Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
Add more filters

Country/Region as subject
Publication year range
1.
2.
Nat Mater ; 19(4): 381-385, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31959951

ABSTRACT

The search continues for nickel oxide-based materials with electronic properties similar to cuprate high-temperature superconductors1-10. The recent discovery of superconductivity in the doped infinite-layer nickelate NdNiO2 (refs. 11,12) has strengthened these efforts. Here, we use X-ray spectroscopy and density functional theory to show that the electronic structure of LaNiO2 and NdNiO2, while similar to the cuprates, includes significant distinctions. Unlike cuprates, the rare-earth spacer layer in the infinite-layer nickelate supports a weakly interacting three-dimensional 5d metallic state, which hybridizes with a quasi-two-dimensional, strongly correlated state with [Formula: see text] symmetry in the NiO2 layers. Thus, the infinite-layer nickelate can be regarded as a sibling of the rare-earth intermetallics13-15, which are well known for heavy fermion behaviour, where the NiO2 correlated layers play an analogous role to the 4f states in rare-earth heavy fermion compounds. This Kondo- or Anderson-lattice-like 'oxide-intermetallic' replaces the Mott insulator as the reference state from which superconductivity emerges upon doping.

3.
Clin Exp Obstet Gynecol ; 41(1): 10-6, 2014.
Article in English | MEDLINE | ID: mdl-24707674

ABSTRACT

OBJECTIVE: The authors evaluated the effectiveness and safety of "neo-metoro" or 'mini-metoro" metreurynters plus oxytocin for labor induction and assessed differences in parturition outcomes, according to the metreurynter used at induction initiation. MATERIALS AND METHODS: The authors retrospectively reviewed 146 consecutive women with live singleton pregnancies, and who underwent induction. Parturition outcomes were vaginal delivery achieved within the planned schedule (VDPS), vaginal delivery finally achieved (VDF), and induction-to-delivery interval (IDI). Women were divided into neo-metoro, mini-metoro, and without metreurynter groups based on metreurynter use at induction initiation. The authors examined the relationships of metreurynter groups with factors, parturition outcomes, and adverse events. In 113 women who underwent two-day induction, the authors calculated IDI and adjusted odds ratio (AOR) for achieving delivery per unit time. RESULTS: VDPS rates were 65% in nulliparous and 81% in multiparous women. VDF rates were 78% in nulliparous and 96% in multiparous women. AORs for VDPS were 0.30 in nulliparous women and 0.18 in Bishop score (BS) 1-3 class. AORs for VDF were 0.04 in BS1-3 class and 0.14 in BS4-5 class. In 113 women undergoing two-day induction, AORs for achieving delivery per unit time were 0.45 in nulliparous women, 0.46 in obese women, and 0.48 in BS1-3 class. Neo-metoro use at induction initiation tended to reduce IDI. CONCLUSIONS: Labor induction using these metreurynters plus oxytocin is safe and effective. The advantages of neo-metoro over mini-metoro use at induction initiation remain unclear; neo-metoro use at induction initiation may reduce IDI.


Subject(s)
Catheters , Labor, Induced/methods , Oxytocics/administration & dosage , Oxytocin/administration & dosage , Administration, Intravaginal , Adult , Combined Modality Therapy , Equipment Design , Female , Fetal Membranes, Premature Rupture/therapy , Humans , Japan , Parity , Pregnancy , Pregnancy Outcome , Proportional Hazards Models , Retrospective Studies
4.
Gynecol Obstet Fertil Senol ; 50(4): 298-306, 2022 04.
Article in French | MEDLINE | ID: mdl-34626849

ABSTRACT

OBJECTIVE: The uPA/PAI-1 assay and the EPClin® test are useful tools that add to clinico-anatomical characteristics to determine the indication of adjuvant chemotherapy in case of intermediate-prognosis invasive breast cancer. The principal purpose of our study was to analyze the concordance of uPA/PAI-1 and EPClin® in classification of patients into two groups: low and high risk of relapse. METHODS: We prospectively included 63 patients treated for intermediate-prognosis invasive breast cancer. All of these patients received a uPA/PAI-1 assay and an EPClin® test. RESULTS: The uPA/PAI-1 assay and EPClin® test were consistent for 56.2% and inconsistent for 43.8%. In the event of a discrepancy, the treatment decision was based in 95.2% of patients on the EPClin® test result. In total, 38 patients were selected for adjuvant chemotherapy after achievement of the two tests. The mean time to report results after surgery was 9 days for the uPA/PAI-1 assay and 35 days for the EPClin® test. No cases of recurrence or death were found, with an average follow-up of 32 months. CONCLUSION: The EPClin® test resulted in more chemotherapy prescriptions than indicated by uPA/PAI-1. However, we can't conclude to the superiority of one of these two tests, survival data and the effectiveness of our study being insufficient. In general, studies comparing different signatures useful to the therapeutic decision of intermediate prognosis breast cancers should be encouraged.


Subject(s)
Breast Neoplasms , Plasminogen Activator Inhibitor 1 , Breast Neoplasms/drug therapy , Female , Humans , Neoplasm Recurrence, Local , Plasminogen Activator Inhibitor 1/therapeutic use , Prognosis , Urokinase-Type Plasminogen Activator/therapeutic use
5.
Nat Med ; 4(7): 839-43, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9662378

ABSTRACT

The p53 tumor suppressor gene, which is induced by DNA damage and/or stress stimuli, causes cells to undergo G1-arrest or apoptotic death; thus it plays an essential role in human carcinogenesis. We have searched for p53-related genes by using degenerate PCR, and have identified two cDNA fragments similar to but distinct from p53: one previously reported, p73, and the other new. We cloned two major splicing variants of the latter gene and named these p51A and p51B (a human homologue of rat Ket). The p51A gene encodes a 448-amino-acid protein with a molecular weight of 50.9 kDa; and p51B, a 641-amino-acid protein with a molecular weight of 71.9 kDa. In contrast with the ubiquitous expression of p53, expression of p51 mRNA was found in a limited number of tissues, including skeletal muscle, placenta, mammary gland, prostate, trachea, thymus, salivary gland, uterus, heart and lung. In p53-deficient cells, p51A induced growth-suppression and apoptosis, and upregulated p21waf-1 through p53 regulatory elements. Mutations in p51 were found in some human epidermal tumors.


Subject(s)
Alternative Splicing , DNA-Binding Proteins/genetics , Phosphoproteins , Trans-Activators , Tumor Suppressor Protein p53/genetics , Amino Acid Sequence , Animals , Apoptosis , Cats , Cell Division , Chickens , Cloning, Molecular , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation , Genes, Tumor Suppressor , Humans , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Salmon , Sequence Homology, Amino Acid , Tissue Distribution , Transcription Factors , Tumor Cells, Cultured , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins
6.
Science ; 373(6551): 213-216, 2021 Jul 09.
Article in English | MEDLINE | ID: mdl-34244413

ABSTRACT

The discovery of superconductivity in infinite-layer nickelates brings us tantalizingly close to a material class that mirrors the cuprate superconductors. We measured the magnetic excitations in these nickelates using resonant inelastic x-ray scattering at the Ni L 3-edge. Undoped NdNiO2 possesses a branch of dispersive excitations with a bandwidth of approximately 200 milli-electron volts, which is reminiscent of the spin wave of strongly coupled, antiferromagnetically aligned spins on a square lattice. The substantial damping of these modes indicates the importance of coupling to rare-earth itinerant electrons. Upon doping, the spectral weight and energy decrease slightly, whereas the modes become overdamped. Our results highlight the role of Mottness in infinite-layer nickelates.

7.
J Thromb Haemost ; 16(5): 960-972, 2018 05.
Article in English | MEDLINE | ID: mdl-29488681

ABSTRACT

Essentials We generated recombinant rhodocytin that could aggregate platelets via CLEC-2. Recombinant wild-type rhodocytin formed heterooctamer with four α- and ß-subunits. Asp 4 in α-subunit of rhodocytin was required for binding to CLEC-2. Inhibitory mutant of rhodocytin blocked podoplanin-dependent hematogenous metastasis. SUMMARY: Background Rhodocytin, a disulfide-linked heterodimeric C-type lectin from Calloselasma rhodostoma consisting of α-subunits and ß-subunits, induces platelet aggregation through C-type lectin-like receptor 2 (CLEC-2). CLEC-2 is a physiological binding partner of podoplanin (PDPN), which is expressed on some tumor cell types, and is involved in tumor cell-induced platelet aggregation and tumor metastasis. Thus, modified rhodocytin may be a possible source of anti-CLEC-2 drugs for both antiplatelet and antimetastasis therapy. However, its molecular function has not been well characterized, because of the lack of recombinant rhodocytin that induces platelet aggregation. Objective To produce recombinant rhodocytin, in order to verify its function with mutagenesis, and to develop an anti-CLEC-2 drug based on the findings. Methods We used Chinese hamster ovary cells to express recombinant rhodocytin (wild-type [WT] and mutant), which was analyzed for induction/inhibition of platelet aggregation with light transmission aggregometry, the formation of multimers with blue native PAGE, and binding to CLEC-2 with flow cytometry. Finally, we investigated whether mutant rhodocytin could suppress PDPN-induced metastasis in an experimental lung metastasis mouse model. Results Functional WT] rhodocytin (αWTßWT) was obtained by coexpression of both subunits. Asp4 in α-subunits of rhodocytin was required for CLEC-2 binding. αWTßWT formed a heterooctamer similarly to native rhodocytin. Moreover, an inhibitory mutant of rhodocytin (αWTßK53A/R56A), forming a heterotetramer, bound to CLEC-2 without inducing platelet aggregation, and blocked CLEC-2-PDPN interaction-dependent platelet aggregation and experimental lung metastasis. Conclusion These findings provide molecular characterization information on rhodocytin, and suggest that mutant rhodocytin could be used as a therapeutic agent to target CLEC-2.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Lectins, C-Type/antagonists & inhibitors , Lung Neoplasms/prevention & control , Membrane Glycoproteins/antagonists & inhibitors , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Recombinant Proteins/pharmacology , Viper Venoms/pharmacology , Animals , CHO Cells , Cricetulus , Female , HEK293 Cells , Humans , Lectins, C-Type/chemistry , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Membrane Glycoproteins/metabolism , Mice, Inbred BALB C , Mice, Nude , Mutation , Protein Multimerization , Signal Transduction/drug effects , Structure-Activity Relationship , Viper Venoms/chemistry , Viper Venoms/genetics , Viper Venoms/metabolism
8.
Mol Cell Biol ; 19(9): 6333-44, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10454580

ABSTRACT

Multiple biological functions have been ascribed to the Ras-related G protein R-Ras. These include the ability to transform NIH 3T3 fibroblasts, the promotion of cell adhesion, and the regulation of apoptotic responses in hematopoietic cells. To investigate the signaling mechanisms responsible for these biological phenotypes, we compared three R-Ras effector loop mutants (S61, G63, and C66) for their relative biological and biochemical properties. While the S61 mutant retained the ability to cause transformation, both the G63 and the C66 mutants were defective in this biological activity. On the other hand, while both the S61 and the C66 mutants failed to promote cell adhesion and survival in 32D cells, the G63 mutant retained the ability to induce these biological activities. Thus, the ability of R-Ras to transform cells could be dissociated from its propensity to promote cell adhesion and survival. Although the transformation-competent S61 mutant bound preferentially to c-Raf, it only weakly stimulated the mitogen-activated protein kinase (MAPK) activity, and a dominant negative mutant of MEK did not significantly perturb R-Ras oncogenicity. Instead, a dominant negative mutant of phosphatidylinositol 3-kinase (PI3-K) drastically inhibited the oncogenic potential of R-Ras. Interestingly, the ability of the G63 mutant to induce cell adhesion and survival was closely associated with the PI3-K-dependent signaling cascades. To further delineate R-Ras downstream signaling events, we observed that while a dominant negative mutant of Akt/protein kinase inhibited the ability of R-Ras to promote cell survival, both dominant negative mutants of Rac and Ral suppressed cell adhesion stimulated by R-Ras. Thus, the biological actions of R-Ras are mediated by multiple effectors, with PI3-K-dependent signaling cascades being critical to its functions.


Subject(s)
GTP Phosphohydrolases/physiology , GTP-Binding Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Proteins/physiology , Proto-Oncogene Proteins , ras Proteins/physiology , 3T3 Cells , Amino Acid Sequence , Animals , COS Cells , Cell Adhesion , Cell Line , Cell Survival , Cell Transformation, Neoplastic , DNA Primers/genetics , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/genetics , GTPase-Activating Proteins , Humans , Mice , Mutation , Proto-Oncogene Proteins c-akt , Signal Transduction , ral GTP-Binding Proteins , ras GTPase-Activating Proteins
9.
Nanoscale ; 9(19): 6471-6477, 2017 May 18.
Article in English | MEDLINE | ID: mdl-28466951

ABSTRACT

We demonstrate the field-effect transistor (FET) operation of a molecularly-thin anatase phase produced through solid state transformation from Ti0.87O2 nanosheets. A monolayer Ti0.87O2 nanosheet with a thickness of 0.7 nm is a two-dimensional oxide insulator in which Ti vacancies are incorporated, rather than oxygen vacancies. Since the fabrication method, in general, largely affects the film quality, the anatase films derived from the Ti0.87O2 nanosheets show interesting characteristics, such as no photocurrent peak at ∼2 eV, which is related to oxygen vacancies, and a larger band gap of 3.8 eV. The 10 nm thick anatase FETs exhibit superior transport characteristics with a maximum mobility of ∼1.3 cm2 V-1 s-1 and a current on/off ratio of ∼105 at room temperature. The molecularly-thin anatase FET may provide new functionalities, such as field-effect control of catalytic properties.

10.
J Thromb Haemost ; 15(2): 219-229, 2017 02.
Article in English | MEDLINE | ID: mdl-27960039

ABSTRACT

A platelet activation receptor, C-type lectin-like receptor 2 (CLEC-2), has been identified as a receptor for a platelet-activating snake venom, rhodocytin. CLEC-2 protein is highly expressed in platelets/megakaryocytes, and at lower levels in liver Kupffer cells. Recently, podoplanin has been revealed as an endogenous ligand for CLEC-2. Podoplanin is expressed in certain types of tumor cells, fibroblastic reticular cells (FRCs) in lymph nodes, kidney podocytes, and lymphatic endothelial cells, but not in vascular endothelial cells. CLEC-2 in platelets cannot have access to podoplanin under normal conditions, but they interact with each other under pathologic conditions or during developmental stages, and play various pathophysiologic roles. CLEC-2 facilitates hematogenous metastasis of podoplanin-expressing tumors. During development, the interaction between CLEC-2 and podoplanin in lymphatic endothelial cells or neuroepithelial cells facilitates blood-lymphatic vessel separation and cerebrovascular patterning and integrity, respectively. In adulthood, platelet CLEC-2 binding to FRCs is crucial for maintenance of the integrity of high endothelial venules in lymph nodes. Podoplanin-expressing FRC-like cells have recently been identified in the bone marrow, and facilitate megakaryocyte proliferation and proplatelet formation by binding to megakaryocyte CLEC-2. Podoplanin is inducibly expressed in liver monocytes and keratinocytes during Salmonella infection and wound healing, and regulates thrombus formation in the liver and controlled wound healing, respectively. By binding to unknown ligands, platelet CLEC-2 regulates the maintenance of vascular integrity during inflammation, thrombus stability under flow, and maintenance of quiescence of hematopoietic stem cells. Podoplanin is expressed in various cells, and additional roles of the CLEC-2-podoplanin interaction will be revealed in the future.


Subject(s)
Lectins, C-Type/metabolism , Membrane Glycoproteins/metabolism , Blood Platelets/metabolism , Cell Proliferation , Endothelial Cells/metabolism , Fibroblasts/metabolism , Humans , Inflammation , Kupffer Cells/metabolism , Liver/embryology , Liver/metabolism , Liver/pathology , Lymph Nodes/pathology , Megakaryocytes/metabolism , Monocytes/metabolism , Platelet Activation/physiology , Platelet Aggregation , Podocytes/metabolism , Protein Binding , Regeneration , Salmonella Infections , Signal Transduction , Thrombosis , Wound Healing
11.
J Thromb Haemost ; 15(3): 513-525, 2017 03.
Article in English | MEDLINE | ID: mdl-28028907

ABSTRACT

Essentials The role of C-type lectin-like receptor-2 (CLEC-2) in cancer progression is unclear. CLEC-2-depleted mouse model is generated by using a rat anti-mouse CLEC-2 monoclonal antibody. CLEC-2 depletion inhibits hematogenous tumor metastasis of podoplanin-expressing B16F10 cells. CLEC-2 depletion prolongs cancer survival by suppressing thrombosis and inflammation. SUMMARY: Background C-type lectin-like receptor 2 (CLEC-2) is a platelet activation receptor of sialoglycoprotein podoplanin, which is expressed on the surface of certain types of tumor cells. CLEC-2-podoplanin interactions facilitate hematogenous tumor metastasis. However, direct evidence of the role of CLEC-2 in hematogenous metastasis and cancer progression is lacking. Objective and methods We generated immunological CLEC-2-depleted mice by using anti-mouse CLEC-2 monoclonal antibody 2A2B10 and investigated whether CLEC-2 promoted hematogenous tumor metastasis and tumor growth and exacerbated the prognosis of mice bearing podoplanin-expressing B16F10 melanoma cells. Results Our results showed that hematogenous metastasis was significantly inhibited in CLEC-2-depleted mice. B16F10 cells co-cultured with wild-type platelets, but not with CLEC-2-deficient platelets, showed increased proliferation. However, B16F10 cell proliferation was not inhibited in CLEC-2-depleted mice. Histological analysis showed that thrombus formation in tumor vessels was significantly inhibited and functional vessel density was significantly increased in CLEC-2-depleted mice. These data suggest that CLEC-2 deficiency may inhibit thrombus formation in tumor vessels and increase the density of functional vessels, thus improving oxygen and nutrient supply to tumors, indirectly promoting tumor proliferation. Furthermore, the overall survival of CLEC-2-depleted mice was significantly prolonged, which may be due to the suppression of thrombus formation in the lungs and subsequent inhibition of systemic inflammation and cachexia. Conclusions These data provide a rationale for the targeted inhibition of CLEC-2 as a new strategy for preventing hematogenous tumor metastasis and for inhibiting cancer-related thromboembolism.


Subject(s)
Lectins, C-Type/metabolism , Neoplasms/pathology , Platelet Activation , Platelet Aggregation , Thrombosis/genetics , Animals , Antibodies, Monoclonal/chemistry , Blood Platelets/metabolism , Blood Platelets/pathology , Cell Proliferation , Disease Progression , Green Fluorescent Proteins/chemistry , Hemoglobins/chemistry , Melanoma, Experimental , Mice , Mice, Knockout , Neoplasm Metastasis , Prognosis , Rats
12.
Aquat Toxicol ; 79(4): 376-83, 2006 Oct 12.
Article in English | MEDLINE | ID: mdl-16930737

ABSTRACT

To investigate the effect of estrogenic compounds on the marine mussel Mytilus edulis, an assay was developed to measure the expression of two vertebrate estrogen responsive genes-estrogen receptor (ER) and vitellogenin (VTG) genes. Expression was measured in M. edulis gonads following a 10-day exposure to 200 ng/l 17beta-estradiol (estradiol). The concentrations of esterified estradiol in mussel tissue increased 15-fold in a time-dependent manner-confirming uptake of the compound by the mussels, however there was no significant increase of free estradiol in mussel tissues during the exposure period. The ER and VTG mRNA levels in the gonads of both sexes were measured at days 1-3, 5, and 10 in control and exposed mussels. However, no significant change in the expression of either the ER or VTG genes was recorded at any of the sampled time points. The results suggest that either a regulatory mechanism exists in a mussel that is able to maintain constant levels of free estradiol by converting the excess estradiol into esterified products which may have reduced affinity for the estrogen receptor, or alternatively, that the ER and VTG genes are unresponsive to estrogens in these organisms. The significance of these findings in terms of the utility of ER and VTG as biomarkers of endocrine disruption in bivalve species is discussed.


Subject(s)
Estradiol/toxicity , Gene Expression/drug effects , Mytilus edulis/drug effects , Receptors, Estrogen/genetics , Vitellogenins/genetics , Water Pollutants, Chemical/toxicity , Amino Acid Sequence , Animals , DNA Primers/chemistry , Estradiol/analysis , Female , Gonads/chemistry , Gonads/drug effects , Male , Molecular Sequence Data , Mytilus edulis/chemistry , RNA, Messenger/analysis , Receptors, Estrogen/biosynthesis , Receptors, Estrogen/drug effects , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Sequence Alignment/veterinary , Vitellogenins/biosynthesis , Vitellogenins/drug effects , Water/analysis
13.
Cancer Res ; 59(23): 5908-11, 1999 Dec 01.
Article in English | MEDLINE | ID: mdl-10606233

ABSTRACT

The p51/p63 gene is a homologue of p53, the product of which acts as a transcriptional activator by binding to p53-responsive elements in the promoter regions of several p53 downstream genes. Recently, we identified four distinct mutations in the p51/p63 gene after screening >200 human tumors and cell lines. Because all of the detected p51/p63 mutations were missense mutations, the pathogenic effect of these mutations is difficult to determine without performing a functional analysis. In this study, we examined the transcriptional activity of tumor-derived p51/p63 missense mutations using a yeast-based assay and compared the data with that of artificial p51/p63 missense mutations at residues corresponding to the positions and substituted residues of p53 mutation "hotspots." Although most of the p51/p63 missense mutations at the p53 hotspot residues were unable to transactivate the promoters used in this study, the tumor-derived p51/p63 missense mutations retained their ability to transactivate the MDM2 and/or the BAX promoter but not the p21/WAF1 promoter. These results suggest that the p51/p63 mutation might be involved in an unknown tumor suppression pathway distinct from that of p53.


Subject(s)
DNA-Binding Proteins/genetics , Genes, p53 , Membrane Proteins , Mutation, Missense , Phosphoproteins/genetics , Promoter Regions, Genetic , Trans-Activators , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Amino Acid Substitution , Cloning, Molecular , DNA-Binding Proteins/metabolism , Genes, Tumor Suppressor , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphoproteins/metabolism , Saccharomyces cerevisiae , Sequence Alignment , Sequence Homology, Amino Acid , Transcription Factors , Tumor Suppressor Proteins
14.
Cancer Res ; 59(12): 2781-6, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10383130

ABSTRACT

p51/p63 is a novel p53 homologue that has been shown to act as a transcriptional activator through the p53-binding sequence of the p21/WAF1 promoter and to induce apoptosis when it is expressed transiently in a human tumor cell line. We developed transcription assay systems for these two related genes in both Saccharomyces cerevisiae and mammalian cells and used them to investigate the functional similarities and differences of these genes. We found that p51/p63 trans-activated the previously identified p53 target genes, but the degree of the transactivation by p51/p63 differed from that by p53. These results suggest that the cellular signal on p51/p63 cross-talks partially but not completely with that of the p53 pathway.


Subject(s)
DNA-Binding Proteins/physiology , Nuclear Proteins , Phosphoproteins , Proto-Oncogene Proteins c-bcl-2 , Trans-Activators , Transcription, Genetic , Transcriptional Activation , Tumor Suppressor Protein p53/physiology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA-Binding Proteins/chemistry , Gene Expression Regulation, Fungal , Gene Expression Regulation, Neoplastic , Genes, Reporter , Genes, Tumor Suppressor , Humans , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2 , Saccharomyces cerevisiae/genetics , Transcription Factors , Tumor Cells, Cultured , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Proteins , bcl-2-Associated X Protein
16.
Oncogene ; 19(16): 2014-22, 2000 Apr 13.
Article in English | MEDLINE | ID: mdl-10803462

ABSTRACT

The GTP-binding protein, R-Ras3/M-Ras, is a novel member of the Ras subfamily of GTPases which shows highest sequence similarity to the TC21 gene. R-Ras3 is highly expressed in both human and mouse brain and ectopic expression of a constitutively active mutant of R-Ras3 induces cellular transformation in NIH3T3 cells. To gain further insight into the normal cellular function of R-Ras3, we examined the ability of R-Ras3 in activating several known intracellular signaling cascades. We observed that R-Ras3 is a relatively weak activator of the mitogen-activated protein kinase/extracellular-signal-regulated kinases (MAPK/ERKs) when compared to the H-Ras oncogene. On the contrary, both R-Ras3 and H-Ras activated the Jun N-terminal kinase (JNK) to a similar extent. Under similar experimental conditions, R-Ras3 significantly stimulated one of the phosphatidylinositol 3-kinase (PI3-K) downstream substrates, Akt/PKB/RAC (Akt), which has been extensively implicated in mediating cell survival signaling. The activation of Akt by R-Ras3 was most likely to be PI3-K-dependent since this biochemical event was blocked by the pharmacological inhibitors, Wortmannin and LY294002, as well as by a dominant negative mutant of PI3-K. More importantly, R-Ras3 affinity-precipitated PI3-K from cell extracts in a GTP-dependent manner, and associated lipid kinase activity was readily detectable in R-Ras3 immune complexes. The biological significance of R-Ras3 in inducing Akt kinase activity is evidenced by the ability of an activated R-Ras3 to confer cell survival in the rat pheochromocytoma cell line, PC12. As expected, this biological activity of R-Ras3 was also abrogated by the addition of LY294002. Thus, R-Ras3 represents a novel G-protein which may play a role in cell survival of neural-derived cells.


Subject(s)
Brain/enzymology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins , ras Proteins/metabolism , 3T3 Cells/metabolism , Androstadienes/pharmacology , Animals , Brain/cytology , COS Cells/metabolism , Cell Survival/drug effects , Chromones/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Genes, ras , Guanosine Triphosphate/metabolism , JNK Mitogen-Activated Protein Kinases , Mice , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Morpholines/pharmacology , Nerve Growth Factor/pharmacology , Neurons/enzymology , Organ Specificity , PC12 Cells/cytology , PC12 Cells/drug effects , PC12 Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Wortmannin , ras Proteins/genetics
17.
Oncogene ; 15(22): 2675-85, 1997 Nov 27.
Article in English | MEDLINE | ID: mdl-9400994

ABSTRACT

Members of the Ras subfamily of GTP-binding proteins, including Ras (H-, K-, and N-), TC21, and R-ras have been shown to display transforming activity, and activating lesions have been detected in human tumors. We have identified an additional member of the Ras gene family which shows significant sequence similarity to the human TC21 gene. This novel human ras-related gene, R-ras3, encodes for a protein of 209 amino acids, and shows approximately 60-75% sequence identity in the N-terminal catalytic domain with members of the Ras subfamily of GTP-binding proteins. An activating mutation corresponding to the leucine 61 oncogenic lesion of the ras oncogenes when introduced into R-ras3, activates its transforming potential. R-ras3 weakly stimulates the mitogen-activated protein kinase (MAPK) activity, but this effect is greatly potentiated by the co-expression of c-raf-1. By the yeast two-hybrid system, R-ras3 interacts only weakly with known Ras effectors, such as Raf and RalGDS, but not with RglII. In addition, R-ras3 displays modest stimulatory effects on trans-activation from different nuclear response elements which bind transcription factors, such as SRF, ETS/TCF, Jun/Fos, and NF-kappaB/Rel. Interestingly, Northern blot analysis of total RNA isolated from various tissues revealed that the 3.8 kilobasepair (kb) transcript of R-ras3 is highly restricted to the brain and heart. The close evolutionary conservation between R-ras3 and Ras family members, in contrast to the significant differences in its biological activities and the pattern of tissue expression, raise the possibility that R-ras3 may control novel cellular functions previously not described for other GTP-binding proteins.


Subject(s)
ras Proteins/genetics , ras Proteins/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Transformation, Neoplastic/genetics , Cloning, Molecular , DNA, Complementary , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Humans , Mice , Molecular Sequence Data , Sequence Homology, Amino Acid , Tissue Distribution
18.
Thromb Res ; 115(5): 409-15, 2005.
Article in English | MEDLINE | ID: mdl-15733975

ABSTRACT

We analyzed the intracellular action of sphingosine 1-phosphate (Sph-1-P), formed from sphingosine (Sph) by sphingosine kinase (SPHK), in platelets. When sphingosine kinase activity was inhibited by N,N-dimethylsphingosine (DMS), Ca2+ mobilization induced by convulxin, an agonist of the collagen receptor glycoprotein VI (GPVI), was moderately but specifically abolished; that induced via G protein-coupled receptors was not affected. Under the same conditions, however, tyrosine phosphorylation of Syk and phospholipase Cgamma2, which is essential for the GPVI-mediated signaling, was not inhibited. Sphingosine kinase activity of the platelet membrane fraction increased specifically upon stimulation with convulxin or collagen. Our results suggest that intracellular sphingosine 1-phosphate is related to Ca2+ mobilization in GPVI-mediated signaling pathways.


Subject(s)
Blood Platelets/drug effects , Calcium Signaling/drug effects , Calcium/metabolism , Lysophospholipids/metabolism , Platelet Membrane Glycoproteins/metabolism , Sphingosine/analogs & derivatives , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Blood Platelets/metabolism , Calcium Signaling/physiology , Collagen/pharmacology , Crotalid Venoms/pharmacology , Humans , Lectins, C-Type , Lysophospholipids/blood , Phosphorylation/drug effects , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Platelet Membrane Glycoproteins/agonists , Receptors, Thrombin/chemistry , Sphingosine/blood , Sphingosine/metabolism , Sphingosine/pharmacology , Tyrosine/drug effects , Tyrosine/metabolism
19.
Exp Hematol ; 24(6): 690-5, 1996 May.
Article in English | MEDLINE | ID: mdl-8635524

ABSTRACT

Megakaryocytes generate cytoplasmic processes (CP) that penetrate endothelial cells in the bone marrow sinus, and these processes may release platelets into the circulation at their terminal stage. Adhesion between the CP and endothelial cells may be important during the extension of CP. We examined the expression of adhesion molecules of the integrin family (CDw49b, CDw49d, CDw49e, CDw49f, CD18, CD11a CD11c, and CD11b), the immunoglobulin superfamily (CD54, CD56, CD58, and CD31), the selectin family (ELAM-1, LECAM-1, and CD62), and CD44, CD41b, and CD42b on platelets, megakaryocytes, and megakaryocytes with CP. No specific adhesion molecules were observed on the megakaryocytes with CP. Three staining patterns of adhesion molecules-homogeneous, speckled, and accumulated-were observed on the megakaryocytes with CP, but not on those without CP. Platelet integrins (i.e., CD41a, CDw49b, CDw49e and CDw49f) and GPIb (CD42b) were strongly and homogeneously stained on the CP. GMP-140 CD62) was weakly stained, in a speckled pattern. CD31 (PECAM-1) was also weakly stained but accumulated selectively on the tip of the CP. ANTI-CD31 suppressed CP formation of megakaryocytes. We speculate that the homodimerization of CD31 expressed on the tips of CP and endothelial cells is important for the extension of the processes and for the migration of megakaryocytes.


Subject(s)
Cell Adhesion Molecules/metabolism , Megakaryocytes/cytology , Antigens, CD/metabolism , Blood Platelets/metabolism , Bone Marrow Cells , Cell Adhesion , Cytoplasm/ultrastructure , Humans , Immunophenotyping , Megakaryocytes/ultrastructure
20.
Exp Hematol ; 27(1): 131-8, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9923451

ABSTRACT

We examined withdrawal effects of recombinant mouse Tpo (rm-Tpo) on the apoptosis of mature and immature megakaryocytes in in vitro experiments. Apoptotic megakaryocytes were detected by double staining for acetylcholinesterase and by the TdT-mediated dUTP-biotin nick end labeling (TUNEL) method. When the purified mature megakaryocytes were cultured with or without rm-Tpo, the numbers of viable megakaryocytes, apoptotic megakaryocytes, and megakaryocytes with cytoplasmic processes were not significantly different between the two groups. In contrast, purified immature megakaryocytes underwent apoptosis when rm-Tpo was absent from the culture system. Murine bone marrow cells were cultured with rm-Tpo (50 U/mL) on days 1-7 to generate immature megakaryocytes and subsequently were cultured with different concentrations of rm-Tpo (0-50 U/mL) on days 8-14. The number of viable megakaryocytes was decreased and that of apoptotic megakaryocytes was increased by rm-Tpo in a dose-dependent manner. These results indicated a clear relation between the rm-Tpo level and the apoptosis of immature megakaryocytes.


Subject(s)
Apoptosis/physiology , Megakaryocytes/cytology , Thrombopoietin/physiology , Animals , Apoptosis/drug effects , Bone Marrow Cells/cytology , Cell Count , Cell Division/physiology , Cell Survival/drug effects , Cells, Cultured/drug effects , Dose-Response Relationship, Drug , Female , In Situ Nick-End Labeling , Mice , Mice, Inbred Strains , Recombinant Proteins , Thrombopoietin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL