Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 287(50): 41706-19, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23095746

ABSTRACT

The gustatory system detects tastants and transmits signals to the brain regarding ingested substances and nutrients. Although tastant receptors and taste signaling pathways have been identified, little is known about their regulation. Because bitter, sweet, and umami taste receptors are G protein-coupled receptors (GPCRs), we hypothesized that regulators of G protein signaling (RGS) proteins may be involved. The recent cloning of RGS21 from taste bud cells has implicated this protein in the regulation of taste signaling; however, the exact role of RGS21 has not been precisely defined. Here, we sought to determine the role of RGS21 in tastant responsiveness. Biochemical analyses confirmed in silico predictions that RGS21 acts as a GTPase-accelerating protein (GAP) for multiple G protein α subunits, including adenylyl cyclase-inhibitory (Gα(i)) subunits and those thought to be involved in tastant signal transduction. Using a combination of in situ hybridization, RT-PCR, immunohistochemistry, and immunofluorescence, we demonstrate that RGS21 is not only endogenously expressed in mouse taste buds but also in lung airway epithelial cells, which have previously been shown to express components of the taste signaling cascade. Furthermore, as shown by reverse transcription-PCR, the immortalized human airway cell line 16HBE was found to express transcripts for tastant receptors, RGS21, and downstream taste signaling components. Over- and underexpression of RGS21 in 16HBE cells confirmed that RGS21 acts to oppose bitter tastant signaling to cAMP and calcium second messenger changes. Our data collectively suggests that RGS21 modulates bitter taste signal transduction.


Subject(s)
Calcium Signaling/physiology , Cyclic AMP/metabolism , GTP-Binding Protein Regulators/biosynthesis , Respiratory Mucosa/metabolism , Taste Buds/metabolism , Taste/physiology , Animals , COS Cells , Calcium/metabolism , Chlorocebus aethiops , Cyclic AMP/genetics , GTP-Binding Protein Regulators/genetics , Humans , Mice , Mice, Transgenic , RGS Proteins , Respiratory Mucosa/cytology , Reverse Transcriptase Polymerase Chain Reaction , Taste Buds/cytology
2.
Ultrasound Med Biol ; 36(4): 677-92, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20350691

ABSTRACT

Acoustic cavitation-mediated wounding (i.e., sonoporation) has great potential to improve medical and laboratory applications requiring intracellular uptake of exogenous molecules; however, the field lacks detailed understanding of cavitation-induced morphologic changes in cells and their relative importance. Here, we present an in-depth study of the effects of acoustic cavitation on cells using electron and confocal microscopy coupled with quantitative flow cytometry. High resolution images of treated cells show that morphologically different types of blebs can occur after wounding conditions caused by ultrasound exposure as well as by mechanical shear and strong laser ablation. In addition, these treatments caused wound-induced nonlytic necrotic death resulting in cell bodies we call wound-derived perikarya (WD-P). However, only cells exposed to acoustic cavitation experienced ejection of intact nuclei and nearly instant lytic necrosis. Quantitative analysis by flow cytometry indicates that wound-derived perikarya are the dominant morphology of nonviable cells, except at the strongest wounding conditions, where nuclear ejection accounts for a significant portion of cell death after ultrasound exposure.


Subject(s)
Cell Membrane/pathology , Cell Membrane/radiation effects , Prostatic Neoplasms/pathology , Sonication/methods , Cell Line , Cell Size/radiation effects , Humans , Male , Radiation Dosage
3.
J Cell Physiol ; 223(3): 746-56, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20205236

ABSTRACT

Actin-myosin contractility modulates focal adhesion assembly, stress fiber formation, and cell migration. We analyzed the contributions of contractility to fibroblast adhesion strengthening using a hydrodynamic adhesion assay and micropatterned substrates to control cell shape and adhesive area. Serum addition resulted in adhesion strengthening to levels 30-40% higher than serum-free cultures. Inhibition of myosin light chain kinase or Rho-kinase blocked phosphorylation of myosin light chain to similar extents and eliminated the serum-induced enhancements in strengthening. Blebbistatin-induced inhibition of myosin II reduced serum-induced adhesion strength to similar levels as those obtained by blocking myosin light chain phosphorylation. Reductions in adhesion strengthening by inhibitors of contractility correlated with loss of vinculin and talin from focal adhesions without changes in integrin binding. In vinculin-null cells, inhibition of contractility did not alter adhesive force, whereas controls displayed a 20% reduction in adhesion strength, indicating that the effects of contractility on adhesive force are vinculin-dependent. Furthermore, in cells expressing FAK, inhibitors of contractility reduced serum-induced adhesion strengthening as well as eliminated focal adhesion assembly. In contrast, in the absence of FAK, these inhibitors did not alter adhesion strength or focal adhesion assembly. These results indicate that contractility modulates adhesion strengthening via FAK-dependent, vinculin-containing focal adhesion assembly.


Subject(s)
Fibroblasts/cytology , Fibroblasts/enzymology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/enzymology , Vinculin/metabolism , Actins/metabolism , Animals , Cell Adhesion , Cell Movement , Humans , Integrins/metabolism , Mice , Myosin Light Chains/metabolism , Myosins/metabolism , NIH 3T3 Cells , Phosphorylation , Protein Binding , Talin/metabolism , Vinculin/deficiency , rho-Associated Kinases/metabolism
4.
J Biomol Screen ; 15(3): 251-60, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20150590

ABSTRACT

Many G-protein-coupled receptors (GPCRs) have been shown to form heteromeric complexes primarily by biochemical methods, including competitive radioligand binding assays or measurements of changes in second-messenger concentration in lysed cells. These results are often cell line specific, and the expression of other cell surface proteins makes it difficult to detect potential functional consequences of GPCR interaction. Here, 2-electrode voltage clamping in Xenopus oocytes was used as a bioassay to explore heterodimerization of bradykinin type 2 receptor (Bk2R) and beta 2 adrenergic receptor (beta(2)AR), using chloride channels as outputs for receptor activation. The data show for the first time that these 2 receptors heterodimerize with functional consequences. Stimulation with bradykinin induced activation of Galphaq- and transactivation of Galphas-coupled pathways in oocytes expressing Bk2R and beta(2)AR. To corroborate these data, potential receptor interaction was examined in PC12 cells, a cell line that endogenously expresses both receptors, and confirmed that stimulation with bradykinin transactivates beta(2)AR. In both oocytes and PC12 cells, transactivation was ablated by Bk2R or beta(2)AR inverse agonists, suggesting that transactivation occurred directly through both receptors. This is the first evidence of Bk2R/beta(2)AR physical interaction, forming a functional heterodimer. The oocyte system may prove highly useful for exploration of GPCR heterodimerization and the functional consequences thereof.


Subject(s)
Biological Assay/methods , Protein Multimerization , Receptors, Adrenergic, beta-2/genetics , Receptors, Bradykinin/metabolism , Transcriptional Activation/genetics , Animals , CHO Cells , Cricetinae , Cricetulus , PC12 Cells , Protein Binding/drug effects , Protein Conformation , Protein Multimerization/drug effects , Rats , Receptor Cross-Talk/drug effects , Receptor, Serotonin, 5-HT2C/metabolism , Receptors, Adrenergic, beta-2/chemistry , Receptors, Bradykinin/chemistry , Signal Transduction/drug effects , Terbutaline/pharmacology , Transcriptional Activation/drug effects , Xenopus
5.
Biochim Biophys Acta ; 1791(8): 746-56, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19362163

ABSTRACT

Serine palmitoyltransferase (SPT) has been localized to the endoplasmic reticulum (ER) by subcellular fractionation and enzymatic assays, and fluorescence microscopy of epitope-tagged SPT; however, our studies have suggested that SPT subunit 1 might be present also in focal adhesions and the nucleus. These additional locations have been confirmed by confocal microscopy using HEK293 and HeLa cells, and for focal adhesions by the demonstration that SPT1 co-immunoprecipitates with vinculin, a focal adhesion marker protein. The focal adhesion localization of SPT1 is associated with cell morphology, and possibly cell migration, because it is seen in most cells before they reach confluence but disappears when they become confluent, and is restored by a standard scratch-wound healing assay. Conversely, elimination of SPT1 using SPTLC1 siRNA causes cell rounding. Thus, in addition to its "traditional" localization in the ER for de novo sphingolipid biosynthesis, SPT1 is present in other cellular compartments, including focal adhesions where it is associated with cell morphology.


Subject(s)
Cell Nucleus/enzymology , Cell Shape , Endoplasmic Reticulum/enzymology , Focal Adhesions/enzymology , Protein Subunits/metabolism , Serine C-Palmitoyltransferase/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Cell Adhesion , Cell Line , Cell Membrane/enzymology , Gene Silencing , Humans , Immunoprecipitation , Protein Transport , RNA, Small Interfering/metabolism , Reproducibility of Results , Sphingolipids/metabolism , Subcellular Fractions/enzymology , Vinculin/metabolism
6.
Biochim Biophys Acta ; 1781(9): 547-57, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18501205

ABSTRACT

Since the molecular cloning of the vzg-1/Edg-2/LPA1 gene, studies have attempted to characterize LPA1 receptor functionality into a single categorical role, different from the other Edg-family LPA receptors. The desire to categorize LPA1 function has highlighted its complexity and demonstrated that the LPA1 receptor does not have one absolute function throughout every system. The central nervous system is highly enriched in the LPA1 receptor, suggesting an integral role in neuronal processes. Metastatic and invasive breast cancer also appears to have LPA-mediated LPA1 receptor functions that enhance phenotypes associated with tumorigenesis. LPA1 possesses a number of motifs conserved among G protein-coupled receptors (GPCRs): a DRY-like motif, a PDZ domain, Ser/Thr predicted sites of phosphorylation, a di-leucine motif, double cysteines in the tail and conserved residues that stabilize structure and determine ligand binding. The third intracellular loop of the LPA1 receptor may be the crux of receptor signaling and attenuation with phosphorylation of Thr-236 potentially a key determinant of basal LPA1 signaling. Mutagenesis data supports the notion that Thr-236 regulates this process since mutating Thr-236 to Ala-236 increased basal and LPA-mediated serum response factor (SRF) signaling activity and Lys-236 further increased this basal signaling. Here we describe progress on defining the major functions of the LPA1 receptor, discuss a context dependent dualistic role as both a negative regulator in cancer and a proto-oncogene, outline its structural components at the molecular amino acid level and present mutagenesis data on the third intracellular loop of the receptor.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Receptors, Lysophosphatidic Acid/chemistry , Receptors, Lysophosphatidic Acid/metabolism , Animals , Central Nervous System/metabolism , Disease Progression , Humans , Neoplasms/genetics , Neoplasms/pathology , Proto-Oncogene Mas , Receptors, Lysophosphatidic Acid/genetics , Signal Transduction
7.
J Biol Chem ; 283(9): 5249-57, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18089565

ABSTRACT

Lysophosphatidic acid (LPA) stimulates cells by activation of five G-protein-coupled receptors, termed LPA 1-5. The LPA 1 receptor is the most widely expressed and is a major regulator of cell migration. In this study, we show that phorbol ester (PMA)-induced internalization of the LPA(1) receptor requires clathrin AP-2 complexes, protein kinase C, and a distal dileucine motif (amino acids 352 and 353) in the cytoplasmic tail but not beta-arrestin. Agonist-dependent internalization of LPA 1, however, requires a cluster of serine residues (amino acids 341-347) located proximal to the dileucine motif, beta-arrestin, and to a lesser extent clathrin AP-2. The serine cluster of LPA 1 is required for beta-arrestin2-GFP translocation to the plasma membrane and signal desensitization. In contrast, the dileucine motif (IL) is required for both basal and PMA-induced internalization. Evidence for the beta-arrestin independence of PMA-induced internalization of LPA 1 comes from the observations that beta-arrestin2-GFP is not recruited to the plasma membrane upon PMA treatment and that LPA 1 is readily internalized in beta-arrestin1/2 knock-out mouse embryonic fibroblasts. These results indicate that distinct molecular mechanisms regulate agonist-dependent and PMA-dependent internalization of the LPA 1 receptor.


Subject(s)
Carcinogens/pharmacology , Lysophospholipids/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Adaptor Protein Complex 2/genetics , Adaptor Protein Complex 2/metabolism , Amino Acid Motifs/physiology , Animals , Arrestins/genetics , Arrestins/metabolism , Cell Movement/drug effects , Cell Movement/physiology , Clathrin/genetics , Clathrin/metabolism , HeLa Cells , Humans , Mice , Mice, Knockout , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Transport/drug effects , Protein Transport/physiology , Receptors, Lysophosphatidic Acid/agonists , Receptors, Lysophosphatidic Acid/genetics , beta-Arrestins
8.
Mol Cancer Res ; 5(11): 1201-11, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18025263

ABSTRACT

Lysophosphatidic acid (LPA) is a bioactive lipid that promotes cancer cell proliferation and motility through activation of cell surface G protein-coupled receptors. Here, we provide the first evidence that LPA reduces the cellular abundance of the tumor suppressor p53 in A549 lung carcinoma cells, which express endogenous LPA receptors. The LPA effect depends on increased proteasomal degradation of p53 and it results in a corresponding decrease in p53-mediated transcription. Inhibition of phosphatidylinositol 3-kinase protected cells from the LPA-induced reduction of p53, which implicates this signaling pathway in the mechanism of LPA-induced loss of p53. LPA partially protected A549 cells from actinomycin D induction of both apoptosis and increased p53 abundance. Expression of LPA(1), LPA(2), and LPA(3) receptors in HepG2 hepatoma cells, which normally do not respond to LPA, also decreased p53 expression and p53-dependent transcription. In contrast, neither inactive LPA(1) (R124A) nor another G(i)-coupled receptor, the M(2) muscarinic acetylcholine receptor, reduced p53-dependent transcription in HepG2 cells. These results identify p53 as a target of LPA action and provide a new dimension for understanding how LPA stimulates cancer cell division, protects against apoptosis, and thereby promotes tumor progression.


Subject(s)
Lysophospholipids/toxicity , Mitogens/toxicity , Neoplasms/pathology , Tumor Suppressor Protein p53/antagonists & inhibitors , Apoptosis/genetics , Carcinoma , Cell Line, Tumor , Cell Nucleus/chemistry , Cell Nucleus/metabolism , DNA Damage , Humans , Lung Neoplasms , Neoplasms/genetics , Neoplasms/metabolism , Receptors, Lysophosphatidic Acid/agonists , Receptors, Lysophosphatidic Acid/metabolism , Transcription, Genetic/drug effects , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/metabolism
9.
Ultrasound Med Biol ; 32(6): 915-24, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16785013

ABSTRACT

Using conditions different from conventional medical imaging or laboratory cell lysis, ultrasound has recently been shown to reversibly increase plasma membrane permeability to drugs, proteins and DNA in living cells and animals independently of cell or drug type, suggesting a ubiquitous mechanism of action. To determine the mechanism of these effects, we examined cells exposed to ultrasound by flow cytometry coupled with electron and fluorescence microscopies. The results show that cavitation generated by ultrasound facilitates cellular incorporation of macromolecules up to 28 nm in radius through repairable micron-scale disruptions in the plasma membrane with lifetimes >1 min, which is a period similar to the kinetics of membrane repair after mechanical wounding. Further data suggest that cells actively reseal these holes using a native healing response involving endogenous vesicle-based membrane resealing. In this way, noninvasively focused ultrasound could deliver drugs and genes to targeted tissues, thereby minimizing side effects, lowering drug dosages, and improving efficacy.


Subject(s)
Drug Delivery Systems/methods , Electroporation/methods , Sonication , Acoustics , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Membrane Permeability , Endocytosis , Erythrocyte Membrane/metabolism , Humans , Male , Microscopy, Electron, Scanning , Tumor Cells, Cultured
10.
J Cell Sci ; 118(Pt 22): 5291-304, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16263766

ABSTRACT

Lysophosphatidic acid (LPA) stimulates heterotrimeric G protein signaling by activating three closely related receptors, termed LPA(1), LPA(2) and LPA(3). Here we show that in addition to promoting LPA(1) signaling, membrane cholesterol is essential for the association of LPA(1) with beta-arrestin, which leads to signal attenuation and clathrin-dependent endocytosis of LPA(1). Reduction of clathrin heavy chain expression, using small interfering RNAs, inhibited LPA(1) endocytosis. LPA(1) endocytosis was also inhibited in beta-arrestin 1 and 2-null mouse embryo fibroblasts (beta-arrestin 1/2 KO MEFs), but was restored upon re-expression of wild-type beta-arrestin 2. beta-arrestin attenuates LPA signaling as LPA(1)-dependent phosphoinositide hydrolysis was significantly elevated in beta-arrestin 1/2 KO MEFs and was reduced to wild-type levels upon re-expression of wild-type beta-arrestin. Interestingly, extraction of membrane cholesterol with methyl-beta-cyclodextrin inhibited LPA(1) signaling, beta-arrestin membrane recruitment and LPA(1) endocytosis. Cholesterol repletion restored all of these functions. However, neither the stimulation of phosphoinositide hydrolysis by the M(1) acetylcholine receptor nor its endocytosis was affected by cholesterol extraction. LPA treatment increased the detergent resistance of LPA(1) and this was inhibited by cholesterol extraction, suggesting that LPA(1) localizes to detergent-resistant membranes upon ligand stimulation. These data indicate that although LPA(1) is internalized by clathrin- and beta-arrestin dependent endocytosis, membrane cholesterol is critical for LPA(1) signaling, membrane recruitment of beta-arrestins and LPA(1) endocytosis.


Subject(s)
Arrestins/metabolism , Cell Membrane/metabolism , Cholesterol/metabolism , Clathrin/metabolism , Endocytosis , Receptors, Lysophosphatidic Acid/metabolism , Animals , Arrestins/deficiency , Cytosol/metabolism , Detergents/pharmacology , Endocytosis/drug effects , Fibroblasts/cytology , HeLa Cells , Humans , Hydrolysis/drug effects , Lysophospholipids/agonists , Lysophospholipids/pharmacology , Membrane Microdomains/drug effects , Mice , Phosphatidylinositols/metabolism , Receptors, Lysophosphatidic Acid/agonists , Receptors, Muscarinic/metabolism , Signal Transduction , beta-Arrestin 1 , beta-Arrestin 2 , beta-Arrestins
11.
Biomaterials ; 26(30): 5972-82, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15890400

ABSTRACT

In this study, we quantified the adsorption of immunoglobulin G (IgG) protein onto several polyelectrolyte-modified sintered porous polyethylene (PPE) membranes. The polymer surfaces had both cationic and anionic charges obtained via the adsorption of polyethylenimine (PEI) and polyacrylic acid (PAA), respectively, onto plasma-activated PPE. The amount of IgG adsorption was determined by measuring the gamma radiation emitted by [125I]-IgG radio labeled protein. By studying the impact of pH and ionic strength on IgG adsorption, we attempted to characterize the role and nature of the electrostatic interactions involved in the adsorption process to better understand how these interactions were influenced by the charge and structure of immobilized polyelectrolyte complexes at modified membrane surfaces. We were able to show that surface modification of PPE membranes with adsorbed PEI monolayers and PEI-PAA bilayers can greatly improve the IgG binding ability of the membrane under optimized conditions. We also showed that the observed improvement in the IgG binding is derived from electrostatic interactions between IgG and the polyelectrolyte surface. In addition, we found that the greatest IgG adsorption occurred when the IgG and the surface possessed predominantly opposite charges, rather than when the surface possessed the greatest electrostatic charge. Finally, we have found that the molecular weight of the terminating polyelectrolyte has a noticeable effect upon the electrostatic interactions between IgG and the PEI-PAA bilayer-modified PPE surfaces.


Subject(s)
Biocompatible Materials/chemistry , Polyethylene/chemistry , Proteins/chemistry , Acrylic Resins/chemistry , Adsorption , Anions , Cations , Electrolytes/chemistry , Gamma Rays , Hydrogen-Ion Concentration , Immunoglobulin G/chemistry , Ions , Lipid Bilayers , Macromolecular Substances/chemistry , Materials Testing , Molecular Weight , Polyethyleneimine/chemistry , Polyethylenes , Polymers/chemistry , Protein Binding , Static Electricity , Surface Properties
12.
Cell Signal ; 16(10): 1141-51, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15240009

ABSTRACT

Lysophosphatidic acid (LPA; 1-acyl-2-hydroxy-sn-glycero-3-phosphate) is a lipid growth factor that stimulates the proliferation of ovarian cancer cells. Recent studies indicate that elevation of cellular cAMP levels inhibits ovarian epithelial cancer cell growth. In this study, we investigated the effects of elevating cellular cAMP levels on LPA stimulation of OVCAR-3 ovarian cancer cell growth and on LPA stimulation of the serum response factor (SRF) transcription factor. Treatment of OVCAR-3 cells with forskolin and isobutylmethylxanthine (IBMX; 3-Isobutyl-1-methylxanthine) inhibited LPA stimulation of growth. LPA stimulation of SRF-mediated transcription was also inhibited in OVCAR-3 cells that were incubated with forskolin, dibutyryl cyclic AMP (db-cAMP), or paired cAMP analogues (N(6)-mono-tert-butylcarbamoyladenosine-3', 5'-cyclic monophosphate [6-MBC-cAMP] and Sp-5,6-DCl-BIMPS), which selectively activate type II protein kinase A. In contrast, incubation with a cAMP analogue (8-(4-chloro-phenylthio)-2'-O-methyadenosine-3',5'-cyclic monophosphate [8CPT-2Me-cAMP]) that specifically activates the cAMP inducible Rap1 exchange factor, Epac, did not inhibit SRF. Similar results were obtained when HepG2 hepatoma cells, which do not express endogenous LPA receptors, were transfected with a single LPA receptor (LPA(1)). We observed that treatment of OVCAR-3 cells with forskolin greatly reduced both F-actin staining and focal adhesion labeling with anti-paxillin antibodies. Treatment of OVCAR-3 cells with the F-actin stabilizing compound, jasplakinolide, prevented the protein kinase A (PKA)-mediated inhibition of SRF. These results suggest that PKA inhibits LPA stimulation of SRF by promoting the dissolution of F-actin and that this is likely to contribute to the cAMP-mediated inhibition of ovarian cancer cell growth.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Cytoskeleton/metabolism , Lysophospholipids/pharmacology , Serum Response Factor/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Carcinoma, Hepatocellular/drug therapy , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Proliferation/drug effects , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinase Type II , Cytoskeletal Proteins/metabolism , Cytoskeleton/drug effects , Female , Humans , Ovarian Neoplasms/drug therapy , Paxillin , Phosphoproteins/metabolism , Tumor Cells, Cultured
13.
J Cell Sci ; 116(Pt 10): 1969-80, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12668728

ABSTRACT

Lysophosphatidic acid (LPA) is a serum-borne phospholipid that exerts a pleiotropic range of effects on cells through activation of three closely related G-protein-coupled receptors termed LPA1/EDG-2, LPA2/EDG-4 and LPA3/EDG-7. Of these receptors, the LPA1 receptor is the most widely expressed. In this study, we investigated the agonist-induced endocytosis of the human LPA1 receptor, bearing an N-terminal FLAG epitope tag, in stably transfected HeLa cells. Treatment with LPA induced the rapid endocytosis of approximately 40% of surface LPA1 within 15 minutes. Internalization was both dose dependent and LPA specific since neither lysophophatidylcholine nor sphingosine-1-phosphate induced LPA1 endocytosis. Removal of agonist following 30 minutes incubation resulted in recycling of LPA1 back to the cell surface. LPA1 internalization was strongly inhibited by dominant-inhibitory mutants of both dynamin2 (K44A) and Rab5a (S34N). In addition, both dynamin2 K44A and Rab5 S34N mildly inhibited LPA1-dependent activation of serum response factor. Finally, our results also indicate that LPA1 exhibits basal, LPA-dependent internalization in the presence of serum-containing medium.


Subject(s)
Dynamin II/metabolism , Endocytosis , Lysophospholipids/metabolism , Receptors, G-Protein-Coupled/metabolism , rab5 GTP-Binding Proteins/metabolism , DNA/metabolism , Dose-Response Relationship, Drug , Epitopes , Fluorescent Antibody Technique, Indirect , HeLa Cells , Humans , Immunoblotting , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/agonists , Receptors, Lysophosphatidic Acid , Serum Response Factor/metabolism , Time Factors , Transfection
14.
J Biol Chem ; 277(36): 33439-46, 2002 Sep 06.
Article in English | MEDLINE | ID: mdl-12093817

ABSTRACT

Upon agonist stimulation, many G protein-coupled receptors such as beta(2)-adrenergic receptors are internalized via beta-arrestin- and clathrin-dependent mechanisms, whereas others, like M(2) muscarinic acetylcholine receptors (mAChRs), are internalized by clathrin- and arrestin-independent mechanisms. To gain further insight into the mechanisms that regulate M(2) mAChR endocytosis, we investigated the post-endocytic trafficking of M(2) mAChRs in HeLa cells and the role of the ADP-ribosylation factor 6 (Arf6) GTPase in regulating M(2) mAChR internalization. Here, we report that M(2) mAChRs are rapidly internalized by a clathrin-independent pathway that is inhibited up to 50% by expression of either GTPase-defective Arf6 Q67L or an upstream Arf6 activator, Galpha(q) Q209L. In contrast, M(2) mAChR internalization was not affected by expression of dominant-negative dynamin 2 K44A, which is a known inhibitor of clathrin-dependent endocytosis. Nevertheless, M(2) mAChRs, which are initially internalized in structures that lack clathrin-dependent endosomal markers, quickly localize to endosomes that contain the clathrin-dependent, early endosomal markers early endosome autoantigen-1, transferrin receptor, and GTPase-defective Rab5 Q79L, which is known to swell early endosomal compartments. These results suggest that M(2) mAChRs initially internalize via an Arf6-associated, clathrin-independent pathway but then quickly merge with the clathrin endocytic pathway at the level of early endosomes.


Subject(s)
Clathrin/metabolism , Endocytosis , Endosomes/metabolism , Receptors, Muscarinic/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Cell Membrane/metabolism , Cells, Cultured , Clathrin/physiology , DNA/metabolism , Dynamin I , Dynamins , Fluorescent Antibody Technique, Indirect , GTP Phosphohydrolases/metabolism , Genes, Dominant , Green Fluorescent Proteins , HeLa Cells , Humans , Kinetics , Luminescent Proteins/metabolism , Membrane Proteins/metabolism , Protein Transport , Receptor, Muscarinic M2 , Receptors, Transferrin/metabolism , Time Factors , Transfection , Vesicular Transport Proteins
15.
Curr Protoc Cell Biol ; Chapter 7: 7.7.1-7.7.6, 2001 Nov.
Article in English | MEDLINE | ID: mdl-18228385

ABSTRACT

The genetics of Drosophila is a powerful tool in the analysis of mutants and mutant proteins. Cultures of cells derived from wild-type or mutant flies can be pulse labeled to biosynthetically label the proteins made by the cells. Immunoprecipitation (and subcellular fractionation) are used to characterize the expression of specific proteins.


Subject(s)
Clinical Laboratory Techniques , Drosophila Proteins/isolation & purification , Metabolism , Animals , Immunoprecipitation , Indicators and Reagents , Isotope Labeling , Research Design
SELECTION OF CITATIONS
SEARCH DETAIL
...