51
|
Borges FT, Michelacci YM, Aguiar JAK, Dalboni MA, Garófalo AS, Schor N. Characterization of glycosaminoglycans in tubular epithelial cells: calcium oxalate and oxalate ions effects. Kidney Int 2005; 68:1630-42. [PMID: 16164640 DOI: 10.1111/j.1523-1755.2005.00577.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND The interaction between tubular epithelial cells and calcium oxalate crystals or oxalate ions is a very precarious event in the lithogenesis. Urine contains ions, glycoproteins and glycosaminoglycans that inhibit the crystallization process and may protect the kidney against lithogenesis. We examined the effect of oxalate ions and calcium oxalate crystals upon the synthesis of glycosaminoglycans in distal [Madin-Darby canine kidney (MDCK)] and proximal (LLC-PK1) tubular cell lines. METHODS Glycosaminoglycan synthesis was analyzed by metabolic labeling with (35)S-sulfate and enzymatic digestion with specific mucopolysaccharidases. Cell death was assessed by fluorescent dyes and crystal endocytosis was analised by flow cytometry. RESULTS The main glycosaminoglycans synthesized by both cells were chondroitin sulfate and heparan sulfate most of them secreted to the culture medium or present at cellular surface. Exposition of MDCK cells to oxalate ions increased apoptosis rate and the incorporation of (35)S-sulfate in chondroitin sulfate and heparan sulfate, while calcium oxalate crystals were endocyted by LLC-PK1, induced necrotic cell death, and increased (35)S-sulfate incorporation in glycosaminoglycans. These effects seem to be specific and due to increased biosynthesis, since hydroxyapatite and other carboxylic acid did not induced cellular death or glycosaminoglycan synthesis and no changes in sulfation degree or molecular weight of glycosaminoglycans could be detected. Thapsigargin inhibited the glycosaminoglycan synthesis induced by calcium oxalate in LLC-PK1, suggesting that this effect was sensitive to the increase in cytosolic calcium. CONCLUSION Tubular cells may increase the synthesis of glycosaminoglycans to protect from the toxic insult of calcium oxalate crystals and oxalate ions, what could partially limit the lithogenesis.
Collapse
Affiliation(s)
- Fernanda T Borges
- Departamento de Medicina, Disciplina de Nefrologia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brasil
| | | | | | | | | | | |
Collapse
|
52
|
Activation of heterotrimeric G-proteins independent of a G-protein coupled receptor and the implications for signal processing. Rev Physiol Biochem Pharmacol 2005. [DOI: 10.1007/s10254-005-0042-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
53
|
Le-Niculescu H, Niesman I, Fischer T, DeVries L, Farquhar MG. Identification and Characterization of GIV, a Novel Gαi/s -interacting Protein Found on COPI, Endoplasmic Reticulum-Golgi Transport Vesicles. J Biol Chem 2005; 280:22012-20. [PMID: 15749703 DOI: 10.1074/jbc.m501833200] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In this report, we characterize GIV (Galpha-interacting vesicle-associated protein), a novel protein that binds members of the Galpha(i) and Galpha subfamilies of heterotrimeric G proteins. The Galpha(s) interaction site was mapped to an 83-amino acid region of GIV that is enriched in highly charged amino acids. BLAST searches revealed two additional mammalian family members, Daple and an uncharacterized protein, FLJ00354. These family members share the highest homology at the Galpha binding domain, are homologous at the N terminus and central coiled coil domain but diverge at the C terminus. Using affinity-purified IgG made against two different regions of the protein, we localized GIV to COPI, endoplasmic reticulum (ER)-Golgi transport vesicles concentrated in the Golgi region in GH3 pituitary cells and COS7 cells. Identification as COPI vesicles was based on colocalization with beta-COP, a marker for these vesicles. GIV also codistributes in the Golgi region with endogenous calnuc and the KDEL receptor, which are cis Golgi markers and with Galpha(i3)-yellow fluorescent protein expressed in COS7 cells. By immunoelectron microscopy, GIV colocalizes with beta-COP and Galpha(i3) on vesicles found in close proximity to ER exit sites and to cis Golgi cisternae. In cell fractions prepared from rat liver, GIV is concentrated in a carrier vesicle fraction (CV2) enriched in ER-Golgi transport vesicles. beta-COP and several Galpha subunits (Galpha(i1-3), Galpha(s)) are also most enriched in CV2. Our results demonstrate the existence of a novel Galpha-interacting protein associated with COPI transport vesicles that may play a role in Galpha-mediated effects on vesicle trafficking within the Golgi and/or between the ER and the Golgi.
Collapse
Affiliation(s)
- Helen Le-Niculescu
- Department of Cellular and Molecular Medicine, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | | | | | | | | |
Collapse
|
54
|
Affiliation(s)
- W E Balch
- Department of Cellular Biology, The Scripps Research Institute, 10666 N. Torrey Pines Road, La Jolla, California 92037, USA
| |
Collapse
|
55
|
Affiliation(s)
- P Melançon
- Department of Chemistry and Biochemistry, University of Colorado at Boulder, Boulder, Colorado 80309-0215, USA
| |
Collapse
|
56
|
Meijer AJ, Codogno P. Regulation and role of autophagy in mammalian cells. Int J Biochem Cell Biol 2005; 36:2445-62. [PMID: 15325584 DOI: 10.1016/j.biocel.2004.02.002] [Citation(s) in RCA: 443] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2003] [Revised: 01/16/2004] [Accepted: 02/05/2004] [Indexed: 01/25/2023]
Abstract
The recent period has witnessed progress in the understanding of the lysosomal autophagic pathway. The discovery of a family of genes conserved from yeast to humans, and involved in the formation of autophagosomes, has unraveled new protein-conjugation systems and has shed light on the importance of autophagy in physiology and pathophysiology. The elucidation of the molecular control of autophagy will also lead to a better understanding of the role of autophagy during cell death. As a great number of extracellular stimuli (starvation, hormonal or therapeutic treatment) as well as intracellular stimuli (accumulation of misfolded proteins, invasion of microorganisms) is able to modulate the autophagic response, it is not surprising that several signaling pathways are involved in the control of autophagy. The mammalian Target of Rapamycin (mTOR) signaling pathway plays a major role in transmitting autophagic stimuli because of its ability to sense nutrient, metabolic and hormonal signals. In addition, autophagy, which is characterized by a flux of membrane from the formation of the autophagosome to the fusion with the lysosome, is regulated by GTPases, similarly to the vesicular transport along the exocytic/endocytic pathway. The aim of the present review is to give an overview of autophagy and to discuss its regulation by activators and effectors of mTOR and GTPases.
Collapse
Affiliation(s)
- Alfred J Meijer
- Department of Biochemistry, Academic Medical Center, University of Amsterdam, Meibergreef 15, 1105 AZ Amsterdam, The Netherlands
| | | |
Collapse
|
57
|
Robay A, Toumaniantz G, Leblais V, Gauthier C. Transfected β3- but Not β2-Adrenergic Receptors Regulate Cystic Fibrosis Transmembrane Conductance Regulator Activity via a New Pathway Involving the Mitogen-Activated Protein Kinases Extracellular Signal-Regulated Kinases. Mol Pharmacol 2004; 67:648-54. [PMID: 15563584 DOI: 10.1124/mol.104.002097] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have shown previously that in a heterologous mammalian expression system A549 cells, beta3-adrenoceptor (beta3-AR) stimulation regulates the activity of cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel. The present investigation was carried out to determine the signaling pathway involved in this regulation. A549 cells were intranuclearly injected with plasmids encoding human CFTR and beta3-AR. CFTR activity was functionally assessed by microcytofluorimetry. The application of 1 microM 4-[3-t-butylamino-2-hydroxypropoxy]benzimidazol-2-1 hydrochloride (CGP-12177), a beta3-AR agonist, produced a CFTR activation that was not abolished by protein kinase A inhibitors. In pertussis toxin-pretreated cells, the CFTR activation induced by CGP-12177 was abolished. The overexpression of beta-adrenoceptor receptor kinase, an inhibitor of betagamma subunits, abolished the CGP-12177-induced CFTR activation, suggesting the involvement of betagamma subunits of Gi/o proteins. The pretreatment of A549 cells with selective inhibitors of either phosphoinositide 3-kinase (PI3K), wortmannin, and 2-(4-morpholinyl)-8-phenyl-1(4H)-benzopyran-4-one hydrochloride (LY294002), or extracellular signal-regulated kinases 1 and 2 (ERK1/2) mitogen-activated protein kinase (MAPK), 2'-amino-3'-methoxyflavone (PD98059), and 1,4-diamino-2,3-dicyano-1,4-bis(2-aminophynyltio)butadiene (U0126), abolished the effects of CGP-12177 on the CFTR activity. Immunohistochemical assays showed that only the cells expressing beta3-AR exhibited MAPK activation in response to CGP-12177. Furthermore, CFTR activity increased in cells pretreated with 10% fetal bovine serum both in A549 cells injected only with CFTR and in T84 cells, which endogenously express CFTR, indicating that CFTR activity can be regulated by the MAPK independently of the beta3-AR stimulation. In conclusion, we have demonstrated that CFTR is regulated through a Gi/o/PI3K/ERK1/2 MAPK signaling cascade dependently or not on an activation of beta3-ARs. This pathway represents a new regulation for CFTR.
Collapse
Affiliation(s)
- Amal Robay
- Institut du Thorax, Institut National de la Sante et de la Recherche Medicale U533, Faculté de Médecine, 1 rue Gaston Veil, BP 53508, F-44035 Nantes, France
| | | | | | | |
Collapse
|
58
|
Cao X, Cismowski MJ, Sato M, Blumer JB, Lanier SM. Identification and characterization of AGS4: a protein containing three G-protein regulatory motifs that regulate the activation state of Gialpha. J Biol Chem 2004; 279:27567-74. [PMID: 15096500 DOI: 10.1074/jbc.m312786200] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activators of G-protein signaling 1-3 (AGS1-3) were identified in a functional screen of mammalian cDNAs that activated G-protein signaling in the absence of a receptor. We report the isolation and characterization of an additional AGS protein (AGS4) from a human prostate leiomyosarcoma cDNA library. AGS4 is identical to G18.1b, which is encoded by a gene within the major histocompatibility class III region of chromosome 6. The activity of AGS4 in the yeast-based functional screen was selective for G(i2)/G(i3) and independent of guanine-nucleotide exchange by G(i)alpha. RNA blots indicated enrichment of AGS4/G18.1b mRNA in heart, placenta, lung, and liver. Immunocytochemistry with AGS4/G18.1b-specific antisera indicated a predominant nonhomogeneous, extranuclear distribution within the cell following expression in COS7 or Chinese hamster ovary cells. AGS4/G18.1b contains three G-protein regulatory motifs downstream of an amino terminus domain with multiple prolines. Glutathione S-transferase (GST)-AGS4/G18.1b fusion proteins interacted with purified G(i)alpha, and peptides derived from each of the G-protein regulatory motifs inhibited guanosine 5'-3-O-(thio)triphosphate (GTPgammaS) binding to purified G(i)alpha(1). AGS4/G18.1b was also complexed with G(i)alpha(3) in COS7 cell lysates following cell transfection. However, AGS4/G18.1b did not alter the generation of inositol phosphates in COS7 cells cotransfected with the Gbetagamma-regulated effector phospholipase C-beta2. These data suggest either that an additional signal is required to position AGS4/G18.1b in the proper cellular location where it can access heterotrimer and promote subunit dissociation or that AGS4 serves as an alternative binding partner for G(i)alpha independent of Gbetagamma participating in G-protein signaling events that are independent of classical G-protein-coupled receptors at the cell surface.
Collapse
Affiliation(s)
- Xiaoqing Cao
- The Guthrie Research Institute, Sayre, Pennsylvania 18840, USA
| | | | | | | | | |
Collapse
|
59
|
Abstract
The trans-Golgi network is the major sorting compartment of the secretory pathway for protein, lipid and membrane traffic. There is a constant flow of membrane and cargo to and from this compartment. Evidence is emerging that the trans-Golgi network has multiple biochemically and functionally distinct subdomains, each of which contributes to the combined sorting and transport requirements of this dynamic compartment. The recruitment of distinct arrays of protein complexes to trans-Golgi network membranes is likely to produce the diversity of structure and biochemistry observed amongst subdomains that serve to generate different carriers or maintain resident trans-Golgi network components. This review discusses how these subdomains may be formed and examines the molecular players involved, including G proteins, clathrin adaptors and golgin tethers. Diversity within these protein families is highlighted and shown to be critical for the functionality of the trans-Golgi network, as a mediator of protein sorting and membrane transport, and for the maintenance of Golgi structure.
Collapse
Affiliation(s)
- Paul A Gleeson
- The Russell Grimwade School of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, Victoria 3010, Australia.
| | | | | | | |
Collapse
|
60
|
Chan D, Strang M, Judson B, Brown WJ. Inhibition of membrane tubule formation and trafficking by isotetrandrine, an antagonist of G-protein-regulated phospholipase A2 enzymes. Mol Biol Cell 2004; 15:1871-80. [PMID: 14767064 PMCID: PMC379283 DOI: 10.1091/mbc.e03-09-0644] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2003] [Revised: 11/30/2003] [Accepted: 01/11/2004] [Indexed: 11/11/2022] Open
Abstract
Previous studies have established a role for cytoplasmic phospholipase A(2) (PLA(2)) activity in tubule-mediated retrograde trafficking between the Golgi complex and the endoplasmic reticulum (ER). However, little else is known about how membrane tubule formation is regulated. This study demonstrates that isotetrandrine (ITD), a biscoclaurine alkaloid known to inhibit PLA(2) enzyme activation by heterotrimeric G-proteins, effectively prevented brefeldin A (BFA)-induced tubule formation from the Golgi complex and retrograde trafficking to the ER. In addition, ITD inhibited BFA-stimulated tubule formation from the trans-Golgi network and endosomes. ITD inhibition of the BFA response was potent (IC(50) approximately 10-20 microM) and rapid (complete inhibition with a 10-15-min preincubation). ITD also inhibited normal retrograde trafficking as revealed by the formation of nocodazole-induced Golgi mini-stacks at ER exit sites. Treatment of cells with ITD alone caused the normally interconnected Golgi ribbons to become fragmented and dilated, but cisternae were still stacked and located in a juxtanuclear position. These results suggest that a G-protein-binding PLA(2) enzyme plays a pivotal role in tubule mediated trafficking between the Golgi and the ER, the maintenance of the interconnected ribbons of Golgi stacks, and tubule formation from endosomes.
Collapse
Affiliation(s)
- Diane Chan
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 18483, USA
| | | | | | | |
Collapse
|
61
|
Sjölinder M, Uhlmann J, Ponstingl H. Characterisation of an evolutionary conserved protein interacting with the putative guanine nucleotide exchange factor DelGEF and modulating secretion. Exp Cell Res 2004; 294:68-76. [PMID: 14980502 DOI: 10.1016/j.yexcr.2003.09.033] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2003] [Indexed: 11/26/2022]
Abstract
A human cDNA library was screened for proteins interacting with the deafness locus putative guanine nucleotide exchange factor (DelGEF) using a yeast two-hybrid system. A protein with a predicted size of 9 kDa was identified as a binding partner, this protein was designated DelGEF interacting protein 1 (DelGIP1). The interaction between DelGEF and DelGIP1 was verified by co-immunoprecipitation of a DelGEF-DelGIP1 complex from cell lysates. Highly conserved homologues of DelGIP1 were identified in higher and lower eukaryotes by database searching. The human DelGIP1 gene is ubiquitously expressed as judged by human multiple tissue Northern blot analysis. DelGEF was recently shown to interact with Sec5, a protein involved in secretion, and to regulate secretion of proteoglycans. Downregulation of endogenous DelGIP1 in HeLa cells induced increased extracellular secretion of proteoglycans indicating a possible role for DelGIP1 in the secretion process.
Collapse
Affiliation(s)
- Mikael Sjölinder
- Division for Molecular Biology of Mitosis, German Cancer Research Center, D-69120 Heidelberg, Germany.
| | | | | |
Collapse
|
62
|
Abstract
One of the most exciting recent discoveries in the area of intracellular protein transport is the finding that many organelles involved in exocytic and endocytic membrane traffic have one or more Ras-like GTP-binding proteins on their cytoplasmic face that are specific for each membranous compartment. These proteins are attractive candidates for regulators of transport vesicle formation and the accurate delivery of transport vesicles to their correct targets.
Collapse
Affiliation(s)
- S R Pfeffer
- Deparment of Biochemistry, Standford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
63
|
Abstract
Among the proteins regulating vesicular traffic, the small, Ras-like GTPases have received particular attention. Several recent reports indicate that another class of GTP-binding (G) protein, the heterotrimeric G proteins, also participates in the regulation of vesicular traffic. Thus, studies using transfected cells and cell-free systems show that a pertussis toxin-sensitive trimeric G protein, G(i3), is involved in the formation of secretory vesicles from the Golgi complex. These results raise the intriguing possibility that signal transduction processes across intracellular membranes play a role in vesicle formation, and provide important clues about the molecular machinery involved in this process.
Collapse
Affiliation(s)
- F A Barr
- Institute for Neurobiology, University of Heidelberg, Im Neuenheimer Feld 364, D-6900 Heidelberg, Germany
| | | | | |
Collapse
|
64
|
Abstract
TGN38/41 is a heterodimeric integral membrane protein that cycles between the trans Golgi network and the cell surface. A tyrosine-containing tetrapeptide motif within its cytoplasmic tail is necessary and sufficient for determining its steady-state location in the TGN. Recent results have shown that TGN38/41 plays an essential role in the formation of exocytic vesicles at the TGN by serving as a receptor for complexes of a cytoplasmic protein known as p62, and one of four small GTP-binding proteins, including rab6. For budding to occur, this complex must bind to the cytoplasmic domain of TGN38/41. We propose here that TGN38/41 may couple the segregation of secretory proteins to the budding of exocytic vesicles at the TGN.
Collapse
Affiliation(s)
- K K Stanley
- Heart Research Institute, 145 Missenden Road, Camperdown, Sydney, NSW 2050, Australia
| | | |
Collapse
|
65
|
Pattingre S, Petiot A, Codogno P. Analyses of Galpha-interacting protein and activator of G-protein-signaling-3 functions in macroautophagy. Methods Enzymol 2004; 390:17-31. [PMID: 15488168 DOI: 10.1016/s0076-6879(04)90002-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Macroautophagy or autophagy is an ubiquitous and conserved degradative pathway of cytosolic components, macromolecules or organelles, into the lysosome. By using biochemical and microscopic methods, which allow one to measure the rate of autophagy, the role of two regulators of Gi3 protein activity, activator of G-protein-signaling-3 (AGS3) and Galpha-interacting protein (GAIP), was studied in the control of autophagy in human colon cancer HT-29 cells. In HT-29 cells, autophagy is under the control of the Gi3 protein and, when bound to the GTP, the Galphai3 protein inhibits autophagy, whereas it stimulates autophagy when bound to the GDP. GAIP, which enhances the intrinsic GTPase-activating protein activity of the Galphai3 protein, stimulates autophagy by favoring the GDP-bound form of Galphai3. We showed that GAIP is phosphorylated on its serine 151 and that this phosphorylation is dependent on the presence of amino acids that modulate Raf-1 activity, the kinase upstream of Erk1/2. AGS3, a guanine nucleotide dissociation inhibitor, stimulates autophagy by binding Galphai3 proteins. The intracellular localization of AGS3 (Golgi apparatus and endoplasmic reticulum, two membranes known to be at the origin of autophagosomes) is consistent with its role in autophagy.
Collapse
Affiliation(s)
- Sophie Pattingre
- Division of Infectious Diseases, Southwestern Medical School, Dallas, Texas 75390, USA
| | | | | |
Collapse
|
66
|
Bäck N, Litonius E, Mains RE, Eipper BA. Fluoride causes reversible dispersal of Golgi cisternae and matrix in neuroendocrine cells. Eur J Cell Biol 2004; 83:389-402. [PMID: 15506563 DOI: 10.1078/0171-9335-00405] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A role for heterotrimeric G proteins in the regulation of Golgi function and formation of secretory granules is generally accepted. We set out to study the effect of activation of heterotrimeric G proteins by aluminum fluoride on secretory granule formation in AtT-20 corticotropic tumor cells and in melanotrophs from the rat pituitary. In AtT-20 cells, treatment with aluminum fluoride or fluoride alone for 60 min induced complete dispersal of Golgi, ER-Golgi intermediate compartment and Golgi matrix markers, while betaCOP immunoreactiviy retained a juxtanuclear position and TGN38 was unaffected. Electron microscopy showed compression of Golgi cisternae followed by conversion of the Golgi stacks into clusters of tubular and vesicular elements. In the melanotroph of the rat pituitary a similar compression of Golgi cisternae was observed, followed by a progressive loss of cisternae from the stacks. As shown in other cells, brefeldin A induced redistribution of the Golgi matrix protein GM130 to punctate structures in the cytoplasm in AtT-20 cells, while mannosidase II immunoreactivity was completely dispersed. Fluoride induced a complete dispersal of mannosidase II and GM130 immunoreactivity. The effect of fluoride was fully reversible with reestablishment of normal mannosidase II and GM130 immunoreactivity within 2 h. After 1 h of recovery, showing varying stages of reassembly, the patterns of mannosidase II and GM130 immunoreactivity were identical in individual cells, indicating that Golgi matrix and cisternae reassemble with similar kinetics during recovery from fluoride treatment. Instead of a specific aluminum fluoride effect on secretory granule formation in the trans-Golgi network, we thus observe a unique form of Golgi dispersal induced by fluoride alone, possibly via its action as a phosphatase inhibitor.
Collapse
Affiliation(s)
- Nils Bäck
- Department of Anatomy, Institute of Biomedicine, Biomedicum Helsinki, University of Helsinki, Finland.
| | | | | | | |
Collapse
|
67
|
Kaushik R, Yu F, Chia W, Yang X, Bahri S. Subcellular localization of LGN during mitosis: evidence for its cortical localization in mitotic cell culture systems and its requirement for normal cell cycle progression. Mol Biol Cell 2003; 14:3144-55. [PMID: 12925752 PMCID: PMC181556 DOI: 10.1091/mbc.e03-04-0212] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Mammalian LGN/AGS3 proteins and their Drosophila Pins orthologue are cytoplasmic regulators of G-protein signaling. In Drosophila, Pins localizes to the lateral cortex of polarized epithelial cells and to the apical cortex of neuroblasts where it plays important roles in their asymmetric division. Using overexpression studies in different cell line systems, we demonstrate here that, like Drosophila Pins, LGN can exhibit enriched localization at the cell cortex, depending on the cell cycle and the culture system used. We find that in WISH, PC12, and NRK but not COS cells, LGN is largely directed to the cell cortex during mitosis. Overexpression of truncated protein domains further identified the Galpha-binding C-terminal portion of LGN as a sufficient domain for cortical localization in cell culture. In mitotic COS cells that normally do not exhibit cortical LGN localization, LGN is redirected to the cell cortex upon overexpression of Galpha subunits of heterotrimeric G-proteins. The results also show that the cortical localization of LGN is dependent on microfilaments and that interfering with LGN function in cultured cell lines causes early disruption to cell cycle progression.
Collapse
Affiliation(s)
- Rachna Kaushik
- Institute of Molecular and Cell Biology, Singapore 117609
| | | | | | | | | |
Collapse
|
68
|
Pattingre S, De Vries L, Bauvy C, Chantret I, Cluzeaud F, Ogier-Denis E, Vandewalle A, Codogno P. The G-protein regulator AGS3 controls an early event during macroautophagy in human intestinal HT-29 cells. J Biol Chem 2003; 278:20995-1002. [PMID: 12642577 DOI: 10.1074/jbc.m300917200] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
AGS3 contains GoLoco or G-protein regulatory motifs in its COOH-terminal half that stabilize the GDP-bound conformation of the alpha-subunit of the trimeric Gi3 protein. The latter is part of a signaling pathway that controls the lysosomal-autophagic catabolism in human colon cancer HT-29 cells. In the present work we show that the mRNA encoding for AGS3 is expressed in human intestinal cell lines (Caco-2 and HT-29) whatever their state of differentiation. Together with the full-length form, minute amounts of the mRNA encoding a NH2-terminal truncated form of AGS3, previously characterized in cardiac tissues, were also detected. Both the endogenous form of AGS3 and a tagged expressed form have a localization compatible with a role in the Galphai3-dependent control of autophagy. Accordingly, expressing its non-Galphai3-interacting NH2-terminal domain or its Galphai3-interacting COOH-terminal domain reversed the stimulatory role of AGS3 on autophagy. On the basis of biochemical and morphometric analysis, we conclude that AGS3 is involved in an early event during the autophagic pathway probably prior to the formation of the autophagosome. These data demonstrate that AGS3 is a novel partner of the Galphai3 protein in the control of a major catabolic pathway.
Collapse
Affiliation(s)
- Sophie Pattingre
- INSERM U504, 16 avenue Paul-Vaillant-Couturier, 94807 Villejuif Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
69
|
Sarma T, Voyno-Yasenetskaya T, Hope TJ, Rasenick MM. Heterotrimeric G-proteins associate with microtubules during differentiation in PC12 pheochromocytoma cells. FASEB J 2003; 17:848-59. [PMID: 12724344 DOI: 10.1096/fj.02-0730com] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Tubulin modifies G-protein signaling and heterotrimeric G-proteins regulate microtubule assembly. Here we report an interplay among G-protein-coupled receptor and receptor tyrosine kinase (such as nerve growth factor-NGF) signaling systems in PC12 pheochromocytoma cells that resulted in a translocation of Galpha(s), Galpha(i1), and Galpha(o) from cell bodies to cellular processes where they appear to localize with tubulin-containing structures. This relocation appeared to depend on the integrity of microtubules, as it was blocked and reversed by nocodazole. Latrunculin, which promotes actin filament depolymerization, had no effect. Both deconvolution microscopy and immunoprecipitation showed a significant increase of Galpha association with microtubules that was coincident with the extension of "neurites." There were distinctions among the Galpha subtypes, with Galpha(s) showing the most profound NGF-induced colocalization with tubulin. Translocation of Galpha was blocked by agents that inhibit the MAP kinases required for neuronal differentiation, suggesting that G-protein relocation is triggered by the intracellular signals for differentiation. Consistent with this, Galpha in Neuro-2A cells, which spontaneously differentiate, showed a similar translocation coincident with differentiation. Thus, diverse signals that promote neuronal differentiation and changes in cell morphology may use specific G-proteins to evoke cytoskeletal rearrangement.
Collapse
Affiliation(s)
- Tulika Sarma
- Department of Physiology, College of Medicine, Chicago, Illinois 60612-7342, USA
| | | | | | | |
Collapse
|
70
|
Abstract
This review outlines recent advances related to the molecular mechanisms and pathways of aquaporin-2 (AQP2) water channel trafficking. AQP2 is a fascinating protein, whose sorting signals can be interpreted by different cell types to achieve apical or basolateral membrane insertion, in both regulated and constitutive trafficking pathways. In addition to the well-known cAMP-mediated, stimulatory effect of vasopressin on AQP2 membrane insertion, other signaling and trafficking events can also lead to AQP2 membrane accumulation via cAMP-independent mechanisms. These include 1) elevation of cGMP, mediated by sodium nitroprusside (a nitric oxide donor), atrial natriuretic factor, and l-arginine (via nitric oxide synthase); 2) disruption of the actin cytoskeleton; and 3) inhibition of the clathrin-mediated endocytotic arm of the AQP2 recycling pathway by dominant-negative dynamin expression and by membrane cholesterol depletion. Recent data also indicate that AQP2 recycles constitutively in epithelial cells, it can be inserted into different membrane domains in different cell types both in vitro and in vivo, and these pathways can be modulated by factors including hypertonicity. The roles of accessory proteins, including small GTPases and soluble N-ethylmaleimide-sensitive factor attachment protein receptor proteins in AQP2 membrane insertion, are also being uncovered. Understanding cAMP-independent mechanisms for membrane insertion of AQP2 is especially relevant to the therapeutic bypassing of the mutated, dysfunctional vasopressin receptor in patients with X-linked nephrogenic diabetes insipidus.
Collapse
Affiliation(s)
- Dennis Brown
- Program in Membrane Biology and Renal Unit, Department of Medicine, Massachusetts General Hospital, Charlestown 02129, USA.
| |
Collapse
|
71
|
Wylie FG, Lock JG, Jamriska L, Khromykh T, Brown DL, Stow JL. GAIP participates in budding of membrane carriers at the trans-Golgi network. Traffic 2003; 4:175-89. [PMID: 12656990 DOI: 10.1034/j.1600-0854.2003.00106.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Galpha interacting protein (GAIP) is a regulator of G protein signaling protein that associates dynamically with vesicles and has been implicated in membrane trafficking, although its specific role is not yet known. Using an in vitro budding assay, we show that GAIP is recruited to a specific population of trans-Golgi network-derived vesicles and that these are distinct from coatomer or clathrin-coated vesicles. A truncation mutant (NT-GAIP) encoding only the N-terminal half of GAIP is recruited to trans-Golgi network membranes during the formation of vesicle carriers. Overexpression of NT-GAIP induces the formation of long, coated tubules, which are stabilized by microtubules. Results from the budding assay and from imaging in live cells show that these tubules remain attached to the Golgi stack rather than being released as carrier vesicles. NT-GAIP expression blocks membrane budding and results in the accumulation of tubular carrier intermediates. NT-GAIP-decorated tubules are competent to load vesicular stomatitis virus protein G-green fluorescent protein as post-Golgi, exocytic cargo and in cells expressing NT-GAIP there is reduced surface delivery of vesicular stomatitis virus protein G-green fluorescent protein. We conclude that GAIP functions as an essential part of the membrane budding machinery for a subset of post-Golgi exocytic carriers derived from the trans-Golgi network.
Collapse
Affiliation(s)
- Fiona G Wylie
- Institute for Molecular Bioscience and Centre for Functional and Applied Genomics, University of Queensland, Brisbane, Queensland, Australia
| | | | | | | | | | | |
Collapse
|
72
|
Sjölinder M, Uhlmann J, Ponstingl H. DelGEF, a homologue of the Ran guanine nucleotide exchange factor RanGEF, binds to the exocyst component Sec5 and modulates secretion. FEBS Lett 2002; 532:211-5. [PMID: 12459492 DOI: 10.1016/s0014-5793(02)03677-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
In order to identify the function of deafness locus putative guanine nucleotide exchange factor (DelGEF), a protein homologous to the nucleotide exchange factor for the small GTPase Ran, a cDNA library was screened for interacting proteins using a yeast two-hybrid system. The human homologue of Sec5, a protein involved in vesicle transport and secretion, was identified as a binding partner. The interaction between DelGEF and Sec5 was found to be dependent on Mg2+ and stimulated by guanosine triphosphate (GTP) or deoxycytidine triphosphate (dCTP). Downregulation of endogenous DelGEF in HeLa cells induced increased extracellular secretion of proteoglycans indicating a possible role for DelGEF in the secretion process.
Collapse
Affiliation(s)
- Mikael Sjölinder
- Division for Molecular Biology of Mitosis, German Cancer Research Center, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | | | | |
Collapse
|
73
|
Qian L, Yang T, Chen H, Xie J, Zeng H, Warren DW, MacVeigh M, Meneray MA, Hamm-Alvarez SF, Mircheff AK. Heterotrimeric GTP-binding proteins in the lacrimal acinar cell endomembrane system. Exp Eye Res 2002; 74:7-22. [PMID: 11878814 DOI: 10.1006/exer.2001.1108] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Secretagogues accelerate traffic in the lysosomal and basal-lateral pathways, as well as in the regulated apical secretory pathway, of lacrimal acinar cells. It has been proposed that alterations of protein segregation in compartments where these traffic pathways intersect may influence autoimmune responses. Heterotrimeric GTP-binding proteins couple secretagogue receptor ligand binding to activation of intracellular signaling cascades, but they are also suggested to participate in endomembrane traffic phenomena. Distributions of G(o), G(i3), G(q), G(11), and two G(s)isoforms were mapped in reconstituted lacrimal acini by confocal immunofluorescence microscopy and in lysates of the reconstituted acini by analytical subcellular fractionation. All G proteins examined were detected at low levels in isolated compartments (blm(i,j)) believed to represent the basal-lateral plasma membrane. G(i3), G(11), and the G(s)isoforms were concentrated in a series of isolated compartments believed to be related to domains of a basal-lateral endosome with sorting and recycling functions (ble-s/r(i,j,k)), a distinct endosomal compartment with basal-lateral membrane-like composition (e-blml), and domains of the trans-Golgi network believed to be involved in traffic to and from the basal-lateral membrane (tgn-blmr). G(o)and G(q)were concentrated in compartments believed to represent a mixture of immature and mature secretory vesicle membranes (isvm and svm) and domains of the trans-Golgi network compartment believed to mediate traffic to secretory vesicles (tgn-svr) and to pre-lysosomes (tgn-lr). Confocal fluorescence microscopy confirmed the presence of both basal-lateral membrane and intracellular pools of the G proteins. Stimulation with 10 microM carbachol for 20min caused a component of the G(o)to redistribute away from the isvm+svm; components of the G(i3), G(q), and G(s)to redistribute away from the tgn-svr+tgn-lr; and a component of the G(i3)to redistribute away from the ble-blml+tgn-blmr. Thus, these proteins may participate in endomembrane traffic steps activated by cholinergic stimulation in addition to playing their classical roles in plasma membrane signal transduction.
Collapse
Affiliation(s)
- Limin Qian
- Department of Physiology and Biophysics, University of Southern California, Los Angeles, CA 90033, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Weiss TS, Chamberlain CE, Takeda T, Lin P, Hahn KM, Farquhar MG. Galpha i3 binding to calnuc on Golgi membranes in living cells monitored by fluorescence resonance energy transfer of green fluorescent protein fusion proteins. Proc Natl Acad Sci U S A 2001; 98:14961-6. [PMID: 11752444 PMCID: PMC64966 DOI: 10.1073/pnas.261572098] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2001] [Indexed: 11/18/2022] Open
Abstract
Galphai3 is found both on the plasma membrane and on Golgi membranes. Calnuc, an EF hand protein, binds both Galphai3 and Ca(2+) and is found both in the Golgi lumen and in the cytoplasm. To investigate whether Galphai3 binds calnuc in living cells and where this interaction takes place we performed fluorescence resonance energy transfer (FRET) analysis between Galphai3 and calnuc in COS-7 cells expressing Galphai3-yellow fluorescent protein (YFP) and calnuc-cyan fluorescent protein (CFP). The tagged proteins have the same localization as the endogenous, nontagged proteins. When Galphai3-YFP and calnuc-CFP are coexpressed, a FRET signal is detected in the Golgi region, but no FRET signal is detected on the plasma membrane. FRET is also seen within the Golgi region when Galphai3 is coexpressed with cytosolic calnuc(DeltaN2-25)-CFP lacking its signal sequence. No FRET signal is detected when Galphai3(DeltaC12)-YFP lacking the calnuc-binding region is coexpressed with calnuc-CFP or when Galphai3-YFP and calnuc(DeltaEF-1,2)-CFP, which is unable to bind Galphai3, are coexpressed. Galphai3(G2AC3A)-YFP lacking its lipid anchors is localized in the cytoplasm, and no FRET signal is detected when it is coexpressed with wild-type calnuc-CFP. These results indicate that cytosolic calnuc binds to Galphai3 on Golgi membranes in living cells and that Galphai3 must be anchored to the cytosolic surface of Golgi membranes via lipid anchors for the interaction to occur. Calnuc has the properties of a Ca(2+) sensor protein capable of binding to and potentially regulating interactions of Galphai3 on Golgi membranes.
Collapse
Affiliation(s)
- T S Weiss
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | |
Collapse
|
75
|
Damiani MT, Colombo MI. Involvement of heterotrimeric G proteins in phagocytosis and recycling from the phagosomal compartment. Exp Cell Res 2001; 271:189-99. [PMID: 11697895 DOI: 10.1006/excr.2001.5354] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Phagocytosis is a receptor-mediated process by which specialized cell types engulf large extracellular particles. Phagosome maturation involves a series of intracellular membrane fusion and budding events resulting in the delivery of particles to compartments enriched in lysosomal hydrolases where they are digested. Substantial amounts of plasma membrane and many phagosomal proteins, such as receptors, rapidly recycle to the plasma membrane following phagosome formation. Despite the importance of this recycling pathway in phagosome maturation and in the retrieval of immunogenic peptides from phagosomes, the molecular machinery involved is largely unknown. To assess the participation of GTPases in phagocytosis and recycling from phagosomes we used aluminum fluoride (AIF(-)(4)), which activates the GDP-bound form of stimulatory and inhibitory trimeric G proteins. AlF(-)(4) inhibited both the uptake to and the recycling from the phagosomal compartment. Cholera toxin, which activates Galphas, and pertussis toxin, which uncouples Gi and Go from receptors, were effective inhibitors of phagocytosis. However, both toxins stimulated recycling from phagosomes. These results suggest that more than one GTP-binding protein participates either directly or indirectly not only in phagocytosis, but also in maturation and recycling from phagosomes, and thereby assign a role for heterotrimeric G proteins in controlling traffic through the phagocytic pathway.
Collapse
Affiliation(s)
- M T Damiani
- Instituto de Histología y Embriología, Universidad Nacional de Cuyo-CONICET, Mendoza, 5500, Argentina
| | | |
Collapse
|
76
|
Lampe PD, Qiu Q, Meyer RA, TenBroek EM, Walseth TF, Starich TA, Grunenwald HL, Johnson RG. Gap junction assembly: PTX-sensitive G proteins regulate the distribution of connexin43 within cells. Am J Physiol Cell Physiol 2001; 281:C1211-22. [PMID: 11546658 DOI: 10.1152/ajpcell.2001.281.4.c1211] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cells expressing connexin43 are able to upregulate gap junction (GJ) communication by enhancing the assembly of new GJs, apparently through increased connexin trafficking. Because G proteins are known to regulate different aspects of protein trafficking, we examined the effects of pertussis toxin (PTX; a specific inhibitor of certain G proteins) on GJ assembly. Dissociated Novikoff hepatoma cells were reaggregated for 60 min to form nascent junctions. PTX inhibited GJ assembly, as indicated by a reduction in dye transfer. Electron microscopy also revealed a 60% decrease in the number of GJ channels per cell interface. Importantly, PTX blocked the twofold enhancement in GJ assembly found in the presence of low-density lipoprotein. Two G(i alpha) proteins (G(i alpha 2) and G(i alpha 3)), which have been implicated in the control of membrane trafficking, reacted with PTX in ADP-ribosylation studies. PTX and/or the trafficking inhibitors, brefeldin A and monensin, inhibited GJ assembly to comparable degrees. In addition, assays for GJ hemichannels demonstrated reduced plasma membrane levels of connexin43 following PTX treatment. These results suggest that PTX-sensitive G proteins regulate connexin43 trafficking, and, as a result of inhibition with PTX, the number of plasma membrane hemichannels available for GJ assembly is reduced.
Collapse
Affiliation(s)
- P D Lampe
- Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA.
| | | | | | | | | | | | | | | |
Collapse
|
77
|
Azzouz N, Gerold P, Kedees MH, Shams-Eldin H, Werner R, Capdeville Y, Schwarz RT. Regulation of Paramecium primaurelia glycosylphosphatidyl-inositol biosynthesis via dolichol phosphate mannose synthesis. Biochimie 2001; 83:801-9. [PMID: 11530213 DOI: 10.1016/s0300-9084(01)01317-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
A set of glycosylinositol-phosphoceramides, belonging to a family of glycosylphosphatidyl-inositols (GPIs) synthesized in a cell-free system prepared from the free-living protozoan Paramecium primaurelia has been described. The final GPI precursor was identified and structurally characterized as: ethanolamine-phosphate-6Man alpha 1-2Man alpha 1-6(mannosylphosphate) Man alpha 1-4glucosamine-inositol-phospho-ceramide. During our investigations on the biosynthesis of the acid-labile modification, the additional mannosyl phosphate substitution, we observed that the use of the nucleotide triphosphate analogue GTP gamma S (guanosine 5-O-(thiotriphosphate)) blocks the biosynthesis of the mannosylated GPI glycolipids. We show that GTP gamma S inhibits the synthesis of dolichol-phosphate-mannose, which is the donor of the mannose residues for GPI biosynthesis. Therefore, we investigated the role of GTP binding regulatory 'G' proteins using cholera and pertussis toxins and an intracellular second messenger cAMP analogue, 8-bromo-cAMP. All the data obtained suggest the involvement of classical heterotrimeric G proteins in the regulation of GPI-anchor biosynthesis through dolichol-phosphate-mannose synthesis via the activation of adenylyl cyclase and protein phosphorylation. Furthermore, our data suggest that GTP gamma S interferes with synthesis of dolichol monophosphate, indicating that the dolichol kinase is regulated by the heterotrimeric G proteins.
Collapse
Affiliation(s)
- N Azzouz
- Zentrum für Hygiene und Medizinische Mikrobiologie, Philipps-Universität Marburg, Robert-Koch-Strasse 17, 35037 Marburg, Germany
| | | | | | | | | | | | | |
Collapse
|
78
|
Saha C, Nigam SK, Denker BM. Expanding role of G proteins in tight junction regulation: Galpha(s) stimulates TJ assembly. Biochem Biophys Res Commun 2001; 285:250-6. [PMID: 11444833 DOI: 10.1006/bbrc.2001.5154] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Multiple signaling mechanisms regulate epithelial cell tight junction (TJ) assembly and maintenance. Several G proteins are likely to regulate these processes, but only G(i/o) have been specifically tested. Treatment of MDCK cells with cholera toxin, a Galpha(s) activator, accelerated TJ development in the calcium switch as measured by the time to half-maximal [T(50) (H)] transepithelial resistance (TER). Galpha(s) was predominantly localized in the lateral membrane, but a fraction colocalizes with ZO-1 in the TJ. MDCK cell lines expressing epitope-tagged Galpha(s) and constitutively active (R201Calpha(s)) showed a similar localization. TJ assembly was significantly faster in R201Calpha(s)-MDCK cell lines (T(50) (H) of 1.7 versus 3.3 h for controls) without detectable differences in cAMP levels. Confocal studies showed R201Calpha(s)-MDCK cells more rapidly localized ZO-1 and occludin into the developing TJ without affecting E-cadherin or Na(+)/K(+) ATPase localization. Endogenous Galpha(s) and R201Calpha(s) were immunoprecipitated with ZO-1 at baseline and during TJ assembly. The data supports a model of multiple Galpha subunits interacting with TJ proteins to regulate the assembly and maintenance of the TJ.
Collapse
Affiliation(s)
- C Saha
- Renal Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
79
|
Miranda KC, Khromykh T, Christy P, Le TL, Gottardi CJ, Yap AS, Stow JL, Teasdale RD. A dileucine motif targets E-cadherin to the basolateral cell surface in Madin-Darby canine kidney and LLC-PK1 epithelial cells. J Biol Chem 2001; 276:22565-72. [PMID: 11312273 DOI: 10.1074/jbc.m101907200] [Citation(s) in RCA: 133] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
E-cadherin is a major adherens junction protein of epithelial cells, with a central role in cell-cell adhesion and cell polarity. Newly synthesized E-cadherin is targeted to the basolateral cell surface. We analyzed targeting information in the cytoplasmic tail of E-cadherin by utilizing chimeras of E-cadherin fused to the ectodomain of the interleukin-2alpha (IL-2alpha) receptor expressed in Madin-Darby canine kidney and LLC-PK(1) epithelial cells. Chimeras containing the full-length or membrane-proximal half of the E-cadherin cytoplasmic tail were correctly targeted to the basolateral domain. Sequence analysis of the membrane-proximal tail region revealed the presence of a highly conserved dileucine motif, which was analyzed as a putative targeting signal by mutagenesis. Elimination of this motif resulted in the loss of Tac/E-cadherin basolateral localization, pinpointing this dileucine signal as being both necessary and sufficient for basolateral targeting of E-cadherin. Truncation mutants unable to bind beta-catenin were correctly targeted, showing, contrary to current understanding, that beta-catenin is not required for basolateral trafficking. Our results also provide evidence that dileucine-mediated targeting is maintained in LLC-PK(1) cells despite the altered polarity of basolateral proteins with tyrosine-based signals in this cell line. These results provide the first direct insights into how E-cadherin is targeted to the basolateral membrane.
Collapse
Affiliation(s)
- K C Miranda
- Institute for Molecular Bioscience, the Department of Biochemistry, and the Department of Physiology & Pharmacology, University of Queensland, Brisbane, Queensland 4072, Australia
| | | | | | | | | | | | | | | |
Collapse
|
80
|
Gkantiragas I, Brügger B, Stüven E, Kaloyanova D, Li XY, Löhr K, Lottspeich F, Wieland FT, Helms JB. Sphingomyelin-enriched microdomains at the Golgi complex. Mol Biol Cell 2001; 12:1819-33. [PMID: 11408588 PMCID: PMC37344 DOI: 10.1091/mbc.12.6.1819] [Citation(s) in RCA: 126] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Sphingomyelin- and cholesterol-enriched microdomains can be isolated as detergent-resistant membranes from total cell extracts (total-DRM). It is generally believed that this total-DRM represents microdomains of the plasma membrane. Here we describe the purification and detailed characterization of microdomains from Golgi membranes. These Golgi-derived detergent-insoluble complexes (GICs) have a low buoyant density and are highly enriched in lipids, containing 25% of total Golgi phospholipids including 67% of Golgi-derived sphingomyelin, and 43% of Golgi-derived cholesterol. In contrast to total-DRM, GICs contain only 10 major proteins, present in nearly stoichiometric amounts, including the alpha- and beta-subunits of heterotrimeric G proteins, flotillin-1, caveolin, and subunits of the vacuolar ATPase. Morphological data show a brefeldin A-sensitive and temperature-sensitive localization to the Golgi complex. Strikingly, the stability of GICs does not depend on its membrane environment, because, after addition of brefeldin A to cells, GICs can be isolated from a fused Golgi-endoplasmic reticulum organelle. This indicates that GIC microdomains are not in a dynamic equilibrium with neighboring membrane proteins and lipids. After disruption of the microdomains by cholesterol extraction with cyclodextrin, a subcomplex of several GIC proteins including the B-subunit of the vacuolar ATPase, flotillin-1, caveolin, and p17 could still be isolated by immunoprecipitation. This indicates that several of the identified GIC proteins localize to the same microdomains and that the microdomain scaffold is not required for protein interactions between these GIC proteins but instead might modulate their affinity.
Collapse
Affiliation(s)
- I Gkantiragas
- Biochemie-Zentrum Heidelberg (BZH), University of Heidelberg, Im Neuenheimer Feld 328, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Roseberry AG, Bünemann M, Elavunkal J, Hosey MM. Agonist-dependent delivery of M(2) muscarinic acetylcholine receptors to the cell surface after pertussis toxin treatment. Mol Pharmacol 2001; 59:1256-68. [PMID: 11306711 DOI: 10.1124/mol.59.5.1256] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The internalization of the M(2) muscarinic cholinergic receptor (mAChR) proceeds through an atypical pathway that is independent of arrestin and clathrin function and shows a unique sensitivity to dynamin when the receptor is expressed in human embryonic kidney 293 cells. In this report we demonstrate that the internalization of the M(2) mAChR was modulated by activation of heterotrimeric G proteins, because treatment with pertussis toxin, which ADP-ribosylates G proteins of the G(i/o) family, caused a significant delay in the onset of internalization of the M(2) mAChR. The effects of pertussis toxin could not be explained by alteration of the agonist-dependent phosphorylation of the M(2) mAChR. The modulation of internalization by pertussis toxin was revealed to be due to recruitment of intracellular receptors to the cell surface upon agonist treatment. Pretreatment with pertussis toxin also greatly increased both the rate and extent of recovery of M(2) mAChRs to the cell surface after agonist-mediated internalization. These results demonstrate a novel aspect involved in the regulation of GPCRs. As with the tightly controlled internalization of GPCRs, the delivery of GPCRs to the cell surface is also highly regulated.
Collapse
Affiliation(s)
- A G Roseberry
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Medical School, Chicago, Illinois, USA
| | | | | | | |
Collapse
|
82
|
Wilson ML, Guild SB. Effects of wortmannin upon the late stages of the secretory pathway of AtT-20 cells. Eur J Pharmacol 2001; 413:55-62. [PMID: 11173063 DOI: 10.1016/s0014-2999(01)00741-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Heterotrimeric GTP-binding (G) proteins, termed Ge, have a role in the late stages of the adrenocorticotrophin (ACTH) secretory pathway in the mouse AtT-20/D16-16 anterior pituitary tumour cell line. The wortmannin sensitivity of Ge-controlled mechanisms in AtT-20 cells was investigated to provide information on the possible mechanisms linking Ge with secretion. Permeabilised cells exposed to calcium ions (10(-9) to 10(-3) M), guanosine 5'-O-(3-thiotriphosphate) (GTP-gamma-S) (10(-8) to 10(-4) M) and mastoparan (10(-8) to 10(-5) M) demonstrated a significant and concentration-dependent stimulation of ACTH secretion from non-stimulated levels for all three agents. Coincubation with wortmannin (10(-5) M) significantly inhibited both calcium-independent and -stimulated secretion. The effect of wortmannin was concentration-dependent being maximal at 10(-6) M. The study shows that wortmannin inhibits both calcium-independent and -stimulated secretion from permeabilised AtT-20 cells indicating a role for phosphatidylinositol-3 kinase in determining the size of the readily releasable pool of ACTH and/or in mediating calcium/Ge-evoked secretion from this pool.
Collapse
Affiliation(s)
- M L Wilson
- Molecular Endocrinology Group, School of Biological and Medical Science, University of St. Andrews, Bute Medical Building, St. Andrews KY16 9TS, UK
| | | |
Collapse
|
83
|
Morel V, Poschet R, Traverso V, Deretic D. Towards the proteome of the rhodopsin-bearing post-Golgi compartment of retinal photoreceptor cells. Electrophoresis 2000; 21:3460-9. [PMID: 11079565 DOI: 10.1002/1522-2683(20001001)21:16<3460::aid-elps3460>3.0.co;2-i] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Polarized sorting of rhodopsin in retinal rod photoreceptor cells is mediated by post-Golgi carrier membranes that bud from the trans-Golgi network and fuse with the specialized domain of the plasma membrane in the rod inner segment. The identity of the majority of the resident proteins of this organelle still remains elusive, despite multifaceted approaches to study this compartment. In the present study we have taken a proteomic approach to the analysis of the post-Golgi carriers. First, we modified the previously established fractionation protocols in order to achieve greater purity of the isolated membranes. Specifically, the new fractionation scheme depleted the post-Golgi fraction of cytosolic proteins that were the most abundant contaminants complicating analysis of two-dimensional (2-D) gel profiles in our previous preparations. The isolated membranes were subjected to 2-D gel electrophoresis, immunoblotting and microsequencing. This analysis showed that the improved subcellular fractionation yielded a fraction highly enriched in rhodopsin-bearing post-Golgi carrier membranes. Two-dimensional mapping revealed 29 proteins that are preferentially found in this fraction and therefore represent candidates for post-Golgi membrane-specific proteins. This preparation of rhodopsin-bearing post-Golgi carriers is a first step towards the proteomics of this important organelle.
Collapse
Affiliation(s)
- V Morel
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor 48105, USA
| | | | | | | |
Collapse
|
84
|
Taylor RS, Wu CC, Hays LG, Eng JK, Yates JR, Howell KE. Proteomics of rat liver Golgi complex: minor proteins are identified through sequential fractionation. Electrophoresis 2000; 21:3441-59. [PMID: 11079564 DOI: 10.1002/1522-2683(20001001)21:16<3441::aid-elps3441>3.0.co;2-g] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The discovery of novel proteins resident to the Golgi complex will fuel our future studies of Golgi structure/function and provide justification for proteomic analysis of this organelle. Our approach to Golgi proteomics was to first isolate and characterize the intact organelle free of proteins in transit by use of tissue pretreated with cycloheximide. Then the stacked Golgi fraction was fractionated into biochemically defined subfractions: Triton X-114 insoluble, aqueous, and detergent phases. The aqueous and detergent phases were further fractionated by anion-exchange column chromatography. In addition, radiolabeled cytosol was incubated with stacked Golgi fractions containing proteins in transit, and the proteins bound to the Golgi stacks in an energy-dependent manner were characterized. All fractions were analyzed by two-dimensional (2-D) gel electrophoresis and identification numbers were given to 588 unique 2-D spots. Tandem mass spectrometry was used to analyze 93 of the most abundant 2-D spots taken from preparative Triton X-114 insoluble, aqueous and detergent phase 2-D gels. Fifty-one known and 22 unknown proteins were identified. This study represents the first installment in the mammalian Golgi proteome database. Our data suggest that cell fractionation followed by biochemical dissection of specific classes of molecules provides a significant advantage for the identification of low abundance proteins in organelles.
Collapse
Affiliation(s)
- R S Taylor
- Department of Cellular and Structural Biology, University of Colorado, School of Medicine, Denver 80262, USA
| | | | | | | | | | | |
Collapse
|
85
|
Sullivan BM, Harrison-Lavoie KJ, Marshansky V, Lin HY, Kehrl JH, Ausiello DA, Brown D, Druey KM. RGS4 and RGS2 bind coatomer and inhibit COPI association with Golgi membranes and intracellular transport. Mol Biol Cell 2000; 11:3155-68. [PMID: 10982407 PMCID: PMC14982 DOI: 10.1091/mbc.11.9.3155] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
COPI, a protein complex consisting of coatomer and the small GTPase ARF1, is an integral component of some intracellular transport carriers. The association of COPI with secretory membranes has been implicated in the maintenance of Golgi integrity and the normal functioning of intracellular transport in eukaryotes. The regulator of G protein signaling, RGS4, interacted with the COPI subunit beta'-COP in a yeast two-hybrid screen. Both recombinant RGS4 and RGS2 bound purified recombinant beta'-COP in vitro. Endogenous cytosolic RGS4 from NG108 cells and RGS2 from HEK293T cells cofractionated with the COPI complex by gel filtration. Binding of beta'-COP to RGS4 occurred through two dilysine motifs in RGS4, similar to those contained in some aminoglycoside antibiotics that are known to bind coatomer. RGS4 inhibited COPI binding to Golgi membranes independently of its GTPase-accelerating activity on G(ialpha). In RGS4-transfected LLC-PK1 cells, the amount of COPI in the Golgi region was considerably reduced compared with that in wild-type cells, but there was no detectable difference in the amount of either Golgi-associated ARF1 or the integral Golgi membrane protein giantin, indicating that Golgi integrity was preserved. In addition, RGS4 expression inhibited trafficking of aquaporin 1 to the plasma membrane in LLC-PK1 cells and impaired secretion of placental alkaline phosphatase from HEK293T cells. The inhibitory effect of RGS4 in these assays was independent of GTPase-accelerating activity but correlated with its ability to bind COPI. Thus, these data support the hypothesis that these RGS proteins sequester coatomer in the cytoplasm and inhibit its recruitment onto Golgi membranes, which may in turn modulate Golgi-plasma membrane or intra-Golgi transport.
Collapse
Affiliation(s)
- B M Sullivan
- Renal Unit, Program in Membrane Biology, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
| | | | | | | | | | | | | | | |
Collapse
|
86
|
De Vries L, Zheng B, Fischer T, Elenko E, Farquhar MG. The regulator of G protein signaling family. Annu Rev Pharmacol Toxicol 2000; 40:235-71. [PMID: 10836135 DOI: 10.1146/annurev.pharmtox.40.1.235] [Citation(s) in RCA: 447] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Regulator of G protein signaling (RGS) proteins are responsible for the rapid turnoff of G protein-coupled receptor signaling pathways. The major mechanism whereby RGS proteins negatively regulate G proteins is via the GTPase activating protein activity of their RGS domain. Structural and mutational analyses have characterized the RGS/G alpha interaction in detail, explaining the molecular mechanisms of the GTPase activating protein activity of RGS proteins. More than 20 RGS proteins have been isolated, and there are indications that specific RGS proteins regulate specific G protein-coupled receptor pathways. This specificity is probably created by a combination of cell type-specific expression, tissue distribution, intracellular localization, posttranslational modifications, and domains other than the RGS domain that link them to other signaling pathways. In this review we discuss what has been learned so far about the role of RGS proteins in regulating G protein-coupled receptor signaling and point out areas that may be fruitful for future research.
Collapse
Affiliation(s)
- L De Vries
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla 92093, USA. ,
| | | | | | | | | |
Collapse
|
87
|
Klussmann E, Maric K, Rosenthal W. The mechanisms of aquaporin control in the renal collecting duct. Rev Physiol Biochem Pharmacol 2000; 141:33-95. [PMID: 10916423 DOI: 10.1007/bfb0119577] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The antidiuretic hormone arginine-vasopressin (AVP) regulates water reabsorption in renal collecting duct principal cells. Central to its antidiuretic action in mammals is the exocytotic insertion of the water channel aquaporin-2 (AQP2) from intracellular vesicles into the apical membrane of principal cells, an event initiated by an increase in cAMP and activation of protein kinase A. Water is then reabsorbed from the hypotonic urine of the collecting duct. The water channels aquaporin-3 (AQP3) and aquaporin-4 (AQP4), which are constitutively present in the basolateral membrane, allow the exit of water from the cell into the hypertonic interstitium. Withdrawal of the hormone leads to endocytotic retrieval of AQP2 from the cell membrane. The hormone-induced rapid redistribution between the interior of the cell and the cell membrane establishes the basis for the short term regulation of water permeability. In addition water channels (AQP2 and 3) of principal cells are regulated at the level of expression (long term regulation). This review summarizes the current knowledge on the molecular mechanisms underlying the short and long term regulation of water channels in principal cells. In the first part special emphasis is placed on the proteins involved in short term regulation of AQP2 (SNARE proteins, Rab proteins, cytoskeletal proteins, G proteins, protein kinase A anchoring proteins and endocytotic proteins). In the second part, physiological and pathophysiological stimuli determining the long term regulation are discussed.
Collapse
Affiliation(s)
- E Klussmann
- Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | | | | |
Collapse
|
88
|
Willard FS, Crouch MF. Nuclear and cytoskeletal translocation and localization of heterotrimeric G-proteins. Immunol Cell Biol 2000; 78:387-94. [PMID: 10947863 DOI: 10.1046/j.1440-1711.2000.00927.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Heterotrimeric GTP-binding proteins (G-proteins) are involved in a diverse array of signalling pathways. They are generally thought to be membrane-bound proteins, which disassociate on receptor activation and binding of GTP. A model to explain this has been proposed, which is often described as 'the G-protein cycle'. The 'G-protein cycle' is discussed in the present paper in relation to evidence that now exists regarding the non- membranous localization of G-proteins. Specifically, the experimental evidence demonstrating association of G-proteins with the cytoskeleton and the nucleus, and the mechanisms by which G-proteins translocate to these sites are reviewed. Furthermore, the possible effector pathways and the physiological function of G-proteins at these sites are discussed.
Collapse
Affiliation(s)
- F S Willard
- Molecular Signalling Group, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | | |
Collapse
|
89
|
Melien O, Sandnes D, Johansen EJ, Christoffersen T. Effects of pertussis toxin on extracellular signal-regulated kinase activation in hepatocytes by hormones and receptor-independent agents: evidence suggesting a stimulatory role of G(i) proteins at a level distal to receptor coupling. J Cell Physiol 2000; 184:27-36. [PMID: 10825231 DOI: 10.1002/(sici)1097-4652(200007)184:1<27::aid-jcp3>3.0.co;2-q] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
It was previously found that pertussis toxin (PTX) pretreatment inhibits the activation of extracellular signal-regulated kinases ERK1 (p44(mapk)) and ERK2 (p42(mapk)) in hepatocytes in response to either agonists that bind to heptahelical receptors or epidermal growth factor (EGF), suggesting a role of G(i) proteins in stimulatory mechanisms for ERK1/2. The present work shows that ERK1/2 is activated in a PTX-sensitive way not only by vasopressin, angiotensin II, prostaglandin (PG) F(2alpha), alpha(1)-adrenergic stimulation, and EGF but also by agents whose actions bypass receptors and stimulate protein kinase C (PKC) and/or elevate intracellular Ca(2+), such as 12-O-tetradecanoyl phorbol-13-acetate (TPA), exogenous phosphatidylcholine-specific phospholipase C (PC-PLC, from Bacillus cereus), thapsigargin, and the Ca(2+) ionophore A23187. Under the same conditions, PTX did not affect agonist stimulation of phosphoinositide-specific phospholipase C (PI-PLC) (IP(3) generation), and did not reduce the activation by these agents of phospholipase D (PLD). The results suggest that in hepatocytes a PTX-sensitive mechanism, presumably involving G(i) proteins, exerts a stimulatory effect on ERK at a level distal to receptor coupling, acting either as an integral part of the signaling pathway(s) or by a permissive, synergistic regulation.
Collapse
Affiliation(s)
- O Melien
- Department of Pharmacology, Faculty of Medicine, University of Oslo, Blindern, Oslo, Norway.
| | | | | | | |
Collapse
|
90
|
Can A, Sayar K, Friedman E, Ambrosio C, Erdemli E, Gurdal H. The effects of agonist stimulation and beta(2)-adrenergic receptor level on cellular distribution of gs(alpha) protein. Cell Signal 2000; 12:303-9. [PMID: 10822171 DOI: 10.1016/s0898-6568(00)00071-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
This study examines the effects of adrenergic ligands, cholera toxin, forskolin, and varying levels of beta(2) adrenergic receptors (beta(2)AR) on the cellular distribution of Gs(alpha) subunits in CHO cells. Localization of Gs(alpha) was evaluated by confocal microscopy and beta(2)AR-mediated signalling was assessed by adenylyl cyclase (AC) activity. In cells expressing 0.2 pmol/mg protein beta(2)ARs (WT18), the localization of Gs(alpha) subunit was restricted to the plasma membrane region. Isoproterenol (ISO), cholera toxin or forskolin elicited redistribution of cellular Gs(alpha) so that Gs(alpha) appeared as intense spots throughout the plasma membrane as well as the cytoplasm. Exposure to a neutral beta(2)AR antagonist, alprenolol, prevented the ISO-stimulated Gs(alpha) translocation from peripheral to inner cytoplasm. In cells expressing high level of beta(2)ARs (8.2 pmol/mg) (WT4), basal and ISO-stimulated AC activities were significantly elevated when compared to the values detected in WT18 clone, suggesting a positive correlation between receptor expression and receptor-mediated signalling. Basal Gs(alpha) distribution in this group was similar to that observed in ISO-, cholera toxin-, or forskolin-stimulated WT18 clone. ISO, cholera toxin, or forskolin did not change the distribution of Gs(alpha) significantly when tested in WT4 clone. No difference in the cellular level of Gs(alpha) protein between WT18 and WT4 clones was detected. Alprenolol did not affect the distribution of Gs(alpha) in WT4 clone. ICI 118,551, a negative beta(2)AR antagonist, altered Gs(alpha) distribution from a dispersed basal pattern to a membrane-confined pattern. The latter appearance was similar to that observed in unstimulated WT18 clone. Taken together, these data suggest that: (1) enhanced beta(2)AR-Gs(alpha) coupling induced by agonist stimulation or by increased expression of beta(2)ARs remodel the cellular distribution of Gs(alpha); (2) the alteration in Gs(alpha) distribution induced by beta(2)AR overexpression provides evidence for agonist-independent interaction of beta(2)AR and Gs(alpha), that can be inhibited by a negative antagonist but not by a neutral antagonist; and (3) forskolin influences the activity state of Gs(alpha) that displays a Gs(alpha) distribution pattern comparable to that observed when Gs(alpha) is activated via beta(2)AR stimulation or directly by cholera toxin.
Collapse
Affiliation(s)
- A Can
- Ankara University Faculty of Medicine, Department of Histology-Embryology, Sihhiye 06100, Ankara, Turkey
| | | | | | | | | | | |
Collapse
|
91
|
Happe S, Cairns M, Roth R, Heuser J, Weidman P. Coatomer vesicles are not required for inhibition of Golgi transport by G-protein activators. Traffic 2000; 1:342-53. [PMID: 11208119 DOI: 10.1034/j.1600-0854.2000.010407.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The G-protein activators guanosine 5'-O-(3-thiodiphosphate) (GTP gamma S) and aluminum fluoride (AIF) are thought to inhibit transport between Golgi cisternae by causing the accumulation of nonfunctional coatomer-coated transport vesicles on the Golgi. Although GTP gamma S and AIF inhibit transport in cell-free intra-Golgi transport systems, blocking coatomer vesicle formation does not. We therefore determined whether inhibition of in vitro Golgi transport by these agents requires coatomer vesicle formation. Depletion of coatomer was found to completely block coated vesicle formation on Golgi cisternae without affecting inhibition of in vitro transport by either GTP gamma S or AIF. Depletion of ADP-ribosylation factor (ARF) prevented inhibition of transport by GTP gamma S, but not by AIF, suggesting that the AIF-sensitive component in transport may not be a GTP-binding protein. Surprisingly, depletion of cytosolic ARF did not prevent the GTP gamma S-induced formation of Golgi-coated vesicles, whereas ARF was required for AIF-induced vesicle formation. Although ARF or coatomer depletion caused an increase in the fenestration of cisternae, no other ultrastructural changes were observed that might explain the inhibition of transport by GTP gamma S or AIF. These findings suggest that ARF-GTP gamma S and AIF act by distinct and coatomer-independent mechanisms to inhibit membrane fusion in cell-free intra-Golgi transport.
Collapse
Affiliation(s)
- S Happe
- Department of Molecular Biology and Biochemistry, St. Louis University Medical School, St Louis, MO 63104, USA
| | | | | | | | | |
Collapse
|
92
|
Yamaguchi T, Nagahama M, Itoh H, Hatsuzawa K, Tani K, Tagaya M. Regulation of the golgi structure by the alpha subunits of heterotrimeric G proteins. FEBS Lett 2000; 470:25-8. [PMID: 10722839 DOI: 10.1016/s0014-5793(00)01284-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Disassembly of the Golgi apparatus is elicited by the action of nordihydroguaiaretic acid (NDGA) and this disassembly is prevented by the activation of heterotrimeric G proteins. In the present study we showed that overexpression of Galpha(z) or Galpha(i2) significantly suppresses the disassembly of the Golgi apparatus induced by NDGA. Overexpression of Gbeta(1)gamma(2), on the other hand, had no effect on NDGA-induced Golgi disassembly. Galpha(z) neither blocked Golgi disassembly induced by brefeldin A or nocodazole, nor interfered with protein transport, suggesting its specificity on the action of NDGA. Our results suggest that the alpha subunits of heterotrimeric G proteins are responsible for the maintenance of the Golgi structure.
Collapse
Affiliation(s)
- T Yamaguchi
- School of Life Science, Tokyo University of Pharmacy and Life Science, Horinouchi 1432-1, Hachioji, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
93
|
Lin P, Fischer T, Weiss T, Farquhar MG. Calnuc, an EF-hand Ca(2+) binding protein, specifically interacts with the C-terminal alpha5-helix of G(alpha)i3. Proc Natl Acad Sci U S A 2000; 97:674-9. [PMID: 10639138 PMCID: PMC15389 DOI: 10.1073/pnas.97.2.674] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Calnuc (nucleobindin) was previously shown to be present both in the cytosol and in the Golgi and to be the major Golgi Ca(2+) binding protein. In this study we verified the existence of the cytosolic pool of calnuc and investigated its interaction with G(alpha)i3. Cytosolic calnuc was released by mild digitonin permeabilization. In pulse-chase experiments, the two pools of calnuc had different mobilities, suggesting different posttranslational modifications. That calnuc interacts with G(alpha)i3 in vivo was verified by the finding that G(alpha)i3 could be crosslinked intracellularly to calnuc and co-immunoprecipitated from NIH 3T3 cells stably overexpressing either activated (Q204L) or inactivated (G203A) G(alpha)i3. Binding was Ca(2+) and Mg(2+)-dependent. Calnuc and G(alpha)i3-GFP codistributed primarily in the Golgi region. By yeast two-hybrid analysis, the binding site on G(alpha)i3 for calnuc was mapped to the C-terminal region because removal of the last 12 amino acids (but not 11) abolished the interaction. Peptide competition indicated that calnuc, with its coiled-coil domain constituted by the two EF-hands, binds to G(alpha)i3's C-terminal alpha5-helix. These results demonstrate that calnuc may play an important role in G protein- and Ca(2+)-regulated signal transduction events.
Collapse
Affiliation(s)
- P Lin
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093-0651, USA
| | | | | | | |
Collapse
|
94
|
Faergeman NJ, Ballegaard T, Knudsen J, Black PN, DiRusso C. Possible roles of long-chain fatty Acyl-CoA esters in the fusion of biomembranes. Subcell Biochem 2000; 34:175-231. [PMID: 10808334 DOI: 10.1007/0-306-46824-7_5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Affiliation(s)
- N J Faergeman
- Department of Biochemistry and Molecular Biology, Albany Medical College, New York 12208, USA
| | | | | | | | | |
Collapse
|
95
|
Scales SJ, Gomez M, Kreis TE. Coat proteins regulating membrane traffic. INTERNATIONAL REVIEW OF CYTOLOGY 1999; 195:67-144. [PMID: 10603575 DOI: 10.1016/s0074-7696(08)62704-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This review focuses on the roles of coat proteins in regulating the membrane traffic of eukaryotic cells. Coat proteins are recruited to the donor organelle membrane from a cytosolic pool by specific small GTP-binding proteins and are required for the budding of coated vesicles. This review first describes the four types of coat complexes that have been characterized so far: clathrin and its adaptors, the adaptor-related AP-3 complex, COPI, and COPII. It then discusses the ascribed functions of coat proteins in vesicular transport, including the physical deformation of the membrane into a bud, the selection of cargo, and the targeting of the budded vesicle. It also mentions how the coat proteins may function in an alternative model for transport, namely via tubular connections, and how traffic is regulated. Finally, this review outlines the evidence that related coat proteins may regulate other steps of membrane traffic.
Collapse
Affiliation(s)
- S J Scales
- Department of Cell Biology, University of Geneva, Switzerland
| | | | | |
Collapse
|
96
|
Martín ME, Hidalgo J, Vega FM, Velasco A. Trimeric G proteins modulate the dynamic interaction of PKAII with the Golgi complex. J Cell Sci 1999; 112 ( Pt 22):3869-78. [PMID: 10547348 DOI: 10.1242/jcs.112.22.3869] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Golgi complex represents a major subcellular location of protein kinase A (PKA) concentration in mammalian cells where it has been previously shown to be involved in vesicle-mediated protein transport processes. We have studied the factors that influence the interaction of PKA typeII subunits with the Golgi complex. In addition to the cytosol, both the catalytic (Calpha) and regulatory (RIIalpha) subunits of PKAII were detected at both sides of the Golgi stack, particularly in elements of the cis- and trans-Golgi networks. PKAII subunits, in contrast, were practically absent from the middle Golgi cisternae. Cell treatment with either brefeldin A, AlF(4-) or at low temperature induced PKAII dissociation from the Golgi complex and redistribution to the cytosol. This suggested the existence of a cycle of association/dissociation of PKAII holoenzyme to the Golgi. The interaction of purified RIIalpha with Golgi membranes was studied in vitro and found not to be affected by brefeldin A while it was sensitive to modulators of heterotrimeric G proteins such as AlF(4-), GTPgammaS, beta(gamma) subunits and mastoparan. RII(alphaa) binding was stimulated by recombinant, myristoylated Galpha(i3) subunit and inhibited by cAMP. Pretreatment of Golgi membranes with bacterial toxins known to catalyze ADP-ribosylation of selected Galpha subunits also modified RIIalpha binding. Taken together the data support a regulatory role for Golgi-associated Galpha proteins in PKAII recruitment from the cytosol.
Collapse
Affiliation(s)
- M E Martín
- Department of Cell Biology, Faculty of Biology, University of Seville, Spain
| | | | | | | |
Collapse
|
97
|
Kolset SO, Vuong TT, Prydz K. Apical secretion of chondroitin sulphate in polarized Madin-Darby canine kidney (MDCK) cells. J Cell Sci 1999; 112 ( Pt 11):1797-801. [PMID: 10318771 DOI: 10.1242/jcs.112.11.1797] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Sugar moieties have been shown to contain sufficient and necessary information to target examples of secreted and transmembrane glycoproteins to the apical surface of epithelial MDCK cells. We have investigated if the sugar chains of proteoglycans, the glycosaminoglycans, also contain structural determinants for apical transport. Here we show that although 75% of the proteoglycan secretion from MDCK cell monolayers is into the basolateral medium, 75% of the proteoglycans of the chondroitin sulphate type are secreted apically. The sorting information in the chondroitin sulphate proteoglycans is localized to the sugar chains, since protein-free chondroitin sulphate chains, initiated on hexyl beta-D-thioxyloside, were also predominantly secreted to the apical medium.
Collapse
Affiliation(s)
- S O Kolset
- Institute for Nutrition Research, University of Oslo, Box 1046 Blindern, Norway.
| | | | | |
Collapse
|
98
|
Luo Y, Denker BM. Interaction of heterotrimeric G protein Galphao with Purkinje cell protein-2. Evidence for a novel nucleotide exchange factor. J Biol Chem 1999; 274:10685-8. [PMID: 10196137 DOI: 10.1074/jbc.274.16.10685] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The heterotrimeric G protein Galphao is ubiquitously expressed throughout the central nervous system, but many of its functions remain to be defined. To search for novel proteins that interact with Galphao, a mouse brain library was screened using the yeast two-hybrid interaction system. Pcp2 (Purkinje cell protein-2) was identified as a partner for Galphao in this system. Pcp2 is expressed in cerebellar Purkinje cells and retinal bipolar neurons, two locations where Galphao is also expressed. Pcp2 was first identified as a candidate gene to explain Purkinje cell degeneration in pcd mice (Nordquist, D. T., Kozak, C. A., and Orr, H. T. (1988) J. Neurosci. 8, 4780-4789), but its function remains unknown as Pcp2 knockout mice are normal (Mohn, A. R., Feddersen, R. M., Nguyen, M. S., and Koller, B. H. (1997) Mol. Cell. Neurosci. 9, 63-76). Galphao and Pcp2 binding was confirmed in vitro using glutathione S-transferase-Pcp2 fusion proteins and in vitro translated [35S]methionine-labeled Galphao. In addition, when Galphao and Pcp2 were cotransfected into COS cells, Galphao was detected in immunoprecipitates of Pcp2. To determine whether Pcp2 could modulate Galphao function, kinetic constants kcat and koff of bovine brain Galphao were determined in the presence and absence of Pcp2. Pcp2 stimulates GDP release from Galphao more than 5-fold without affecting kcat. These findings define a novel nucleotide exchange function for Pcp2 and suggest that the interaction between Pcp2 and Galphao is important to Purkinje cell function.
Collapse
Affiliation(s)
- Y Luo
- Renal Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
99
|
Jing J, Chikvashvili D, Singer-Lahat D, Thornhill WB, Reuveny E, Lotan I. Fast inactivation of a brain K+ channel composed of Kv1.1 and Kvbeta1.1 subunits modulated by G protein beta gamma subunits. EMBO J 1999; 18:1245-56. [PMID: 10064591 PMCID: PMC1171215 DOI: 10.1093/emboj/18.5.1245] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Modulation of A-type voltage-gated K+ channels can produce plastic changes in neuronal signaling. It was shown that the delayed-rectifier Kv1.1 channel can be converted to A-type upon association with Kvbeta1.1 subunits; the conversion is only partial and is modulated by phosphorylation and microfilaments. Here we show that, in Xenopus oocytes, expression of Gbeta1gamma2 subunits concomitantly with the channel (composed of Kv1.1 and Kvbeta1.1 subunits), but not after the channel's expression in the plasma membrane, increases the extent of conversion to A-type. Conversely, scavenging endogenous Gbetagamma by co-expression of the C-terminal fragment of the beta-adrenergic receptor kinase reduces the extent of conversion to A-type. The effect of Gbetagamma co-expression is occluded by treatment with dihydrocytochalasin B, a microfilament-disrupting agent shown previously by us to enhance the extent of conversion to A-type, and by overexpression of Kvbeta1.1. Gbeta1gamma2 subunits interact directly with GST fusion fragments of Kv1.1 and Kvbeta1.1. Co-expression of Gbeta1gamma2 causes co-immunoprecipitation with Kv1.1 of more Kvbeta1.1 subunits. Thus, we suggest that Gbeta1gamma2 directly affects the interaction between Kv1.1 and Kvbeta1.1 during channel assembly which, in turn, disrupts the ability of the channel to interact with microfilaments, resulting in an increased extent of A-type conversion.
Collapse
Affiliation(s)
- J Jing
- Department of Physiology and Pharmacology, Sackler School of Medicine, Tel-Aviv University, 69978 Ramat Avivl, Israel
| | | | | | | | | | | |
Collapse
|
100
|
Babiá T, Ayala I, Valderrama F, Mato E, Bosch M, Santarén JF, Renau-Piqueras J, Kok JW, Thomson TM, Egea G. N-Ras induces alterations in Golgi complex architecture and in constitutive protein transport. J Cell Sci 1999; 112 ( Pt 4):477-89. [PMID: 9914160 DOI: 10.1242/jcs.112.4.477] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aberrant glycosylation of proteins and lipids is a common feature of many tumor cell types, and is often accompanied by alterations in membrane traffic and an anomalous localization of Golgi-resident proteins and glycans. These observations suggest that the Golgi complex is a key organelle for at least some of the functional changes associated with malignant transformation. To gain insight into this possibility, we have analyzed changes in the structure and function of the Golgi complex induced by the conditional expression of the transforming N-Ras(K61) mutant in the NRK cell line. A remarkable and specific effect associated with this N-Ras-induced transformation was a conspicuous rearrangement of the Golgi complex into a collapsed morphology. Ultrastructural and stereological analyses demonstrated that the Golgi complex was extensively fragmented. The collapse of the Golgi complex was also accompanied by a disruption of the actin cytoskeleton. Functionally, N-Ras-transformed KT8 cells showed an increase in the constitutive protein transport from the trans-Golgi network to the cell surface, and did not induce the appearance of aberrant cell surface glycans. The Golgi complex collapse, the actin disassembly, and the increased constitutive secretion were all partially inhibited by the phospholipase A2 inhibitor 4-bromophenylacyl bromide. The results thus suggest the involvement of the actin cytoskeleton in the shape of the Golgi complex, and intracellular phospholipase A2 in its architecture and secretory function.
Collapse
Affiliation(s)
- T Babiá
- Departament de Biologia Cel.lular, Facultat de Medicina, IDIBAPS, Universitat de Barcelona, C/Casanova, 08036 Barcelona (Spain).
| | | | | | | | | | | | | | | | | | | |
Collapse
|