun Iwanami, and Masaki Mogi. "Regulation of angiotensin II receptors beyond the classical pathway." Clinical Science 123, no. 4 (April 20, 2012): 193–203. http://dx.doi.org/10.1042/cs20110677.

Full text
Abstract:
The RAS (renin–angiotensin system) plays a role not only in the cardiovascular system, including blood pressure regulation, but also in the central nervous system. AngII (angiotensin II) binds two major receptors: the AT1 receptor (AngII type 1 receptor) and AT2 receptor (AngII type 2 receptor). It has been recognized that AT2 receptor activation not only opposes AT1 receptor actions, but also has unique effects beyond inhibitory cross-talk with AT1 receptor signalling. Novel pathways beyond the classical actions of RAS, the ACE (angiotensin-converting enzyme)/AngII/AT1 receptor axis, have been highlighted: the ACE2/Ang-(1–7) [angiotensin-(1–7)]/Mas receptor axis as a new opposing axis against the ACE/AngII/AT1 receptor axis, novel AngII-receptor-interacting proteins and various AngII-receptor-activation mechanisms including dimer formation. ATRAP (AT1-receptor-associated protein) and ATIP (AT2-receptor-interacting protein) are well-characterized AngII-receptor-associated proteins. These proteins could regulate the functions of AngII receptors and thereby influence various pathophysiological states. Moreover, the possible cross-talk between PPAR (peroxisome-proliferator-activated receptor)-γ and AngII receptor subtypes is an intriguing issue to be addressed in order to understand the roles of RAS in the metabolic syndrome, and interestingly some ARBs (AT1-receptor blockers) have been reported to have an AT1-receptor-blocking action with a partial PPAR-γ agonistic effect. These emerging concepts concerning the regulation of AngII receptors are discussed in the present review.
APA, Harvard, Vancouver, ISO, and other styles
12

Zhao, Mingming, Justyna A. Janas, Masaru Niki, Pier Paolo Pandolfi, and Linda Van Aelst. "Dok-1 Independently Attenuates Ras/Mitogen-Activated Protein Kinase and Src/c-Myc Pathways To Inhibit Platelet-Derived Growth Factor-Induced Mitogenesis." Molecular and Cellular Biology 26, no. 7 (April 1, 2006): 2479–89. http://dx.doi.org/10.1128/mcb.26.7.2479-2489.2006.

Full text
Abstract:
ABSTRACT The Dok adaptor proteins play key regulatory roles in receptor and non-receptor kinase-initiated signaling pathways. Dok-1, the prototype member of this family, negatively regulates cell proliferation elicited by numerous growth factors, including platelet-derived growth factor (PDGF). However, how Dok-1 exerts its negative effect on mitogenesis has remained elusive. Using Dok-1 knockout cells and Dok-1 mutants deficient in binding to specific Dok-1-interacting proteins, we show that Dok-1 interferes with PDGF-stimulated c-myc induction and Ras/mitogen-activated protein kinase (MAPK) activation by tethering different signaling components to the cell membrane. Specifically, Dok-1 attenuates PDGF-elicited c-myc induction by recruiting Csk to active Src kinases, whereupon their activities and consequent c-myc induction are diminished. On the other hand, Dok-1 negatively regulates PDGF-induced MAPK activation by acting on Ras-GAP and at least one other Dok-1-interacting protein. Importantly, we demonstrate that Dok-1's actions on both of these signaling pathways contribute to its inhibitory effect on mitogenesis. Our data suggest a mechanistic basis for the inhibitory effect of Dok-1 on growth factor-induced mitogenesis and its role as a tumor suppressor.
APA, Harvard, Vancouver, ISO, and other styles
13

Bhattacharya, M., A. V. Babwah, and S. S. G. Ferguson. "Small GTP-binding protein-coupled receptors." Biochemical Society Transactions 32, no. 6 (October 26, 2004): 1040–44. http://dx.doi.org/10.1042/bst0321040.

Full text
Abstract:
Heterotrimeric GPCRs (G-protein-coupled receptors) form the largest group of integral membrane receptor proteins and mediate diverse physiological processes. In addition to signalling via heterotrimeric G-proteins, GPCRs can also signal by interacting with various small G-proteins to regulate downstream effector pathways. The small G-protein superfamily is structurally classified into at least five families: the Ras, Rho/Rac/cdc42, Rab, Sar1/Arf and Ran families. They are monomeric G-proteins with molecular masses over the range 20–30 kDa, which function as molecular switches to control many eukaryotic cell functions. Several studies have provided evidence of crosstalk between GPCRs and small G-proteins. It is well documented that GPCR signalling through heterotrimeric G-proteins can lead to the activation of Ras and Rho GTPases. In addition, RhoA, Rabs, ARFs and ARF GEFs (guanine nucleotide-exchange factors) can associate directly with GPCRs, and GPCRs may also function as GEFs for small GTPases. In this review, we summarize the recent progress made in understanding the interaction between GPCRs and small GTPases, focusing on understanding how the association of small G-proteins with GPCRs and GPCR-regulatory proteins may influence GPCR signalling and intracellular trafficking.
APA, Harvard, Vancouver, ISO, and other styles
14

Radziwill, G., R. A. Erdmann, U. Margelisch, and K. Moelling. "The Bcr Kinase Downregulates Ras Signaling by Phosphorylating AF-6 and Binding to Its PDZ Domain." Molecular and Cellular Biology 23, no. 13 (July 1, 2003): 4663–72. http://dx.doi.org/10.1128/mcb.23.13.4663-4672.2003.

Full text
Abstract:
ABSTRACT The protein kinase Bcr is a negative regulator of cell proliferation and oncogenic transformation. We identified Bcr as a ligand for the PDZ domain of the cell junction and Ras-interacting protein AF-6. The Bcr kinase phosphorylates AF-6, which subsequently allows efficient binding of Bcr to AF-6, showing that the Bcr kinase is a regulator of the PDZ domain-ligand interaction. Bcr and AF-6 colocalize in epithelial cells at the plasma membrane. In addition, Bcr, AF-6, and Ras form a trimeric complex. Bcr increases the affinity of AF-6 to Ras, and a mutant of AF-6 that lacks a specific phosphorylation site for Bcr shows a reduced binding to Ras. Wild-type Bcr, but not Bcr mutants defective in binding to AF-6, interferes with the Ras-dependent stimulation of the Raf/MEK/ERK pathway. Since AF-6 binds to Bcr via its PDZ domain and to Ras via its Ras-binding domain, we propose that AF-6 functions as a scaffold-like protein that links Bcr and Ras to cellular junctions. We suggest that this trimeric complex is involved in downregulation of Ras-mediated signaling at sites of cell-cell contact to maintain cells in a nonproliferating state.
APA, Harvard, Vancouver, ISO, and other styles
15

ARESTA, Sandra, Marie-France de TAND-HEIM, Florence BÉRANGER, and Jean de GUNZBURG. "A novel Rho GTPase-activating-protein interacts with Gem, a member of the Ras superfamily of GTPases." Biochemical Journal 367, no. 1 (October 1, 2002): 57–65. http://dx.doi.org/10.1042/bj20020829.

Full text
Abstract:
Gem is a Ras-related protein whose expression is induced in several cell types upon activation by extracellular stimuli. With the aim of isolating the cellular partners of Gem that mediate its biological activity we performed a yeast two-hybrid screen and identified a novel protein of 970 amino acids, Gmip, that interacts with Gem through its N-terminal half, and presents a cysteine-rich domain followed by a Rho GTPase-activating protein (RhoGAP) domain in its C-terminal half. The RhoGAP domain of Gmip stimulates in vitro the GTPase activity of RhoA, but is inactive towards other Rho family proteins such as Rac1 and Cdc42; it is also specific for RhoA in vivo. The same is true for the full-length protein, which is furthermore able to down-regulate RhoA-dependent stress fibres in Ref-52 rat fibroblasts. These findings suggest that the signalling pathways controlled by two proteins of the Ras superfamily, RhoA and Gem, are linked via the action of the RhoGAP protein Gmip (Gem-interacting protein).
APA, Harvard, Vancouver, ISO, and other styles
16

Fürstenau, U., M. Schwaninger, R. Blume, I. Kennerknecht, and W. Knepel. "Characterization of a novel protein kinase C response element in the glucagon gene." Molecular and Cellular Biology 17, no. 4 (April 1997): 1805–16. http://dx.doi.org/10.1128/mcb.17.4.1805.

Full text
Abstract:
To maintain glucose levels in blood within narrow limits, the synthesis and secretion of pancreatic islet hormones are controlled by a variety of neural, hormonal, and metabolic messengers that act through multiple signal transduction pathways. Glucagon gene transcription is stimulated by cyclic AMP and depolarization-induced calcium influx. In this study, the effect of protein kinase C on glucagon gene transcription was investigated. After transient transfection of a glucagon-reporter fusion gene into the glucagon-producing islet cell line alphaTC2, activation of protein kinase C by 12-O-tetradecanoylphorbol-13-acetate (TPA) stimulated glucagon gene transcription. By 5' deletions, 3' deletions, internal deletion, and oligonucleotide cassette insertion, the TPA-responsive element was mapped to the G2 element (from -165 to -200). Like TPA, overexpression of oncogenic Ras (V-12 Ras) stimulated G2-mediated transcription whereas overexpression of a dominant negative Ras mutant (N-17 Ras) blocked the effect of TPA. A mutational analysis of G2 function and nuclear protein binding indicated that protein kinase C and Ras responsiveness is conferred to the glucagon gene by HNF-3beta functionally interacting with a protein that binds to a closely associated site with sequence similarity to binding sites of Ets family proteins. HNF-3beta belongs to the winged-helix family of transcription factors and has been implicated in the control of cell-specific and developmental gene expression. The results of the present study show that the cell lineage-specific transcription factor HNF-3beta is an essential component of a novel protein kinase C response element in the glucagon gene.
APA, Harvard, Vancouver, ISO, and other styles
17

Athanasopoulos, Panagiotis S., Wright Jacob, Sebastian Neumann, Miriam Kutsch, Dirk Wolters, Eng K. Tan, Zoë Bichler, Christian Herrmann, and Rolf Heumann. "Identification of protein phosphatase 2A as an interacting protein of leucine-rich repeat kinase 2." Biological Chemistry 397, no. 6 (June 1, 2016): 541–54. http://dx.doi.org/10.1515/hsz-2015-0189.

Full text
Abstract:
Abstract Mutations in the gene coding for the multi-domain protein leucine-rich repeat kinase 2 (LRRK2) are the leading cause of genetically inherited Parkinson’s disease (PD). Two of the common found mutations are the R1441C and G2019S. In this study we identified protein phosphatase 2A (PP2A) as an interacting partner of LRRK2. We were able to demonstrate that the Ras of complex protein (ROC) domain is sufficient to interact with the three subunits of PP2A in human neuroblastoma SH-SY5Y cells and in HeLa cells. The alpha subunit of PP2A is interacting with LRRK2 in the perinuclear region of HeLa cells. Silencing the catalytic subunit of PP2A by shRNA aggravated cellular degeneration induced by the pathogenic R1441C-LRRK2 mutant expressed in neuroblastoma SH-SY5Y cells. A similar enhancement of apoptotic nuclei was observed by downregulation of the catalytic subunit of PP2A in cultured cortical cells derived from neurons overexpressing the pathogenic mutant G2019S-LRRK2. Conversely, pharmacological activation of PP2A by sodium selenate showed a partial neuroprotection from R1441C-LRRK2-induced cellular degeneration. All these data suggest that PP2A is a new interacting partner of LRRK2 and reveal the importance of PP2A as a potential therapeutic target in PD.
APA, Harvard, Vancouver, ISO, and other styles
18

Krishnadev, Oruganty, Shveta Bisht, and Narayanaswamy Srinivasan. "Prediction of Protein-Protein Interactions Between Human Host and Two Mycobacterial Organisms." International Journal of Knowledge Discovery in Bioinformatics 1, no. 1 (January 2010): 1–13. http://dx.doi.org/10.4018/jkdb.2010100201.

Full text
Abstract:
The genomes of many human pathogens have been sequenced but the protein-protein interactions across a pathogen and human are still poorly understood. The authors apply a simple homology-based method to predict protein-protein interactions between human host and two mycobacterial organisms viz., M.tuberculosis and M.leprae. They focused on secreted proteins of pathogens and cellular membrane proteins to restrict to uncovering biologically significant and feasible interactions. Predicted interactions include five mycobacterial proteins of yet unknown function, thus suggesting a role for these proteins in pathogenesis. The authors predict interaction partners for secreted mycobacterial antigens such as MPT70, serine proteases and other proteins interacting with human proteins, such as toll-like receptors, ras signalling proteins and immune maintenance proteins, that are implicated in pathogenesis. These results suggest that the list of predicted interactions is suitable for further analysis and forms a useful step in the understanding of pathogenesis of these mycobacterial organisms.
APA, Harvard, Vancouver, ISO, and other styles
19

Aronheim, A., E. Zandi, H. Hennemann, S. J. Elledge, and M. Karin. "Isolation of an AP-1 repressor by a novel method for detecting protein-protein interactions." Molecular and Cellular Biology 17, no. 6 (June 1997): 3094–102. http://dx.doi.org/10.1128/mcb.17.6.3094.

Full text
Abstract:
Transcription factor AP-1 transduces environmental signals to the transcriptional machinery. To ensure a quick response yet maintain tight control over AP-1 target genes, AP-1 activity is likely to be negatively regulated in nonstimulated cells. To identify proteins that interact with the Jun subunits of AP-1 and repress its activity, we developed a novel screen for detecting protein-protein interactions that is not based on a transcriptional readout. In this system, the mammalian guanyl nucleotide exchange factor (GEF) Sos is recruited to the Saccharomyces cerevisiae plasma membrane harboring a temperature-sensitive Ras GEF, Cdc25-2, allowing growth at the nonpermissive temperature. Using the Sos recruitment system, we identified new c-Jun-interacting proteins. One of these, JDP2, heterodimerizes with c-Jun in nonstimulated cells and represses AP-1-mediated activation.
APA, Harvard, Vancouver, ISO, and other styles
20

Ordentlich, Peter, Arthur Lin, Chun-Pyn Shen, Chris Blaumueller, Kenji Matsuno, Spyros Artavanis-Tsakonas, and Tom Kadesch. "Notch Inhibition of E47 Supports the Existence of a Novel Signaling Pathway." Molecular and Cellular Biology 18, no. 4 (April 1, 1998): 2230–39. http://dx.doi.org/10.1128/mcb.18.4.2230.

Full text
Abstract:
ABSTRACT E47 is a widely expressed transcription factor that activates B-cell-specific immunoglobulin gene transcription and is required for early B-cell development. In an effort to identify processes that regulate E47, and potentially B-cell development, we found that activated Notch1 and Notch2 effectively inhibit E47 activity. Only the intact E47 protein was inhibited by Notch—fusion proteins containing isolated DNA binding and activation domains were unaffected—suggesting that Notch targets an atypical E47 cofactor. Although overexpression of the coactivator p300 partially reversed E47 inhibition, results of several assays indicated that p300/CBP is not a general target of Notch. Notch inhibition of E47 did not correlate with its ability to activate CBF1/RBP-Jκ, the mammalian homolog of Suppressor of Hairless, a protein that associates physically with Notch and defines the only known Notch signaling pathway in drosophila. Importantly, E47 was inhibited independently of CBF1/RPB-Jκ by Deltex, a second Notch-interacting protein. We provide evidence that Notch and Deltex may act on E47 by inhibiting signaling through Ras because (i) full E47 activity was found to be dependent on Ras and (ii) both Notch and Deltex inhibited GAL4-Jun, a hybrid transcription factor whose activity is dependent on signaling from Ras to SAPK/JNK.
APA, Harvard, Vancouver, ISO, and other styles
21

Lee, Susan, Carole A. Parent, Robert Insall, and Richard A. Firtel. "A Novel Ras-interacting Protein Required for Chemotaxis and Cyclic Adenosine Monophosphate Signal Relay inDictyostelium." Molecular Biology of the Cell 10, no. 9 (September 1999): 2829–45. http://dx.doi.org/10.1091/mbc.10.9.2829.

Full text
Abstract:
We have identified a novel Ras-interacting protein fromDictyostelium, RIP3, whose function is required for both chemotaxis and the synthesis and relay of the cyclic AMP (cAMP) chemoattractant signal. rip3 null cells are unable to aggregate and lack receptor activation of adenylyl cyclase but are able, in response to cAMP, to induce aggregation-stage, postaggregative, and cell-type-specific gene expression in suspension culture. In addition, rip3 null cells are unable to properly polarize in a cAMP gradient and chemotaxis is highly impaired. We demonstrate that cAMP stimulation of guanylyl cyclase, which is required for chemotaxis, is reduced ∼60% in rip3 null cells. This reduced activation of guanylyl cyclase may account, in part, for the defect in chemotaxis. When cells are pulsed with cAMP for 5 h to mimic the endogenous cAMP oscillations that occur in wild-type strains, the cells will form aggregates, most of which, however, arrest at the mound stage. Unlike the response seen in wild-type strains, the rip3 null cell aggregates that form under these experimental conditions are very small, which is probably due to the rip3 null cell chemotaxis defect. Many of the phenotypes of the rip3 null cell, including the inability to activate adenylyl cyclase in response to cAMP and defects in chemotaxis, are very similar to those of strains carrying a disruption of the gene encoding the putative Ras exchange factor AleA. We demonstrate that aleA null cells also exhibit a defect in cAMP-mediated activation of guanylyl cyclase similar to that of rip3 null cells. A double-knockout mutant (rip3/aleA null cells) exhibits a further reduction in receptor activation of guanylyl cyclase, and these cells display almost no cell polarization or movement in cAMP gradients. As RIP3 preferentially interacts with an activated form of theDictyostelium Ras protein RasG, which itself is important for cell movement, we propose that RIP3 and AleA are components of a Ras-regulated pathway involved in integrating chemotaxis and signal relay pathways that are essential for aggregation.
APA, Harvard, Vancouver, ISO, and other styles
22

Cao, Haoyuan, Jiandong Zhang, and Wei Wang. "DAB2IP Plays Important Clinical Significance and Correlates With Immune Infiltration in Renal Cell Carcinoma." Technology in Cancer Research & Treatment 19 (January 1, 2020): 153303382093668. http://dx.doi.org/10.1177/1533033820936682.

Full text
Abstract:
Background: Disabled homolog 2-interacting protein is a new member of the Ras GTPase superfamily involved in the regulation of cell proliferation, apoptosis, and metastasis. However, the expression of disabled homolog 2-interacting protein in renal cell carcinoma, its correlation with cancer prognosis, and tumor infiltrating lymphocytes remains unclear. Methods: The expression of disabled homolog 2-interacting protein was analyzed by UALCAN database, GEPIA database and the evaluation of disabled homolog 2-interacting protein effects on clinical prognosis. Prognostic factor analysis was used to identify the correlations between disabled homolog 2-interacting protein and cancer immune infiltration via the TIMER database. In addition, COXPRESdb database was used to analyze the enrichment of disabled homolog 2-interacting protein co-expression genes. Results: Compared to the normal tissues, the messenger RNA expression levels of DAB2IP are higher in 8 while lower in 15 types of tumor tissues. Furthermore, disabled homolog 2-interacting protein has high expression in kidney chromophobe and low expression in both kidney renal clear cell carcinoma and kidney renal papillary cell carcinoma. The messenger RNA expression levels of disabled homolog 2-interacting protein decrease gradually due to the increasing tumor staging which positively correlates with disease-free survival and overall survival in both kidney renal clear cell carcinoma and kidney renal papillary cell carcinoma. The expression levels of disabled homolog 2-interacting protein also positively correlate with the tumor purity of kidney chromophobe, kidney renal clear cell carcinoma, and kidney renal papillary cell carcinoma samples. Besides, the expression of disabled homolog 2-interacting protein in renal cell carcinoma has negative correlation with the immune infiltration, and the immune infiltration of B cells and CD8+ T cells affects the prognosis of kidney renal papillary cell carcinoma. Enrichment analysis of disabled homolog 2-interacting protein co-expressed genes suggested that its biological role was mainly in regulating GTPase activity. Conclusions: These findings suggest that disabled homolog 2-interacting protein functions as a tumor suppressor in the progression of renal cell carcinoma, and the expression of disabled homolog 2-interacting protein is related to the immune infiltrating cells and affects the survival of renal cell carcinoma. Disabled homolog 2-interacting protein can be a novel clinical biomarker for patients with renal cell carcinoma, which also provides new insights for the future treatments of renal cell carcinoma.
APA, Harvard, Vancouver, ISO, and other styles
23

Kelly, Paul A., and Zohra Rahmani. "DYRK1A Enhances the Mitogen-activated Protein Kinase Cascade in PC12 Cells by Forming a Complex with Ras, B-Raf, and MEK1." Molecular Biology of the Cell 16, no. 8 (August 2005): 3562–73. http://dx.doi.org/10.1091/mbc.e04-12-1085.

Full text
Abstract:
Dual-specificity tyrosine-phosphorylated and regulated kinase 1A (Dyrk1A) is the human homologue of the Drosophila mnb (minibrain) gene. In Drosophila, mnb is involved in postembryonic neurogenesis. In human, DYRK1A maps within the Down syndrome critical region of chromosome 21 and is overexpressed in Down syndrome embryonic brain. Despite its potential involvement in the neurobiological alterations observed in Down syndrome patients, the biological functions of the serine/threonine kinase DYRK1A have not been identified yet. Here, we report that DYRK1A overexpression potentiates nerve growth factor (NGF)-mediated PC12 neuronal differentiation by up-regulating the Ras/MAP kinase signaling pathway independently of its kinase activity. Furthermore, we show that DYRK1A prolongs the kinetics of ERK activation by interacting with Ras, B-Raf, and MEK1 to facilitate the formation of a Ras/B-Raf/MEK1 multiprotein complex. These data indicate that DYRK1A may play a critical role in Ras-dependent transducing signals that are required for promoting or maintaining neuronal differentiation and suggest that overexpression of DYRK1A may contribute to the neurological abnormalities observed in Down syndrome patients.
APA, Harvard, Vancouver, ISO, and other styles
24

Khoza, Thembisile, Ian Dubery, and Lizelle Piater. "Identification of Candidate Ergosterol-Responsive Proteins Associated with the Plasma Membrane of Arabidopsis thaliana." International Journal of Molecular Sciences 20, no. 6 (March 14, 2019): 1302. http://dx.doi.org/10.3390/ijms20061302.

Full text
Abstract:
The impact of fungal diseases on crop production negatively reflects on sustainable food production and overall economic health. Ergosterol is the major sterol component in fungal membranes and regarded as a general elicitor or microbe-associated molecular pattern (MAMP) molecule. Although plant responses to ergosterol have been reported, the perception mechanism is still unknown. Here, Arabidopsis thaliana protein fractions were used to identify those differentially regulated following ergosterol treatment; additionally, they were subjected to affinity-based chromatography enrichment strategies to capture and categorize ergosterol-interacting candidate proteins using liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS). Mature plants were treated with 250 nM ergosterol over a 24 h period, and plasma membrane-associated fractions were isolated. In addition, ergosterol was immobilized on two different affinity-based systems to capture interacting proteins/complexes. This resulted in the identification of defense-related proteins such as chitin elicitor receptor kinase (CERK), non-race specific disease resistance/harpin-induced (NDR1/HIN1)-like protein, Ras-related proteins, aquaporins, remorin protein, leucine-rich repeat (LRR)- receptor like kinases (RLKs), G-type lectin S-receptor-like serine/threonine-protein kinase (GsSRK), and glycosylphosphatidylinositol (GPI)-anchored protein. Furthermore, the results elucidated unknown signaling responses to this MAMP, including endocytosis, and other similarities to those previously reported for bacterial flagellin, lipopolysaccharides, and fungal chitin.
APA, Harvard, Vancouver, ISO, and other styles
25

Pitoniak, Andrew, Colin A. Chavel, Jacky Chow, Jeremy Smith, Diawoye Camara, Sheelarani Karunanithi, Boyang Li, Kennith H. Wolfe, and Paul J. Cullen. "Cdc42p-Interacting Protein Bem4p Regulates the Filamentous-Growth Mitogen-Activated Protein Kinase Pathway." Molecular and Cellular Biology 35, no. 2 (November 10, 2014): 417–36. http://dx.doi.org/10.1128/mcb.00850-14.

Full text
Abstract:
The ubiquitous Rho (Ras homology) GTPase Cdc42p can function in different settings to regulate cell polarity and cellular signaling. How Cdc42p and other proteins are directed to function in a particular context remains unclear. We show that the Cdc42p-interacting protein Bem4p regulates the mitogen-activated protein kinase (MAPK) pathway that controls filamentous growth inSaccharomyces cerevisiae. Bem4p controlled the filamentous-growth pathway but not other MAPK pathways (mating or high-osmolarity glycerol response [HOG]) that also require Cdc42p and other shared components. Bem4p associated with the plasma membrane (PM) protein, Sho1p, to regulate MAPK activity and cell polarization under nutrient-limiting conditions that favor filamentous growth. Bem4p also interacted with the major activator of Cdc42p, the guanine nucleotide exchange factor (GEF) Cdc24p, which we show also regulates the filamentous-growth pathway. Bem4p interacted with the pleckstrin homology (PH) domain of Cdc24p, which functions in an autoinhibitory capacity, and was required, along with other pathway regulators, to maintain Cdc24p at polarized sites during filamentous growth. Bem4p also interacted with the MAPK kinase kinase (MAPKKK) Ste11p. Thus, Bem4p is a new regulator of the filamentous-growth MAPK pathway and binds to general proteins, like Cdc42p and Ste11p, to promote a pathway-specific response.
APA, Harvard, Vancouver, ISO, and other styles
26

Mitin, Natalia Y., Melissa B. Ramocki, Alfred J. Zullo, Channing J. Der, Stephen F. Konieczny, and Elizabeth J. Taparowsky. "Identification and Characterization of Rain, a Novel Ras-interacting Protein with a Unique Subcellular Localization." Journal of Biological Chemistry 279, no. 21 (March 18, 2004): 22353–61. http://dx.doi.org/10.1074/jbc.m312867200.

Full text
APA, Harvard, Vancouver, ISO, and other styles
27

Elumalai, Elakkiya, and Suresh Kumar Muthuvel. "Network Analysis of Dengue NS1 Interacting Core Human Proteins Driving Dengue Pathogenesis." Current Chemical Biology 15, no. 4 (December 2021): 287–300. http://dx.doi.org/10.2174/2212796816666211216115753.

Full text
Abstract:
Aim: We aimed to identify critical human proteins involved in cathepsin L regulation Background: It has been shown that Dengue Virus (DENV) NS1 activates cathepsin L (CTSL). The CTSL activates heparanase, which cleaves heparan sulfate proteoglycans and causes dengue pathogenesis. NS1 directly interacts with PTBP1 and Gab proteins. Gab protein activates the Ras signaling pathway. Still, no known direct interaction partners are linking GAB1 to cathepsin L. Objective: Our objective includes three main points.1-Network analysis of NS1 interacting human proteins 2- Identification of protein-drug and protein-disease interactions 3- Identification of core proteins involved in cathepsin L regulation. Method: We collected NS1 interacting Human proteins from DenHunt, Int-Act Molecular Interaction Database, Virus Mentha, Virus Pathogen Database and Analysis Resource (ViPR), and Virus MINT. We employed Pesca, cytohubba, and centiscape as the significant plug-ins in Cytoscape for network analysis. To study protein-diseases and protein-drugs interaction, we used NetworkAnalyst. Result: Based on the prior knowledge on the interaction of NS1 with GAB1 and PTBP1 human proteins, we found several core proteins that drive dengue pathogenesis. The proteins EED, NXF1, and MOV10, are the mediators between PTBP1 and CTSL. Similarly, DNM2, GRB2, PXN, PTPRC, and NTRK1 mediate GAB1 and PTBP1. The common first neighbors of MOV10, NXF1, and EED were identified, and the common primary pathways in all three subnetworks were mRNA processing and protein translation. The common interaction partners were considered for drug and disease network analysis. These proteins were; PARP1, NFKB2, HDAC2, SLC25A4, ATP5A1, EPN1, CTSL, UBR4, CLK3, and ARPC4. PARP1 was the highly connected node in the protein-drug network. The highest degree protein, LMNA, was associated with many diseases. The NXF1 is connected with LMNA. Here, we reported one essential protein, namely, NXF1 protein, which links PTBP1 with CTSL. The NXF1 is also connected with TPM3, which is connected to CTSL. Conclusion: We listed functionally important proteins which are involved in cathepsin L activation. Based on network properties, we proposed, NXF1 and TPM3 are the important high centrality proteins in dengue infection.
APA, Harvard, Vancouver, ISO, and other styles
28

Kozminski, Keith G., Ann J. Chen, Avital A. Rodal, and David G. Drubin. "Functions and Functional Domains of the GTPase Cdc42p." Molecular Biology of the Cell 11, no. 1 (January 2000): 339–54. http://dx.doi.org/10.1091/mbc.11.1.339.

Full text
Abstract:
Cdc42p, a Rho family GTPase of the Ras superfamily, is a key regulator of cell polarity and morphogenesis in eukaryotes. Using 37 site-directed cdc42 mutants, we explored the functions and interactions of Cdc42p in the budding yeast Saccharomyces cerevisiae. Cytological and genetic analyses of thesecdc42 mutants revealed novel and diverse phenotypes, showing that Cdc42p possesses at least two distinct essential functions and acts as a nodal point of cell polarity regulation in vivo. In addition, mapping the functional data for each cdc42mutation onto a structural model of the protein revealed as functionally important a surface of Cdc42p that is distinct from the canonical protein-interacting domains (switch I, switch II, and the C terminus) identified previously in members of the Ras superfamily. This region overlaps with a region (α5-helix) recently predicted by structural models to be a specificity determinant for Cdc42p-protein interactions.
APA, Harvard, Vancouver, ISO, and other styles
29

Cao, Huijuan, Hao Gong, Tianqiao Song, Mina Yu, Xiayan Pan, Junjie Yu, Zhongqiang Qi, Yan Du, and Yongfeng Liu. "The Adaptor Protein UvSte50 Governs Fungal Pathogenicity of Ustilaginoidea Virens via the MAPK Signaling Pathway." Journal of Fungi 8, no. 9 (September 11, 2022): 954. http://dx.doi.org/10.3390/jof8090954.

Full text
Abstract:
The mitogen-activated protein kinase (MAPK) signaling pathways regulate diverse cellular processes and have been partially characterized in the rice false smut fungus Ustilaginoidea virens. UvSte50 has been identified as a homolog to Saccharomyces cerevisiae Ste50, which is known to be an adaptor protein for MAPK cascades. ΔUvste50 was found to be defective in conidiation, sensitive to hyperosmotic and oxidative stresses, and non-pathogenic. The mycelial expansion of ΔUvste50 inside spikelets of rice terminated at stamen filaments, eventually resulting in a lack of formation of false smut balls on spikelets. We determined that UvSte50 directly interacts with both UvSte7 (MAPK kinase; MEK) and UvSte11 (MAPK kinase kinase; MEKK), where the Ras-association (RA) domain of UvSte50 is indispensable for its interaction with UvSte7. UvSte50 also interacts with UvHog1, a MAP kinase of the Hog1-MAPK pathway, which is known to have important roles in hyphal growth and stress responses in U. virens. In addition, affinity capture–mass spectrometry analysis and yeast two-hybrid assay were conducted, through which we identified the interactions of UvSte50 with UvRas2, UvAc1 (adenylate cyclase), and UvCap1 (cyclase-associated protein), key components of the Ras/cAMP signaling pathway in U. virens. Together, UvSte50 functions as an adaptor protein interacting with multiple components of the MAPK and Ras/cAMP signaling pathways, thus playing critical role in plant infection by U. virens.
APA, Harvard, Vancouver, ISO, and other styles
30

Mott, Helen R., and Darerca Owen. "Structure and function of RLIP76 (RalBP1): an intersection point between Ras and Rho signalling." Biochemical Society Transactions 42, no. 1 (January 23, 2014): 52–58. http://dx.doi.org/10.1042/bst20130231.

Full text
Abstract:
RLIP76 (Ral-interacting protein of 76 kDa) [also known as RalBP1 (Ral-binding protein 1)] is an effector for the Ral family small GTPases. RLIP76 has been implicated in a number of cell processes, including receptor-mediated endocytosis, cell migration, mitochondrial division and metabolite transport. RLIP76 has two recognizable domains in the centre of the protein sequence: a GAP (GTPase-activating protein) domain for the Rho family G-proteins and an RBD (Ral-binding domain). The remainder of RLIP76 has no discernable homology with other proteins. The RBD forms a simple coiled-coil of two α-helices, which interacts with RalB by binding to both of the nucleotide-sensitive ‘switch’ regions. Both of these RLIP76 helices are involved in the interaction with Ral, but the interhelix loop is left free. This is the location of one of the two ATP-binding sites that have been identified in RLIP76 and suggests that Ral interaction would not prevent ATP binding. The structure of the RhoGAP–RBD dyad shows that the two domains are fixed in their orientation by a relatively rigid linker. This domain arrangement allows the two domains to engage Rho family and Ral small G-proteins simultaneously at the membrane. This suggests that RLIP76 is a node for Rho and Ras family signalling.
APA, Harvard, Vancouver, ISO, and other styles
31

Kim, Ji-Eun, Duk-Shin Lee, Tae-Hyun Kim, and Tae-Cheon Kang. "CDDO-Me Attenuates CA1 Neuronal Death by Facilitating RalBP1-Mediated Mitochondrial Fission and 4-HNE Efflux in the Rat Hippocampus Following Status Epilepticus." Antioxidants 11, no. 5 (May 18, 2022): 985. http://dx.doi.org/10.3390/antiox11050985.

Full text
Abstract:
Ras-related protein Ral-A (RalA)-binding protein 1 (RalBP1, also known as Ral-interacting protein of 76 kDa (RLIP76) or Ral-interacting protein 1 (RLIP1 or RIP1)) is involved in the efflux of 4-hydroxynonenal (4-HNE, an end product of lipid peroxidation), as well as mitochondrial fission. In the present study, we found that 2-cyano-3,12-dioxo-oleana-1,9(11)-dien-28-oic acid methyl ester (CDDO-Me) attenuated CA1 neuronal death and aberrant mitochondrial elongations in these neurons coupled with enhanced RalBP1 expression and reduced 4-HNE levels following status epilepticus (SE). RalBP1 knockdown did not affect mitochondrial dynamics and CA1 neuronal death under physiological and post-SE conditions. Following SE, however, cotreatment of RalBP1 siRNA diminished the effect of CDDO-Me on 4-HNE levels, mitochondrial hyperfusion in CA1 neurons, and CA1 neuronal death. These findings indicate that CDDO-Me may ameliorate CA1 neuronal death by facilitating RalBP1-mediated 4-HNE efflux and mitochondrial fission following SE. Therefore, our findings suggest that increased RalBP1 expression/activity may be one of the considerable targets to protect neurons from SE.
APA, Harvard, Vancouver, ISO, and other styles
32

Becker, C. F. W., C. L. Hunter, R. Seidel, S. B. H. Kent, R. S. Goody, and M. Engelhard. "Total chemical synthesis of a functional interacting protein pair: The protooncogene H-Ras and the Ras-binding domain of its effector c-Raf1." Proceedings of the National Academy of Sciences 100, no. 9 (April 18, 2003): 5075–80. http://dx.doi.org/10.1073/pnas.0831227100.

Full text
APA, Harvard, Vancouver, ISO, and other styles
33

Carmena, Ana, Stephan Speicher, and Mary Baylies. "The PDZ Protein Canoe/AF-6 Links Ras-MAPK, Notch and Wingless/Wnt Signaling Pathways by Directly Interacting with Ras, Notch and Dishevelled." PLoS ONE 1, no. 1 (December 20, 2006): e66. http://dx.doi.org/10.1371/journal.pone.0000066.

Full text
APA, Harvard, Vancouver, ISO, and other styles
34

Zhang, Tiantian, Soyoung Choi, Tuo Zhang, Zhengming Chen, Yudan Chi, Shixia Huang, Jenny Z. Xiang, and Yi-Chieh Nancy Du. "miR-431 Promotes Metastasis of Pancreatic Neuroendocrine Tumors by Targeting DAB2 Interacting Protein, a Ras GTPase Activating Protein Tumor Suppressor." American Journal of Pathology 190, no. 3 (March 2020): 689–701. http://dx.doi.org/10.1016/j.ajpath.2019.11.007.

Full text
APA, Harvard, Vancouver, ISO, and other styles
35

Niemela, Julie E., Lianghao Lu, Thomas A. Fleisher, Joie Davis, Iusta Caminha, Marc Natter, Laurel A. Beer, et al. "Somatic KRAS mutations associated with a human nonmalignant syndrome of autoimmunity and abnormal leukocyte homeostasis." Blood 117, no. 10 (March 10, 2011): 2883–86. http://dx.doi.org/10.1182/blood-2010-07-295501.

Full text
Abstract:
Abstract Somatic gain-of-function mutations in members of the RAS subfamily of small guanosine triphosphatases are found in > 30% of all human cancers. We recently described a syndrome of chronic nonmalignant lymphadenopathy, splenomegaly, and autoimmunity associated with a mutation in NRAS affecting hematopoietic cells, and initially we classified the disease as a variant of the autoimmune lymphoproliferative syndrome. Here, we demonstrate that somatic mutations in the related KRAS gene can also be associated with a nonmalignant syndrome of autoimmunity and breakdown of leukocyte homeostasis. The activating KRAS mutation impaired cytokine withdrawal–induced T-cell apoptosis through the suppression of the proapoptotic protein BCL-2 interacting mediator of cell death and facilitated proliferation through p27kip1 down-regulation. These defects could be corrected in vitro by mitogen-activated protein kinase/extracellular signal-regulated kinase kinase 1 or phosphatidyl inositol-3 kinase inhibition. We suggest the use of the term RAS-associated autoimmune leukoproliferative disease to differentiate this disorder from autoimmune lymphoproliferative syndrome.
APA, Harvard, Vancouver, ISO, and other styles
36

Roth Flach, Rachel J., Chang-An Guo, Laura V. Danai, Joseph C. Yawe, Sharvari Gujja, Yvonne J. K. Edwards, and Michael P. Czech. "Endothelial Mitogen-Activated Protein Kinase Kinase Kinase Kinase 4 Is Critical for Lymphatic Vascular Development and Function." Molecular and Cellular Biology 36, no. 12 (April 4, 2016): 1740–49. http://dx.doi.org/10.1128/mcb.01121-15.

Full text
Abstract:
The molecular mechanisms underlying lymphatic vascular development and function are not well understood. Recent studies have suggested a role for endothelial cell (EC) mitogen-activated protein kinase kinase kinase kinase 4 (Map4k4) in developmental angiogenesis and atherosclerosis. Here, we show that constitutive loss of EC Map4k4 in mice causes postnatal lethality due to chylothorax, suggesting that Map4k4 is required for normal lymphatic vascular function. Mice constitutively lacking EC Map4k4 displayed dilated lymphatic capillaries, insufficient lymphatic valves, and impaired lymphatic flow; furthermore, primary ECs derived from these animals displayed enhanced proliferation compared with controls. Yeast 2-hybrid analyses identified the Ras GTPase-activating protein Rasa1, a known regulator of lymphatic development and lymphatic endothelial cell fate, as a direct interacting partner for Map4k4. Map4k4 silencing in ECs enhanced basal Ras and extracellular signal-regulated kinase (Erk) activities, and primary ECs lacking Map4k4 displayed enhanced lymphatic EC marker expression. Taken together, these results reveal that EC Map4k4 is critical for lymphatic vascular development by regulating EC quiescence and lymphatic EC fate.
APA, Harvard, Vancouver, ISO, and other styles
37

Predicala, Rey, and Yan Zhou. "The role of Ran-binding protein 3 during influenza A virus replication." Journal of General Virology 94, no. 5 (May 1, 2013): 977–84. http://dx.doi.org/10.1099/vir.0.049395-0.

Full text
Abstract:
Influenza A virus vRNP nuclear export is CRM1-dependent. Ran-binding protein 3 (RanBP3) is a Ran-interacting protein that is best known for its role as a cofactor of CRM1-mediated cargo nuclear export. In this study, we investigated the role of RanBP3 during the influenza A virus life cycle. We found that RanBP3 was phosphorylated at Ser58 in the early and late phases of infection. Knockdown of RanBP3 expression led to vRNP nuclear retention, suggesting that RanBP3 is involved in vRNP nuclear export. Moreover, we demonstrated that the function of RanBP3 during vRNP nuclear export is regulated by phosphorylation at Ser58, and that RanBP3 phosphorylation is modulated by both PI3K/Akt and Ras/ERK/RSK pathways in the late phase of viral infection.
APA, Harvard, Vancouver, ISO, and other styles
38

D'Arcangelo, G., R. Habas, S. Wang, S. Halegoua, and S. R. Salton. "Activation of codependent transcription factors is required for transcriptional induction of the vgf gene by nerve growth factor and Ras." Molecular and Cellular Biology 16, no. 9 (September 1996): 4621–31. http://dx.doi.org/10.1128/mcb.16.9.4621.

Full text
Abstract:
Nerve growth factor (NGF) treatment of PC12 cells leads to the elaboration of a neuronal phenotype, including the induction of neuronally expressed genes such as vgf. To study vgf transcription, we have created chimeric vgf/beta-globin genes in which vgf promoter sequences drive the expression of the beta-globin reporter gene or of a chimeric beta-globin gene fused to 3' untranslated vgf gene sequences. We have found that the level of inducibility of the latter construct by NGF resembles that of the endogenous vgf gene. Using transient transfection of the chimeric reporter genes into PC12 cells, into PC12 subclones expressing activated or dominantly interfering mutant Ras proteins, and into PC12 variants expressing specific NGF receptor/Trk mutants, we show that transcriptional regulation of the vgf promoter by NGF is mediated through a Ras-dependent signaling pathway. By mutational analysis of the vgf promoter, we have identified three promoter elements involved in mediating transcriptional induction by NGF and Ras. In addition to the cyclic AMP-responsive element (CRE), which binds to ATF-1, ATF-2, and CRE-binding protein in PC12 nuclear extracts, a novel CCAAT element and its binding proteins were identified, which, like the CRE, is necessary but not sufficient for the Ras-dependent induction of the vgf gene by NGF. We also identify a G(S)G element unusually located between the TATA box and transcriptional start site, which binds the NGF- and Ras-induced transcription factor, NGFI-A, and amplifies the transcriptional response. Integrating data from studies of vgf promoter regulation and NGF signal transduction, we present a model for vgf gene induction in which transcriptional activation is achieved through the persistent, direct activation of multiple interacting transcription factors binding to CRE and CCAAT elements, coordinated with the delayed transcription factor action at a G(S)G element resulting from the induced expression of NGFI-A.
APA, Harvard, Vancouver, ISO, and other styles
39

Pham, Nam, and Daniela Rotin. "Nedd4 Regulates Ubiquitination and Stability of the Guanine-Nucleotide Exchange Factor CNrasGEF." Journal of Biological Chemistry 276, no. 50 (October 11, 2001): 46995–7003. http://dx.doi.org/10.1074/jbc.m108373200.

Full text
Abstract:
Cyclic nucleotide ras GEF (CNrasGEF) is a guanine-nucleotide exchange factor previously isolated in a screen for Nedd4-WW domain interacting proteins (Pham, N., Cheglakov, I., Koch, C. A., de Hoog, C. L., Moran, M. F., and Rotin, D. (2000)Curr. Biol.10, 555–558). It activates Ras in a cAMP-dependent manner and Rap-1 independent of cAMP. Here we show that CNrasGEF is a likely substrate of the ubiquitin protein ligase Nedd4. CNrasGEF possesses two PY motifs at its C terminus that are responsible for binding to Nedd4in vitro. Moreover, Nedd4 and CNrasGEF co-immunoprecipitate from 293T cells expressing ectopic CNrasGEF and endogenous Nedd4, and this co-immunoprecipitation is abrogated in PY motif-mutated CNrasGEF (CNrasGEFΔ2PY). CNrasGEF is ubiquitinated in cells, and this ubiquitination is augmented upon overexpression of wt-Nedd4 but is inhibited in cells overexpressing a catalytically inactive Nedd4 (Nedd4(CS)) or in cells expressing CNrasGEFΔ2PY, which cannot bind Nedd4. Moreover, pulse-chase experiments have demonstrated that the half-life of CNrasGEF is reduced 5-fold (from ∼10 to ∼2 h) in cells co-expressing Nedd4 with CNrasGEF but not with CNrasGEFΔ2PY (t0.5∼ 14 h). CNrasGEF is also stabilized in cells co-expressing Nedd4(CS) or following treatment with lactacystin, indicating proteasomal degradation of this protein. Deletion/mutation of the CDC25 domain to abrogate Ras (or Rap-1) binding leads to impaired ubiquitination of CNrasGEF, suggesting that such binding is critical for ubiquitination. Treatment of cells with the cAMP analogue 8-bromo-cAMP does not affect the ability of CNrasGEF to bind Nedd4 nor its level of ubiquitination, suggesting that Ras bindingper seand not its activation is the critical step in triggering ubiquitination of CNrasGEF. These results suggest that CNrasGEF is a substrate for Nedd4, which regulates its ubiquitination and stability in cells.
APA, Harvard, Vancouver, ISO, and other styles
40

Hu, Lingli, Lulu Li, Chen Yan, Yuxue Cao, Xiaohong Duan, and Jing Sun. "Baicalin Inhibits Airway Smooth Muscle Cells Proliferation through the RAS Signaling Pathway in Murine Asthmatic Airway Remodeling Model." Oxidative Medicine and Cellular Longevity 2023 (February 13, 2023): 1–12. http://dx.doi.org/10.1155/2023/4144138.

Full text
Abstract:
Background. Studies that looked at asthma airway remodeling pathogenesis and prevention have led to the discovery of the rat sarcoma viral oncogene (RAS) signaling pathway as a key mechanism that controls airway smooth muscle cell (ASMC) proliferation. Baicalin has great anti-inflammatory, proliferation-inhibited, and respiratory disease-relieving properties. However, the inhibitory effects and mechanisms of baicalin on ASMC-mediated airway remodeling in mice are still poorly understood. Methods. After establishing the asthmatic mice model by ovalbumin (OVA) and interfering with baicalin, airway remodeling characteristics such as airway resistance, mRNA, and protein expression levels of remodeling-related cytokines were measured by histopathological assessment, quantitative real-time polymerase chain reaction (qPCR), enzyme-linked immunosorbent assay (ELISA), and western blot. Further efforts on detailed mechanisms were used antibody arrays to compare the expression and activation of proteins involved in the RAS signaling pathway. In addition, validation experiments were performed in ASMC proliferation model and low-expression cells of the target gene by using shRNA. Results. In OVA-induced asthmatic mice model, baicalin significantly reduced the infiltration of inflammatory cells in lung tissue, attenuated airway resistance, and decreased mRNA and protein expression levels of remodeling-related cytokines such as interleukin-13 (IL-13), vascular endothelial growth factor (VEGF), transforming growth factor-beta 1 (TGF-β1), matrix metallopeptidase 9 (MMP9), and tissue inhibitor of metalloproteinase 1 (TIMP1). The results of antibody arrays involved in RAS signaling pathway revealed that OVA and baicalin administration altered the activation of protein kinase C alpha type (PKC-α), A-rapidly accelerated fibrosarcoma (A-RAF), mitogen-activated protein kinase 2 (MEK2), extracellular regulated MAP kinase (ERK), MAPK interacting serine/threonine kinase 1 (MNK1), and ETS transcription factor 1 (ELK1). The above results were further verified in the ASMC proliferation model. A-RAF silencing (shA-RAF) could promote ASMC proliferation and downregulate p-MEK2, p-ERK, p-MNK1, and p-ELK1 expression. Conclusion. The effects of baicalin against airway remodeling and ASMC proliferation might partially be achieved by suppressing the RAS signaling pathway. Baicalin may be a new therapeutic option for managing airway remodeling in asthma patients.
APA, Harvard, Vancouver, ISO, and other styles
41

Rocchetti, Maria Teresa, Grazia Tamma, Domenica Lasorsa, Ida Valentina Suriano, Annamaria D'Apollo, Massimo Papale, Lisa Mastrofrancesco, et al. "Altered urinary excretion of aquaporin 2 in IgA nephropathy." European Journal of Endocrinology 165, no. 4 (October 2011): 657–64. http://dx.doi.org/10.1530/eje-11-0512.

Full text
Abstract:
ObjectiveThe intrarenal renin–angiotensin system (RAS) activation plays a pivotal role in immunoglobulin A nephropathy (IgAN) pathogenesis, which is still largely undefined. Recently, vasopressin (AVP) has been advocated to contribute to the genesis and progression of chronic kidney diseases (CKD) directly, and indirectly, via RAS activation. Our aim is to explore the intrarenal activity of AVP, its relationship with RAS activity, as well as its modulation by therapies in IgAN.DesignIn this observational study, we measured plasma copeptin, a surrogate marker of AVP, the urine excretion of aquaporin 2 (AQP2), a protein reflecting renal AVP action, and angiotensinogen (AGT), a parameter of renal RAS activation, and their relationship with renal function in 44 IgAN patients at the time of renal biopsy, without any drug therapy, and after 6-month treatment with ACEi or steroid+ACEi. Twenty-one patients with other CKD and 40 healthy subjects were recruited as controls.MethodsELISAs were used to measure all variables of interest.ResultsAt baseline, IgAN patients showed higher urinary levels of AQP2, compared with controls and patients with other CKD. Urinary AQP2 and AGT levels strongly correlated with the presence of arterial hypertension. Steroids+ACEi caused the decrease of all the variables examined. The fall of urinary AQP2 and AGT following drug treatments was associated with the decrease of daily proteinuria.ConclusionOur findings would support the involvement of AVP–AQP2 axis, interacting with the RAS, in the progression of IgAN and candidate AQP2 as a possible novel marker of the disease.
APA, Harvard, Vancouver, ISO, and other styles
42

Dar, Khalid Bashir, Aashiq Hussain Bhat, Shajrul Amin, Syed Anjum, Bilal Ahmad Reshi, Mohammad Afzal Zargar, Akbar Masood, and Showkat Ahmad Ganie. "Exploring Proteomic Drug Targets, Therapeutic Strategies and Protein - Protein Interactions in Cancer: Mechanistic View." Current Cancer Drug Targets 19, no. 6 (June 21, 2019): 430–48. http://dx.doi.org/10.2174/1568009618666180803104631.

Full text
Abstract:
Protein-Protein Interactions (PPIs) drive major signalling cascades and play critical role in cell proliferation, apoptosis, angiogenesis and trafficking. Deregulated PPIs are implicated in multiple malignancies and represent the critical targets for treating cancer. Herein, we discuss the key protein-protein interacting domains implicated in cancer notably PDZ, SH2, SH3, LIM, PTB, SAM and PH. These domains are present in numerous enzymes/kinases, growth factors, transcription factors, adaptor proteins, receptors and scaffolding proteins and thus represent essential sites for targeting cancer. This review explores the candidature of various proteins involved in cellular trafficking (small GTPases, molecular motors, matrix-degrading enzymes, integrin), transcription (p53, cMyc), signalling (membrane receptor proteins), angiogenesis (VEGFs) and apoptosis (BCL-2family), which could possibly serve as targets for developing effective anti-cancer regimen. Interactions between Ras/Raf; X-linked inhibitor of apoptosis protein (XIAP)/second mitochondria-derived activator of caspases (Smac/DIABLO); Frizzled (FRZ)/Dishevelled (DVL) protein; beta-catenin/T Cell Factor (TCF) have also been studied as prospective anticancer targets. Efficacy of diverse molecules/ drugs targeting such PPIs although evaluated in various animal models/cell lines, there is an essential need for human-based clinical trials. Therapeutic strategies like the use of biologicals, high throughput screening (HTS) and fragment-based technology could play an imperative role in designing cancer therapeutics. Moreover, bioinformatic/computational strategies based on genome sequence, protein sequence/structure and domain data could serve as competent tools for predicting PPIs. Exploring hot spots in proteomic networks represents another approach for developing targetspecific therapeutics. Overall, this review lays emphasis on a productive amalgamation of proteomics, genomics, biochemistry, and molecular dynamics for successful treatment of cancer.
APA, Harvard, Vancouver, ISO, and other styles
43

Guo, Xiaoli, Ginny G. Farías, Rafael Mattera, and Juan S. Bonifacino. "Rab5 and its effector FHF contribute to neuronal polarity through dynein-dependent retrieval of somatodendritic proteins from the axon." Proceedings of the National Academy of Sciences 113, no. 36 (August 24, 2016): E5318—E5327. http://dx.doi.org/10.1073/pnas.1601844113.

Full text
Abstract:
An open question in cell biology is how the general intracellular transport machinery is adapted to perform specialized functions in polarized cells such as neurons. Here we illustrate this adaptation by elucidating a role for the ubiquitous small GTPase Ras-related protein in brain 5 (Rab5) in neuronal polarity. We show that inactivation or depletion of Rab5 in rat hippocampal neurons abrogates the somatodendritic polarity of the transferrin receptor and several glutamate receptor types, resulting in their appearance in the axon. This loss of polarity is not caused primarily by increased transport from the soma to the axon but rather by decreased retrieval from the axon to the soma. Retrieval is also dependent on the Rab5 effector Fused Toes (FTS)–Hook–FTS and Hook-interacting protein (FHIP) (FHF) complex, which interacts with the minus-end–directed microtubule motor dynein and its activator dynactin to drive a population of axonal retrograde carriers containing somatodendritic proteins toward the soma. These findings emphasize the importance of both biosynthetic sorting and axonal retrieval for the polarized distribution of somatodendritic receptors at steady state.
APA, Harvard, Vancouver, ISO, and other styles
44

Stang, Espen, Frøydis D. Blystad, Maja Kazazic, Vibeke Bertelsen, Tonje Brodahl, Camilla Raiborg, Harald Stenmark, and Inger Helene Madshus. "Cbl-dependent Ubiquitination Is Required for Progression of EGF Receptors into Clathrin-coated Pits." Molecular Biology of the Cell 15, no. 8 (August 2004): 3591–604. http://dx.doi.org/10.1091/mbc.e04-01-0041.

Full text
Abstract:
Ligand binding causes the EGF receptor (EGFR) to become ubiquitinated by Cbl upon association with the adaptor protein Grb2. We have investigated the role of ubiquitin and Grb2 in ligand-induced endocytosis of the EGFR. Incubation of cells with EGF on ice caused translocation of Grb2 and Cbl from the cytosol to the rim of coated pits. Grb2 with point mutations in both SH3 domains inhibited recruitment of the EGFR to clathrin-coated pits, in a Ras-independent manner. On overexpression of the Cbl-binding protein Sprouty, ubiquitination of the EGFR was inhibited, the EGFR was recruited only to the rim of coated pits, and endocytosis of the EGFR was inhibited. Conjugation-defective ubiquitin similarly inhibited recruitment of EGF-EGFR to clathrin-coated pits. Even though this does not prove that cargo must be ubiquitinated, this indicates the importance of interaction of ubiquitinated protein(s) with proteins harboring ubiquitin-interacting domains. We propose that Grb2 mediates transient anchoring of the EGFR to an Eps15-containing molecular complex at the rim of coated pits and that Cbl-induced ubiquitination of the EGFR allows relocation of EGFR from the rim to the center of clathrin-coated pits.
APA, Harvard, Vancouver, ISO, and other styles
45

Sanchez, Pilar, Guillermo De Carcer, Ignacio V. Sandoval, Jorge Moscat, and María T. Diaz-Meco. "Localization of Atypical Protein Kinase C Isoforms into Lysosome-Targeted Endosomes through Interaction with p62." Molecular and Cellular Biology 18, no. 5 (May 1, 1998): 3069–80. http://dx.doi.org/10.1128/mcb.18.5.3069.

Full text
Abstract:
ABSTRACT An increasing number of independent studies indicate that the atypical protein kinase C (PKC) isoforms (aPKCs) are critically involved in the control of cell proliferation and survival. The aPKCs are targets of important lipid mediators such as ceramide and the products of the PI 3-kinase. In addition, the aPKCs have been shown to interact with Ras and with two novel proteins, LIP (lambda-interacting protein; a selective activator of λ/ιPKC) and the product ofpar-4 (a gene induced during apoptosis), which is an inhibitor of both λ/ιPKC and ζPKC. LIP and Par-4 interact with the zinc finger domain of the aPKCs where the lipid mediators have been shown to bind. Here we report the identification of p62, a previously described phosphotyrosine-independent p56 lck SH2-interacting protein, as a molecule that interacts potently with the V1 domain of λ/ιPKC and, albeit with lower affinity, with ζPKC. We also show in this study that ectopically expressed p62 colocalizes perfectly with both λ/ιPKC and ζPKC. Interestingly, the endogenous p62, like the ectopically expressed protein, displays a punctate vesicular pattern and clearly colocalizes with endogenous λ/ιPKC and endogenous ζPKC. P62 colocalizes with Rab7 and partially with lamp-1 and limp-II as well as with the epidermal growth factor (EGF) receptor in activated cells, but not with Rab5 or the transferrin receptor. Of functional relevance, expression of dominant negative λ/ιPKC, but not of the wild-type enzyme, severely impairs the endocytic membrane transport of the EGF receptor with no effect on the transferrin receptor. These findings strongly suggest that the aPKCs are anchored by p62 in the lysosome-targeted endosomal compartment, which seems critical for the control of the growth factor receptor trafficking. This is particularly relevant in light of the role played by the aPKCs in mitogenic cell signaling events.
APA, Harvard, Vancouver, ISO, and other styles
46

Taya, Shinichiro, Takaharu Yamamoto, Kyoko Kano, Yoji Kawano, Akihiro Iwamatsu, Tomoko Tsuchiya, Keiji Tanaka, et al. "The Ras Target AF-6 is a Substrate of the Fam Deubiquitinating Enzyme." Journal of Cell Biology 142, no. 4 (August 24, 1998): 1053–62. http://dx.doi.org/10.1083/jcb.142.4.1053.

Full text
Abstract:
The Ras target AF-6 has been shown to serve as one of the peripheral components of cell–cell adhesions, and is thought to participate in cell–cell adhesion regulation downstream of Ras. We here purified an AF-6-interacting protein with a molecular mass of ∼220 kD (p220) to investigate the function of AF-6 at cell–cell adhesions. The peptide sequences of p220 were identical to the amino acid sequences of mouse Fam. Fam is homologous to a deubiquitinating enzyme in Drosophila, the product of the fat facets gene. Recent genetic analyses indicate that the deubiquitinating activity of the fat facets product plays a critical role in controlling the cell fate. We found that Fam accumulated at the cell–cell contact sites of MDCKII cells, but not at free ends of plasma membranes. Fam was partially colocalized with AF-6 and interacted with AF-6 in vivo and in vitro. We also showed that AF-6 was ubiquitinated in intact cells, and that Fam prevented the ubiquitination of AF-6.
APA, Harvard, Vancouver, ISO, and other styles
47

Li, Zheng, Li Li, Haifeng Zhang, Huanjiao Jenny Zhou, Weidong Ji, and Wang Min. "Short AIP1 (ASK1-Interacting Protein-1) Isoform Localizes to the Mitochondria and Promotes Vascular Dysfunction." Arteriosclerosis, Thrombosis, and Vascular Biology 40, no. 1 (January 2020): 112–27. http://dx.doi.org/10.1161/atvbaha.119.312976.

Full text
Abstract:
Objective: Vascular endothelial cells (ECs) normally maintain vascular homeostasis and are regulated by proinflammatory cytokines and reactive oxygen species. A human genome-wide association study identified that AIP1 (ASK1 [apoptosis signal-regulating kinase 1]-interacting protein-1; also identified as DAB2IP ) gene variants confer susceptibility to cardiovascular disease, but the underlying mechanism is unknown. Approach and Results: We detected a normal AIP1 form (named AIP1A) in the healthy aorta, but a shorter form of AIP1 (named AIP1B) was found in diseased aortae that contained atherosclerotic plaques and graft arteriosclerosis. AIP1B transcription in resting ECs was suppressed through epigenetic inhibition by RIF1 (Rap1 [ras-related protein 1]-interacting factor 1)/H3K9 (histone H3 lysine 9) methyltransferase-mediated H3K9 trimethylation, and this inhibition was released by proinflammatory cytokines. AIP1A, but not AIP1B, was downregulated by proteolytic degradation through a Smurf1 (SMAD [suppressor of mothers against decapentaplegic miscellaneous] ubiquitylation regulatory factor 1)-dependent pathway in ECs under inflammation. Therefore, AIP1B was the major form present during inflammatory conditions. AIP1B, which lacks the N-terminal pleckstrin homology domain of AIP1A, localized to the mitochondria and augmented TNFα (tumor necrosis factor alpha)-induced mitochondrial reactive oxygen species generation and EC activation. AIP1B-ECTG (EC-specific AIP1B transgenic) mice exhibited augmented reactive oxygen species production, EC activation, and neointima formation in vascular remodeling models. Conclusions: Our current study suggests that a shift from anti-inflammatory AIP1A to proinflammatory AIP1B during chronic inflammation plays a key role in inflammatory vascular diseases.
APA, Harvard, Vancouver, ISO, and other styles
48

Rudenko, Iakov N., Alice Kaganovich, Rebekah G. Langston, Aleksandra Beilina, Kelechi Ndukwe, Ravindran Kumaran, Allissa A. Dillman, Ruth Chia, and Mark R. Cookson. "The G2385R risk factor for Parkinson's disease enhances CHIP-dependent intracellular degradation of LRRK2." Biochemical Journal 474, no. 9 (April 24, 2017): 1547–58. http://dx.doi.org/10.1042/bcj20160909.

Full text
Abstract:
Autosomal dominant mutations in leucine-rich repeat kinase 2 (LRRK2) are associated with Parkinson's disease (PD). Most pathogenic LRRK2 mutations result in amino acid substitutions in the central ROC (Ras of complex proteins)–C-terminus of ROC–kinase triple domain and affect enzymatic functions of the protein. However, there are several variants in LRRK2, including the risk factor G2385R, that affect PD pathogenesis by unknown mechanisms. Previously, we have shown that G2385R LRRK2 has decreased kinase activity in vitro and altered affinity to LRRK2 interactors. Specifically, we found an increased binding to the chaperone Hsp90 (heat shock protein 90 kDa) that is known to stabilize LRRK2, suggesting that G2385R may have structural effects on LRRK2. In the present study, we further explored the effects of G2385R on LRRK2 in cells. We found that G2385R LRRK2 has lower steady-state intracellular protein levels compared with wild-type LRRK2 due to increased protein turnover of the mutant protein. Mechanistically, this is a consequence of a higher affinity of G2385R compared with the wild-type protein for two proteins involved in proteasomal degradation, Hsc70 and carboxyl-terminus of Hsc70-interacting protein (CHIP). Overexpression of CHIP decreased intracellular protein levels of both G2385R mutant and wild-type LRRK2, while short interfering RNA CHIP knockdown had the opposite effect. We suggest that the G2385R substitution tilts the equilibrium between refolding and proteasomal degradation toward intracellular degradation. The observation of lower steady-state protein levels may explain why G2385R is a risk factor rather than a penetrant variant for inherited PD.
APA, Harvard, Vancouver, ISO, and other styles
49

Miller, Kristi E., Wing-Cheong Lo, Ching-Shan Chou, and Hay-Oak Park. "Temporal regulation of cell polarity via the interaction of the Ras GTPase Rsr1 and the scaffold protein Bem1." Molecular Biology of the Cell 30, no. 20 (September 15, 2019): 2543–57. http://dx.doi.org/10.1091/mbc.e19-02-0106.

Full text
Abstract:
The Cdc42 guanosine triphosphatase (GTPase) plays a central role in polarity development in species ranging from yeast to humans. In budding yeast, a specific growth site is selected in the G1 phase. Rsr1, a Ras GTPase, interacts with Cdc42 and its associated proteins to promote polarized growth at the proper bud site. Yet how Rsr1 regulates cell polarization is not fully understood. Here, we show that Rsr1-GDP interacts with the scaffold protein Bem1 in early G1, likely hindering the role of Bem1 in Cdc42 polarization and polarized secretion. Consistent with these in vivo observations, mathematical modeling predicts that Bem1 is unable to promote Cdc42 polarization in early G1 in the presence of Rsr1-GDP. We find that a part of the Bem1 Phox homology domain, which overlaps with a region interacting with the exocyst component Exo70, is necessary for the association of Bem1 with Rsr1-GDP. Overexpression of the GDP-locked Rsr1 interferes with Bem1-dependent Exo70 polarization. We thus propose that Rsr1 functions in spatial and temporal regulation of polarity establishment by associating with distinct polarity factors in its GTP- and GDP-bound states.
APA, Harvard, Vancouver, ISO, and other styles
50

Sugiyama, Takayuki, Donna P. Frazier, Pankaj Taneja, Robert D. Kendig, Rachel L. Morgan, Lauren A. Matise, Sarah J. Lagedrost, and Kazushi Inoue. "Signal Transduction Involving the Dmp1 Transcription Factor and its Alteration in Human Cancer." Clinical medicine. Oncology 2 (January 2008): CMO.S548. http://dx.doi.org/10.4137/cmo.s548.

Full text
Abstract:
Summary Dmp1 (cyclin D-interacting myb-like protein 1; also called Dmtf1) is a transcription factor that has been isolated in a yeast two-hybrid screen through its binding property to cyclin D2. Dmp1 directly binds to and activates the Arf promoter and induces Arf-p53-dependent cell cycle arrest in primary cells. D-type cyclins usually inhibit Dmp1-mediated transcription in a Cdk-independent fashion; however, Dmp1 shows synergistic effects with D-cyclins on the Arf promoter. Ras or Myc oncogene-induced tumor formation is accelerated in both Dmp1+/- and Dmp1-/- mice with no significant differences between Dmp1+/- and Dmp1-/-. Thus, Dmp1 is haplo-insufficient for tumor suppression. Tumors from Dmp1-/- or Dmp1+/- mice often retain wild-type Arf and p53, suggesting that Dmp1 is a physiological regulator of the Arf-p53 pathway. The Dmp1 promoter is activated by oncogenic Ras-Raf signaling, while it is repressed by physiological mitogenic stimuli, overexpression of E2F proteins, and genotoxic stimuli mediated by NF-κB. The human DMP1 gene (h DMP1) is located on chromosome 7q21 and is hemizygously deleted in approximately 40% of human lung cancers, especially those that retain normal INK4a/ARF and P53 loci. Thus, h DMP1 is clearly involved in human carcinogenesis, and tumors with h DMP1 deletion may constitute a discrete disease entity.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography