Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 134
Filter
2.
Eur J Clin Nutr ; 58(9): 1253-6, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15054441

ABSTRACT

OBJECTIVE: To determine whether trigonelline contributes to the effect of coffee on homocysteine (Hcy). DESIGN AND INTERVENTIONS: This was a randomised crossover study. Subjects consumed 50 mg trigonelline, 5 g of instant coffee (approximately 50 mg trigonelline) or water, consumed as a single dose in 100 ml, with 1 week between each treatment. Blood samples were drawn fasting and hourly for 8 h. Urine samples were collected pretreatment and every 2 h for 8 h. SETTING: Christchurch Clinical Studies Trust, Christchurch, New Zealand. SUBJECTS: Eight healthy male subjects. RESULTS: Instant coffee raised plasma Hcy concentrations compared with water (P=0.019) and trigonelline (P=0.037). Plasma Hcy concentrations were not different between water and trigonelline treatments (P=0.789). The change in plasma Hcy concentration was higher (mean+/-s.e.) 4 h (0.7+/-0.2 micromol/l, P=0.006), 5 h (0.7+/-0.2 micromol/l, P=0.013) and 7 h (0.7+/-0.2 micromol/l, P=0.024) following coffee consumption. Urinary glycine betaine excretion was increased by coffee but not by trigonelline. CONCLUSION: Ingestion of instant coffee acutely elevated plasma Hcy; however, trigonelline is not responsible for this rise. SPONSORSHIP: Supported by the Health Research Council, the Canterbury Medical Foundation, the Foundation of Research, Science and Technology.


Subject(s)
Alkaloids/pharmacology , Coffee , Homocysteine/blood , Adult , Area Under Curve , Cross-Over Studies , Fasting , Humans , Male , Placebos
3.
Proc Natl Acad Sci U S A ; 98(26): 14889-94, 2001 Dec 18.
Article in English | MEDLINE | ID: mdl-11742073

ABSTRACT

One aspect of the function of the beta-arrestins is to serve as scaffold or adapter molecules coupling G-protein coupled receptors (GPCRs) to signal transduction pathways distinct from traditional second messenger pathways. Here we report the identification of Dishevelled 1 and Dishevelled 2 (Dvl1 and Dvl2) as beta-arrestin1 (betaarr1) interacting proteins. Dvl proteins participate as key intermediates in signal transmission from the seven membrane-spanning Frizzled receptors leading to inhibition of glycogen synthase kinase-3beta (GSK-3beta), stabilization of beta-catenin, and activation of the lymphoid enhancer factor (LEF) transcription factor. We find that phosphorylation of Dvl strongly enhances its interaction with betaarr1, suggesting that regulation of Dvl phosphorylation and subsequent interaction with betaarr1 may play a key role in the activation of the LEF transcription pathway. Because coexpression of the Dvl kinases, CK1epsilon and PAR-1, with Dvl synergistically activates LEF reporter gene activity, we reasoned that coexpression of betaarr1 with Dvl might also affect LEF-dependent gene activation. Interestingly, whereas betaarr1 or Dvl alone leads to low-level stimulation of LEF (2- to 5-fold), coexpression of betaarr1 with either Dvl1 or Dvl2 leads to a synergistic activation of LEF (up to 16-fold). Additional experiments with LiCl as an inhibitor of GSK-3beta kinase activity indicate that the step affected by betaarr1 is upstream of GSK-3beta and most likely at the level of Dvl. These results identify betaarr1 as a regulator of Dvl-dependent LEF transcription and suggest that betaarr1 might serve as an adapter molecule that can couple Frizzled receptors and perhaps other GPCRs to these important transcription pathways.


Subject(s)
Arrestins/physiology , DNA-Binding Proteins/genetics , Proteins/metabolism , Transcription Factors/genetics , Transcription, Genetic/physiology , Adaptor Proteins, Signal Transducing , Animals , Arrestins/metabolism , Cell Line , Dishevelled Proteins , Humans , Lymphoid Enhancer-Binding Factor 1 , Mice , Phosphoproteins , Phosphorylation , Protein Binding , beta-Arrestins
4.
Sci STKE ; 2001(69): pe1, 2001 Feb 13.
Article in English | MEDLINE | ID: mdl-11752639

ABSTRACT

Recent data suggest that internalized receptor and arrestin complexes are actively involved in signal transduction. Miller and Lefkowitz discuss evidence from the Drosophila visual system that suggests that intracellular rhodopsin and arrestin2 complexes induce apoptosis. Experiments with activated mammalian G protein-coupled receptor and arrestin complexes point to a mechanism by which proliferative or proapoptotic signals can be mediated largely independent from G protein activation.


Subject(s)
Apoptosis/physiology , Arrestins/physiology , Signal Transduction/physiology , Animals , Humans
5.
J Biol Chem ; 276(45): 42509-13, 2001 Nov 09.
Article in English | MEDLINE | ID: mdl-11533043

ABSTRACT

beta-Arrestins are multifunctional adaptor proteins known to regulate internalization of agonist-stimulated G protein-coupled receptors by linking them to endocytic proteins such as clathrin and AP-2. Here we describe a previously unappreciated mechanism by which beta-arrestin orchestrates the process of receptor endocytosis through the activation of ADP-ribosylation factor 6 (ARF6), a small GTP-binding protein. Involvement of ARF6 in the endocytic process is demonstrated by the ability of GTP-binding defective and GTP hydrolysis-deficient mutants to inhibit internalization of the beta(2)-adrenergic receptor. The importance of regulation of ARF6 function is shown by the ability of the ARF GTPase-activating protein GIT1 to inhibit and of the ARF nucleotide exchange factor, ARNO, to enhance receptor endocytosis. Endogenous beta-arrestin is found in complex with ARNO. Upon agonist stimulation of the receptor, beta-arrestin also interacts with the GDP-liganded form of ARF6, thereby facilitating ARNO-promoted GTP loading and activation of the G protein. Thus, the agonist-driven formation of a complex including beta-arrestin, ARNO, and ARF6 provides a molecular mechanism that explains how the agonist-stimulated receptor recruits a small G protein necessary for the endocytic process and controls its activation.


Subject(s)
ADP-Ribosylation Factors/physiology , Arrestins/physiology , Endocytosis , Receptors, Adrenergic, beta-2/metabolism , ADP-Ribosylation Factor 6 , Animals , Cells, Cultured , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Guanosine Triphosphate/metabolism , beta-Arrestins
6.
Toxicol Appl Pharmacol ; 175(3): 217-25, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11559020

ABSTRACT

Apoptosis was measured in rat alveolar macrophage NR8383 cells challenged in vitro with respirable quartz or kaolin dust and with the dusts pretreated with dipalmitoyl phosphatidylcholine (DPPC) to model conditioning of respired dusts by interaction with a primary phospholipid component of pulmonary surfactant. Quartz dust is known to induce apoptosis in vitro and in vivo. For this study, quartz and kaolin were compared as dusts of similar cytotoxicity in some in vitro assays but of differing pathogenic potential: quartz can cause significant pulmonary fibrosis while kaolin generally does not. NR8383 cells exposed to native quartz at concentrations from 50 to 400 microg/ml for 6 h showed a dose-dependent increase in apoptosis measured by the TdT-mediated dUTP-fluorescein nick end labeling (TUNEL), cell death ELISA, and DNA ladder formation assays, while native kaolin induced significant response only at the higher concentrations and only in the TUNEL and ELISA assays. For cell challenge from 6 h to 5 days at 100 microg/ml of dust, quartz was active at all times while kaolin was active only at 5 days. DPPC pre-treatment suppressed quartz activity until 3 days and kaolin activity through 5 days. Cellular release of lactate dehydrogenase, measured in parallel experiments to compare dust apoptotic and necrotic activities, indicated that components of serum as well as surfactant may affect kaolin in vitro expression of those activities.


Subject(s)
1,2-Dipalmitoylphosphatidylcholine/pharmacology , Apoptosis/drug effects , Kaolin/toxicity , Macrophages, Alveolar/drug effects , Quartz/toxicity , Animals , Cell Line , DNA/analysis , DNA Fragmentation , Dose-Response Relationship, Drug , Drug Combinations , Drug Interactions , Dust , Enzyme-Linked Immunosorbent Assay , In Situ Nick-End Labeling , L-Lactate Dehydrogenase/metabolism , Macrophages, Alveolar/enzymology , Macrophages, Alveolar/pathology , Male , Rats , Rats, Sprague-Dawley
7.
J Biol Chem ; 276(47): 43663-7, 2001 Nov 23.
Article in English | MEDLINE | ID: mdl-11546805

ABSTRACT

The insulin and the endothelin type A (ETA) receptor both can couple into the heterotrimeric G protein alpha(q/11) (Galpha(q/11)), leading to Galpha(q/11) tyrosine phosphorylation, phosphatidylinositol 3-kinase activation, and subsequent stimulation of glucose transport. In this study, we assessed the potential role of Src kinase in ET-1 signaling to glucose transport in 3T3-L1 adipocytes. Src kinase inhibitor PP2 blocked ET-1-induced Src kinase activity, Galpha(q/11) tyrosine phosphorylation, and glucose transport stimulation. To determine which Src family kinase member was involved, we microinjected anti-c-Src, -c-Fyn, or -c-Yes antibody into these cells and found that only anti-c-Yes antibody blocked GLUT4 translocation (70% decreased). Overexpression or microinjection of a dominant negative mutant (K298M) of Src kinase also inhibited ET-1-induced Galpha(q/11) tyrosine phosphorylation and GLUT4 translocation. In co-immunoprecipitation experiments, we found that beta-arrestin 1 associated with the ETA receptor in an agonist-dependent manner and that beta-arrestin 1 recruited Src kinase to a molecular complex that included the ETA receptor. Microinjection of beta-arrestin 1 antibody inhibited ET-1- but not insulin-stimulated GLUT4 translocation. In conclusion, 1) the Src kinase Yes can induce tyrosine phosphorylation of Galpha(q/11) in response to ET-1 stimulation, and 2) beta-arrestin 1 and Src kinase form a molecular complex with the ETA receptor to mediate ET-1 signaling to Galpha(q/11) with subsequent glucose transport stimulation.


Subject(s)
Arrestins/physiology , Endothelin-1/pharmacology , Glucose/metabolism , Muscle Proteins , Proto-Oncogene Proteins/metabolism , src-Family Kinases/metabolism , 3T3 Cells , Animals , Arrestins/metabolism , Biological Transport , GTP-Binding Protein alpha Subunits, Gq-G11 , Glucose Transporter Type 4 , Heterotrimeric GTP-Binding Proteins/metabolism , Mice , Microscopy, Fluorescence , Monosaccharide Transport Proteins/metabolism , Proto-Oncogene Proteins c-yes , Signal Transduction , beta-Arrestin 1 , beta-Arrestins
8.
J Biol Chem ; 276(30): 27770-7, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11356842

ABSTRACT

Accumulating evidence indicates that the beta-arrestins act as scaffold molecules that couple G-protein-coupled receptors to mitogen-activated protein (MAP) kinase signaling pathways. Recently, we identified the c-Jun N-terminal kinase 3 (JNK3) as a beta-arrestin2-interacting protein in yeast-two hybrid and co-immunoprecipitation studies. Beta-arrestin2 acts as a scaffold to enhance signaling to JNK3 stimulated by overexpression of the MAP3 kinase ASK1 or by agonist activation of the angiotensin 1A receptor. Whereas beta-arrestin2 is a very strong activator of JNK3 signaling, beta-arrestin1 is very weak in this regard. The data also indicate that the specific step enhanced by beta-arrestin2 involves phosphorylation of JNK3 by the MAP2 kinase MKK4. We reasoned that defining the region (or domain) in beta-arrestin2 responsible for high level JNK3 activation would provide insight into the mechanism by which beta-arrestin2 enhances the activity of this signaling pathway. Using chimeric beta-arrestins, we have determined that sequences in the carboxyl-terminal region of beta-arrestin2 are important for the enhancement of JNK3 phosphorylation. More detailed analysis of the carboxyl-terminal domains of the beta-arrestins indicated that beta-arrestin2, but not beta-arrestin1, contains a sequence (RRSLHL) highly homologous to the conserved docking motif present in many MAP kinase-binding proteins. Replacement of the beta-arrestin2 RRS residues with the corresponding KP residues present in beta-arrestin1 dramatically reduced both JNK3 interaction and enhancement of JNK3 phosphorylation. Conversely, replacement of the KP residues in beta-arrestin1 with RRS significantly increased both JNK3 binding and enhancement of JNK3 phosphorylation. These results delineate a mechanism by which beta-arrestin2 functions as a scaffold protein in the JNK3 signaling pathway and implicate the conserved docking site in beta-arrestin2 as an important factor in binding JNK3 and stimulating the phosphorylation of JNK3 by MKK4.


Subject(s)
Arabidopsis Proteins , Arrestins/chemistry , Mitogen-Activated Protein Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , COS Cells , Dose-Response Relationship, Drug , Enzyme Activation , Immunoblotting , Mitogen-Activated Protein Kinase 10 , Mitogen-Activated Protein Kinase Kinases/metabolism , Models, Biological , Molecular Sequence Data , Phosphorylation , Plant Proteins/metabolism , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Receptors, Adrenergic, beta-2/metabolism , Sequence Homology, Amino Acid , Signal Transduction , beta-Arrestins
9.
Curr Opin Cell Biol ; 13(2): 139-45, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11248546

ABSTRACT

beta-arrestins play previously unsuspected and important roles as adapters and scaffolds that localize signaling proteins to ligand-activated G-protein-coupled receptors. As with the paradigmatic role of the beta-arrestins in uncoupling receptors from G proteins (desensitization), these novel functions involve the interaction of beta-arrestin with phosphorylated heptahelical receptors. beta-arrestins interact with at least two main classes of signaling proteins. First, interaction with molecules such as clathrin, AP-2 and NSF directs the clathrin-mediated internalization of G-protein-coupled receptors. Second, interaction with molecules such as Src, Raf, Erk, ASK1 and JNK3 appears to regulate several pathways that result in the activation of MAP kinases. These recent discoveries indicate that the beta-arrestins play widespread roles as scaffolds and/or adapter molecules that organize a variety of complex signaling pathways emanating from heptahelical receptors. It is likely that additional roles for the beta-arrestins remain to be discovered.


Subject(s)
Arrestins/physiology , GTP-Binding Proteins/metabolism , Receptors, Cell Surface/physiology , Animals , Endocytosis , Humans , Receptors, Cell Surface/metabolism , Signal Transduction , beta-Arrestins
10.
Proc Natl Acad Sci U S A ; 98(5): 2449-54, 2001 Feb 27.
Article in English | MEDLINE | ID: mdl-11226259

ABSTRACT

Using both confocal immunofluorescence microscopy and biochemical approaches, we have examined the role of beta-arrestins in the activation and targeting of extracellular signal-regulated kinase 2 (ERK2) following stimulation of angiotensin II type 1a receptors (AT1aR). In HEK-293 cells expressing hemagglutinin-tagged AT1aR, angiotensin stimulation triggered beta-arrestin-2 binding to the receptor and internalization of AT1aR-beta-arrestin complexes. Using red fluorescent protein-tagged ERK2 to track the subcellular distribution of ERK2, we found that angiotensin treatment caused the redistribution of activated ERK2 into endosomal vesicles that also contained AT1aR-beta-arrestin complexes. This targeting of ERK2 reflects the formation of multiprotein complexes containing AT1aR, beta-arrestin-2, and the component kinases of the ERK cascade, cRaf-1, MEK1, and ERK2. Myc-tagged cRaf-1, MEK1, and green fluorescent protein-tagged ERK2 coprecipitated with Flag-tagged beta-arrestin-2 from transfected COS-7 cells. Coprecipitation of cRaf-1 with beta-arrestin-2 was independent of MEK1 and ERK2, whereas the coprecipitation of MEK1 and ERK2 with beta-arrestin-2 was significantly enhanced in the presence of overexpressed cRaf-1, suggesting that binding of cRaf-1 to beta-arrestin facilitates the assembly of a cRaf-1, MEK1, ERK2 complex. The phosphorylation of ERK2 in beta-arrestin complexes was markedly enhanced by coexpression of cRaf-1, and this effect is blocked by expression of a catalytically inactive dominant inhibitory mutant of MEK1. Stimulation with angiotensin increased the binding of both cRaf-1 and ERK2 to beta-arrestin-2, and the association of beta-arrestin-2, cRaf-1, and ERK2 with AT1aR. These data suggest that beta-arrestins function both as scaffolds to enhance cRaf-1 and MEK-dependent activation of ERK2, and as targeting proteins that direct activated ERK to specific subcellular locations.


Subject(s)
Arrestins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Angiotensin II/pharmacology , Animals , Cell Line , Enzyme Activation , Humans , Microscopy, Confocal , beta-Arrestin 2 , beta-Arrestins
11.
Science ; 290(5496): 1574-7, 2000 Nov 24.
Article in English | MEDLINE | ID: mdl-11090355

ABSTRACT

beta-Arrestins, originally discovered in the context of heterotrimeric guanine nucleotide binding protein-coupled receptor (GPCR) desensitization, also function in internalization and signaling of these receptors. We identified c-Jun amino-terminal kinase 3 (JNK3) as a binding partner of beta-arrestin 2 using a yeast two-hybrid screen and by coimmunoprecipitation from mouse brain extracts or cotransfected COS-7 cells. The upstream JNK activators apoptosis signal-regulating kinase 1 (ASK1) and mitogen-activated protein kinase (MAPK) kinase 4 were also found in complex with beta-arrestin 2. Cellular transfection of beta-arrestin 2 caused cytosolic retention of JNK3 and enhanced JNK3 phosphorylation stimulated by ASK1. Moreover, stimulation of the angiotensin II type 1A receptor activated JNK3 and triggered the colocalization of beta-arrestin 2 and active JNK3 to intracellular vesicles. Thus, beta-arrestin 2 acts as a scaffold protein, which brings the spatial distribution and activity of this MAPK module under the control of a GPCR.


Subject(s)
Arrestins/metabolism , MAP Kinase Kinase 4 , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, Angiotensin/metabolism , Angiotensin II/metabolism , Angiotensin II/pharmacology , Animals , Arrestins/genetics , COS Cells , Cell Line , Cell Nucleus/metabolism , Cytosol/enzymology , Cytosol/metabolism , Endosomes/enzymology , Endosomes/metabolism , Enzyme Activation , Humans , MAP Kinase Kinase Kinase 5 , Mice , Mitogen-Activated Protein Kinase 10 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism , Rats , Receptor, Angiotensin, Type 1 , Recombinant Fusion Proteins/metabolism , Transfection , Two-Hybrid System Techniques , beta-Arrestin 2 , beta-Arrestins
12.
J Biol Chem ; 275(49): 38659-66, 2000 Dec 08.
Article in English | MEDLINE | ID: mdl-10995758

ABSTRACT

The beta(1)-adrenergic receptor (beta(1)AR) is the most abundant subtype of beta-adrenergic receptor in the mammalian brain and is known to potently regulate synaptic plasticity. To search for potential neuronal beta(1)AR-interacting proteins, we screened a rat brain cDNA library using the beta(1)AR carboxyl terminus (beta(1)AR-CT) as bait in the yeast two-hybrid system. These screens identified PSD-95, a multiple PDZ domain-containing scaffolding protein, as a specific binding partner of the beta(1)AR-CT. This interaction was confirmed by in vitro fusion protein pull-down and blot overlay experiments, which demonstrated that the beta(1)AR-CT binds specifically to the third PDZ domain of PSD-95. Furthermore, the full-length beta(1)AR associates with PSD-95 in cells, as determined by co-immunoprecipitation experiments and immunofluorescence co-localization studies. The interaction between beta(1)AR and PSD-95 is mediated by the last few amino acids of the beta(1)AR, and mutation of the beta(1)AR carboxyl terminus eliminated the binding and disrupted the co-localization of the beta(1)AR and PSD-95 in cells. Agonist-induced internalization of the beta(1)AR in HEK-293 cells was markedly attenuated by PSD-95 co-expression, whereas co-expression of PSD-95 has no significant effect on either desensitization of the beta(1)AR or beta(1)AR-induced cAMP accumulation. Furthermore, PSD-95 facilitated the formation of a complex between the beta(1)AR and N-methyl-d-aspartate receptors, as assessed by co-immunoprecipitation. These data reveal that PSD-95 is a specific beta(1)AR binding partner that modulates beta(1)AR function and facilitates physical association of the beta(1)AR with synaptic proteins, such as the N-methyl-d-aspartate receptors, which are known to be regulated by beta(1)AR stimulation.


Subject(s)
Nerve Tissue Proteins/metabolism , Receptors, Adrenergic, beta-1/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Brain/metabolism , COS Cells , Cell Line , Chlorocebus aethiops , Cloning, Molecular , Disks Large Homolog 4 Protein , Gene Library , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Nerve Tissue Proteins/chemistry , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Protein Transport , Rats , Receptors, Adrenergic, beta-1/chemistry , Receptors, Adrenergic, beta-1/genetics , Receptors, N-Methyl-D-Aspartate/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae , Transfection
13.
J Biol Chem ; 275(13): 9572-80, 2000 Mar 31.
Article in English | MEDLINE | ID: mdl-10734107

ABSTRACT

Many G protein-coupled receptors (GPCRs) activate MAP kinases by stimulating tyrosine kinase signaling cascades. In some systems, GPCRs stimulate tyrosine phosphorylation by inducing the "transactivation" of a receptor tyrosine kinase (RTK). The mechanisms underlying GPCR-induced RTK transactivation have not been clearly defined. Here we report that GPCR activation mimics growth factor-mediated stimulation of the epidermal growth factor receptor (EGFR) with respect to many facets of RTK function. beta(2)-Adrenergic receptor (beta(2)AR) stimulation of COS-7 cells induces EGFR dimerization, tyrosine autophosphorylation, and EGFR internalization. Coincident with EGFR transactivation, isoproterenol exposure induces the formation of a multireceptor complex containing both the beta(2)AR and the "transactivated" EGFR. beta(2)AR-mediated EGFR phosphorylation and subsequent beta(2)AR stimulation of extracellular signal-regulated kinase (ERK) 1/2 are sensitive to selective inhibitors of both EGFR and Src kinases, indicating that both kinases are required for EGFR transactivation. beta(2)AR-dependent signaling to ERK1/2, like direct EGF stimulation of ERK1/2 activity, is sensitive to inhibitors of clathrin-mediated endocytosis, suggesting that signaling downstream of both the EGF-activated and the GPCR-transactivated EGFRs requires a productive engagement of the complex with the cellular endocytic machinery. Thus, RTK transactivation is revealed to be a process involving both association of receptors of distinct classes and the interaction of the transactivated RTK with the cells endocytic machinery.


Subject(s)
ErbB Receptors/metabolism , Mitogen-Activated Protein Kinases/metabolism , Receptors, Adrenergic, beta-2/physiology , Animals , COS Cells , Clathrin/physiology , Endocytosis/physiology , Enzyme Activation/physiology , ErbB Receptors/genetics , Ligands , Phosphorylation , Protein Binding , Receptors, Adrenergic, beta-2/metabolism , Transcriptional Activation
14.
J Biol Chem ; 275(3): 2239-45, 2000 Jan 21.
Article in English | MEDLINE | ID: mdl-10636931

ABSTRACT

The dermatonecrotic toxin produced by Pasteurella multocida is one of the most potent mitogenic substances known for fibroblasts in vitro. Exposure to recombinant P. multocida toxin (rPMT) causes phospholipase C-mediated hydrolysis of inositol phospholipids, calcium mobilization, and activation of protein kinase C via a poorly characterized mechanism involving G(q/11) family heterotrimeric G proteins. To determine whether the regulation of G protein pathways contributes to the mitogenic effects of rPMT, we have examined the mechanism whereby rPMT stimulates the Erk mitogen-activated protein kinase cascade in cultured HEK-293 cells. Treatment with rPMT resulted in a dose and time-dependent increase in Erk 1/2 phosphorylation that paralleled its stimulation of inositol phospholipid hydrolysis. Both rPMT- and alpha-thrombin receptor- stimulated Erk phosphorylation were selectively blocked by cellular expression of two peptide inhibitors of G(q/11) signaling, the dominant negative mutant G protein-coupled receptor kinase, GRK2(K220R), and the Galpha(q) carboxyl-terminal peptide, Galpha(q)-(305-359). Like alpha-thrombin receptor-mediated Erk activation, the effect of rPMT was insensitive to the protein kinase C inhibitor GF109203X, but was blocked by the epidermal growth factor receptor-specific tyrphostin, AG1478 and by dominant negative mutants of mSos1 and Ha-Ras. These data indicate that rPMT employs G(q/11) family heterotrimeric G proteins to induce Ras-dependent Erk activation via protein kinase C-independent "transactivation" of the epidermal growth factor receptor.


Subject(s)
Bacterial Proteins , Bacterial Toxins/metabolism , Enzyme Activation , ErbB Receptors/metabolism , GTP-Binding Proteins/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogens/metabolism , Cell Division/drug effects , Cell Line , Cyclic AMP-Dependent Protein Kinases/pharmacology , Dose-Response Relationship, Drug , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Gene Expression Regulation, Enzymologic , Humans , Hydrolysis , Lysophospholipids/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Mutation , Phosphatidylinositols/metabolism , Receptors for Activated C Kinase , Receptors, Cell Surface/antagonists & inhibitors , Recombinant Proteins/metabolism , Thrombin/pharmacology , Time Factors , Transcriptional Activation , Transfection , Virulence Factors, Bordetella/metabolism , beta-Adrenergic Receptor Kinases
15.
J Biol Chem ; 275(15): 11312-9, 2000 Apr 14.
Article in English | MEDLINE | ID: mdl-10753943

ABSTRACT

beta-Arrestins can act as adapter molecules, coupling G-protein-coupled receptors to proteins involved in mitogenic as well as endocytic pathways. We have previously identified c-SRC as a molecule that is rapidly recruited to the beta2-adrenergic receptor in a beta-arrestin1-dependent manner. Recruitment of c-SRC to the receptor appears to be involved in pathways leading to receptor internalization and mitogen-activated protein kinase activation. This recruitment of c-SRC to the receptor involves an interaction between the amino-terminal proline-rich region of beta-arrestin1 and the Src homology 3 (SH3) domain of c-SRC, but deletion of the proline-rich domain does not totally ablate the interaction. We have found that a major interaction also exists between beta-arrestin1 and the catalytic or kinase domain (SH1) of c-SRC. We therefore hypothesized that a catalytically inactive mutant of the isolated catalytic subunit, SH1(kinase dead) (SH1(KD)), would specifically block those cellular actions of c-SRC that are mediated by beta-arrestin1 recruitment to the G-protein-coupled receptor. In contrast, the majority of cellular phosphorylations catalyzed by c-SRC, which do not involve interaction with the SH1 domain, would be predicted to be unaffected. The SH1(KD) mutant did indeed block beta2-adrenergic receptor internalization and receptor-stimulated tyrosine phosphorylation of dynamin, actions previously shown to be c-SRC-dependent. In contrast, SAM-68 and whole cell tyrosine phosphorylation by c-SRC was unaffected, indicating that the SH1(KD) mutant did not inhibit c-SRC tyrosine kinase activity in general. These results not only clarify the nature of the beta-arrestin1/c-SRC interaction but also implicate beta-arrestin1 as an important mediator of receptor internalization by recruiting tyrosine kinase activity to the cell surface to phosphorylate key endocytic intermediates, such as dynamin.


Subject(s)
Arrestins/physiology , Endocytosis , Protein-Tyrosine Kinases/physiology , Receptors, Adrenergic, beta-2/metabolism , CSK Tyrosine-Protein Kinase , Catalytic Domain , Cell Line , Dynamins , ErbB Receptors/physiology , GTP Phosphohydrolases/metabolism , Humans , Phosphorylation , Protein-Tyrosine Kinases/chemistry , Structure-Activity Relationship , Tyrosine/metabolism , beta-Arrestins , src Homology Domains , src-Family Kinases
16.
Virology ; 264(1): 159-66, 1999 Nov 10.
Article in English | MEDLINE | ID: mdl-10544141

ABSTRACT

The Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) interacts with the tumor necrosis factor receptor (TNFR)-associated factor (TRAF) molecules, which are important for LMP1-mediated signaling. Two domains of LMP1 can independently activate NF-kB, carboxyl-terminal activating region 1 (CTAR1) and CTAR2. The activation of NF-kB by CTAR1 occurs through direct interaction of LMP1 with the TRAF molecules, whereas CTAR2 interacts with the TNFR-associated death domain protein (TRADD) to activate NF-kB and the c-Jun N-terminal kinase (JNK). A20, which is induced by LMP1 through NF-kB, can block NF-kB activation from both domains of LMP1 and inhibit JNK activation from CTAR2. A20 also has been shown to associate with TRAF1 and TRAF2. In this study, an interaction between LMP1 and A20 was detected that was increased by TRAF2 overexpression. A20 did not affect the association of TRAF1 with TRAF2 but did displace TRAF1 from the LMP1 complex. The interaction of LMP1 and TRADD was decreased in the presence of A20, and the LMP1-A20 association was decreased by TRADD, suggesting that A20 and TRADD both interact with LMP1 and may compete for binding. These data indicate that A20 alters the interactions between LMP1 and the TRAF molecules and TRADD, affecting the activation of NF-kB, JNK, and perhaps other TRAF-mediated signaling events.


Subject(s)
Herpesvirus 4, Human/physiology , Proteins/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins , Viral Matrix Proteins/metabolism , Animals , Binding Sites , Carcinoma, Non-Small-Cell Lung , Cysteine Endopeptidases , DNA-Binding Proteins , Humans , Intracellular Signaling Peptides and Proteins , Lung Neoplasms , Mice , Models, Chemical , NF-kappa B/metabolism , Nuclear Proteins , Proteins/chemistry , Proteins/isolation & purification , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , TNF Receptor-Associated Death Domain Protein , TNF Receptor-Associated Factor 1 , Tumor Cells, Cultured , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/isolation & purification
17.
Trends Microbiol ; 7(11): 453-8, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10542425

ABSTRACT

The epidermal growth factor receptor (EGFR) is a potent stimulator of the mitogen-activated protein kinase (MAPK) signaling pathway. Chronic stimulation of the EGFR and of multiple steps in the MAPK signaling pathway is involved in the development of cancer. Several tumor viruses encode proteins that induce EGFR expression or stimulate EGFR-mediated signaling and are thus likely to play an important role in the transformation of virus-infected cells.


Subject(s)
Cell Transformation, Neoplastic , ErbB Receptors/metabolism , MAP Kinase Signaling System , Neoplasms/metabolism , Oncogene Proteins, Viral/metabolism , Oncogenic Viruses/metabolism , Animals , ErbB Receptors/genetics , Gene Expression , Humans , Oncogene Proteins, Viral/genetics , Oncogenic Viruses/genetics
18.
Proc Natl Acad Sci U S A ; 96(22): 12559-64, 1999 Oct 26.
Article in English | MEDLINE | ID: mdl-10535961

ABSTRACT

Several G-protein coupled receptors, such as the beta1-adrenergic receptor (beta1-AR), contain polyproline motifs within their intracellular domains. Such motifs in other proteins are known to mediate protein-protein interactions such as with Src homology (SH)3 domains. Accordingly, we used the proline-rich third intracellular loop of the beta1-AR either as a glutathione S-transferase fusion protein in biochemical "pull-down" assays or as bait in the yeast two-hybrid system to search for interacting proteins. Both approaches identified SH3p4/p8/p13 (also referred to as endophilin 1/2/3), a SH3 domain-containing protein family, as binding partners for the beta1-AR. In vitro and in human embryonic kidney (HEK) 293 cells, SH3p4 specifically binds to the third intracellular loop of the beta1-AR but not to that of the beta2-AR. Moreover, this interaction is mediated by the C-terminal SH3 domain of SH3p4. Functionally, overexpression of SH3p4 promotes agonist-induced internalization and modestly decreases the Gs coupling efficacy of beta1-ARs in HEK293 cells while having no effect on beta2-ARs. Thus, our studies demonstrate a role of the SH3p4/p8/p13 protein family in beta1-AR signaling and suggest that interaction between proline-rich motifs and SH3-containing proteins may represent a previously underappreciated aspect of G-protein coupled receptor signaling.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Receptors, Adrenergic, beta-1/metabolism , Adrenergic beta-1 Receptor Agonists , Animals , Cattle , Cell Line , GTP-Binding Protein alpha Subunits, Gs/metabolism , Humans , Proline/metabolism , Protein Binding , Receptors, Adrenergic, beta-1/chemistry , src Homology Domains
19.
J Toxicol Environ Health A ; 57(8): 529-41, 1999 Aug 27.
Article in English | MEDLINE | ID: mdl-10515572

ABSTRACT

Chrysotile fibers (NIEHS intermediate length) were treated with ultrapure HCl to alter the fiber surface chemistry without substantially changing fiber morphology or dimensions. The objective of the study was to determine whether fiber surface chemistry is an important variable in fiber genotoxicity in vitro. The modified fibers, along with native chrysotile fibers, were used to challenge Chinese hamster lung fibroblasts (V79) in vitro using the micronucleus induction genotoxicity assay. Fiber dimensions were assessed using scanning electron microscopy by measuring the distribution of fiber lengths in 3 length ranges: less than 3 microm, 3-10 microm, and greater than 10 microm. For both treated and native fiber samples, 500 fibers were examined. Results indicate that acid-treated fibers were about 20% shorter than untreated chrysotile. Surface chemistry alterations were verified by zeta-potential reversal, x-ray photoelectron spectroscopy (XPS), and scanning electron microscopy/energy-dispersive x-ray spectroscopy (SEM-EDS) elemental analysis. Scanning Auger spectrometry indicated the presence of Mg, O, and Si in both treated and native chrysotile samples, which confirmed the surface purity of both fiber samples. Both XPS and SEM-EDS analysis demonstrated substantial depletion of Mg from fiber surfaces. Results of the micronucleus assay showed a positive concentration-related response for both samples, with toxicity evident only at the highest concentration. No significant difference was found for the treated and untreated chrysotile samples. These results indicate that the surface chemistry is not an important variable in the in vitro genotoxicity of chrysotile asbestos in V79 cells as detected by the micronucleus assay under the conditions used in this study, and support a model of chemically nonspecific chromosomal and spindle damage effects.


Subject(s)
Asbestos, Serpentine/toxicity , DNA Damage/drug effects , Lung/drug effects , Mutagens/toxicity , Animals , Asbestos, Serpentine/chemistry , Cell Survival/drug effects , Cells, Cultured , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Electron Probe Microanalysis , Fibroblasts/drug effects , Fibroblasts/pathology , Lung/pathology , Magnesium/analysis , Micronuclei, Chromosome-Defective/drug effects , Micronucleus Tests , Microscopy, Electron, Scanning , Mutagens/chemistry , Silicon/analysis , Spectrometry, X-Ray Emission
20.
Stat Med ; 18(8): 975-87, 1999 Apr 30.
Article in English | MEDLINE | ID: mdl-10363335

ABSTRACT

Previous work has shown that cross-over trial data can be analysed using within-subject linear functions. Scores that result from linear functions are graphed in quantile comparison plots in order to visualize the differences between factor levels. An example suggests how this visualization can be used to identify outliers or to provide a more specific interpretation of results. Additional examples indicate how this approach can be used to track carry-over differences or interaction effects. This article is a US Government work and is in the public domain in the United States.


Subject(s)
Clinical Protocols , Cross-Over Studies , Linear Models , Animals , Blood Glucose/analysis , Blood Pressure , Humans , Hypertension/therapy , Insulin/pharmacology , Insulin/therapeutic use , Rabbits , Radiography, Thoracic/classification , Radiography, Thoracic/statistics & numerical data
SELECTION OF CITATIONS
SEARCH DETAIL