101
|
Abstract
ADP-ribosylation factors (ARFs) are critical regulators of vesicular trafficking pathways and act at multiple intracellular sites. ADP-ribosylation factor-GTPase-activating proteins (ARFGAPs) are proposed to contribute to site-specific regulation. In yeast, two distinct proteins, Glo3p and Gcs1p, together provide overlapping, essential ARFGAP function required for coat protein (COP)-I-dependent trafficking. In mammalian cells, only the Gcs1p orthologue, named ARFGAP1, has been characterized in detail. However, Glo3p is known to make the stronger contribution to COP I traffic in yeast. Here, based on a conserved signature motif close to the carboxy terminus, we identify ARFGAP2 and ARFGAP3 as the human orthologues of yeast Glo3p. By immunofluorescence (IF), ARFGAP2 and ARFGAP3 are closely colocalized with coatomer subunits in NRK cells in the Golgi complex and peripheral punctate structures. In contrast to ARFGAP1, both ARFGAP2 and ARFGAP3 are associated with COP-I-coated vesicles generated from Golgi membranes in the presence of GTP-γ-S in vitro. ARFGAP2 lacking its zinc finger domain directly binds to coatomer. Expression of this truncated mutant (ΔN-ARFGAP2) inhibits COP-I-dependent Golgi-to-endoplasmic reticulum transport of cholera toxin (CTX-K63) in vivo. Silencing of ARFGAP1 or a combination of ARFGAP2 and ARFGAP3 in HeLa cells does not decrease cell viability. However, silencing all three ARFGAPs causes cell death. Our data provide strong evidence that ARFGAP2 and ARFGAP3 function in COP I traffic.
Collapse
Affiliation(s)
- Gabriella Frigerio
- Department of Clinical Biochemistry, Cambridge Institute for Medical Research, University of CambridgeHills Road, Cambridge CB2 2XY, United Kingdom
- European Bioinformatics Institute, Wellcome Trust Genome CampusHinxton, Cambridge CB10 1SD, United Kingdom
| | - Neil Grimsey
- Department of Clinical Biochemistry, Cambridge Institute for Medical Research, University of CambridgeHills Road, Cambridge CB2 2XY, United Kingdom
| | - Martin Dale
- Department of Clinical Biochemistry, Cambridge Institute for Medical Research, University of CambridgeHills Road, Cambridge CB2 2XY, United Kingdom
| | - Irina Majoul
- Department of Clinical Biochemistry, Cambridge Institute for Medical Research, University of CambridgeHills Road, Cambridge CB2 2XY, United Kingdom
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of LondonEgham TW20 0EX, United Kingdom
| | - Rainer Duden
- Department of Clinical Biochemistry, Cambridge Institute for Medical Research, University of CambridgeHills Road, Cambridge CB2 2XY, United Kingdom
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of LondonEgham TW20 0EX, United Kingdom
- Rainer Duden,
| |
Collapse
|
102
|
Laroche G, Giguère PM, Dupré E, Dupuis G, Parent JL. The N-terminal coiled-coil domain of the cytohesin/ARNO family of guanine nucleotide exchange factors interacts with Galphaq. Mol Cell Biochem 2007; 306:141-52. [PMID: 17846866 DOI: 10.1007/s11010-007-9564-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2007] [Accepted: 07/12/2007] [Indexed: 01/06/2023]
Abstract
Cytohesins are guanine-nucleotide exchange factors (GEF) for the Arf family of GTPases. One member of the Arf family, ARF6, plays an active role in the intracellular trafficking of G protein-coupled receptors. We have previously reported that Galphaq signaling leads to the activation of ARF6, possibly through a direct interaction with cytohesin-2/ARNO. Here, we report that Galphaq can directly interact with cytohesin-1, another Arf-GEF of the ARNO/cytohesin family. Cytohesin-1 preferentially associated with a constitutively active mutant of Galphaq (Galphaq-Q209L) compared to wild-type Galphaq in HEK293 cells. Stimulation of TPbeta, a Galphaq-coupled receptor, to activate Galphaq resulted in the promotion of a protein complex between Galphaq and cytohesin-1. Confocal immunofluorescence microscopy revealed that wild-type Galphaq and cytohesin-1 co-localized in intracellular compartments and at or near the plasma membrane. In contrast, expression of Galphaq-Q209L induced a drastic increase in the localization of cytohesin-1 at the plasma membrane. Expression of a dominant-negative mutant of cytohesin-1 reduced by 40% the agonist-induced internalization of TPbeta, a process that we previously demonstrated to be dependent on Galphaq-mediated signaling and Arf6 activation. Using deletion mutants, we show that cytohesin-1 interacts with Galphaq through its N-terminal coiled-coil domain. Cytohesin-1 and cytohesin-2/ARNO mutants lacking the coiled-coil domain were unable to relay Galphaq-mediated activation of Arf6. This is the first report of an interaction between the coiled-coil domain of the cytohesin/ARNO family of Arf-GEFs and a member of the heterotrimeric G proteins.
Collapse
Affiliation(s)
- Geneviève Laroche
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, Centre de Recherche Clinique Etienne-Lebel, University of Sherbrooke, 3001, 12th Avenue North, J1H 5N4 Fleurimont, Sherbrooke, QC, Canada
| | | | | | | | | |
Collapse
|
103
|
Li CC, Chiang TC, Wu TS, Pacheco-Rodriguez G, Moss J, Lee FJS. ARL4D recruits cytohesin-2/ARNO to modulate actin remodeling. Mol Biol Cell 2007; 18:4420-37. [PMID: 17804820 PMCID: PMC2043562 DOI: 10.1091/mbc.e07-02-0149] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
ARL4D is a developmentally regulated member of the ADP-ribosylation factor/ARF-like protein (ARF/ARL) family of Ras-related GTPases. Although the primary structure of ARL4D is very similar to that of other ARF/ARL molecules, its function remains unclear. Cytohesin-2/ARF nucleotide-binding-site opener (ARNO) is a guanine nucleotide-exchange factor (GEF) for ARF, and, at the plasma membrane, it can activate ARF6 to regulate actin reorganization and membrane ruffling. We show here that ARL4D interacts with the C-terminal pleckstrin homology (PH) and polybasic c domains of cytohesin-2/ARNO in a GTP-dependent manner. Localization of ARL4D at the plasma membrane is GTP- and N-terminal myristoylation-dependent. ARL4D(Q80L), a putative active form of ARL4D, induced accumulation of cytohesin-2/ARNO at the plasma membrane. Consistent with a known action of cytohesin-2/ARNO, ARL4D(Q80L) increased GTP-bound ARF6 and induced disassembly of actin stress fibers. Expression of inactive cytohesin-2/ARNO(E156K) or small interfering RNA knockdown of cytohesin-2/ARNO blocked ARL4D-mediated disassembly of actin stress fibers. Similar to the results with cytohesin-2/ARNO or ARF6, reduction of ARL4D suppressed cell migration activity. Furthermore, ARL4D-induced translocation of cytohesin-2/ARNO did not require phosphoinositide 3-kinase activation. Together, these data demonstrate that ARL4D acts as a novel upstream regulator of cytohesin-2/ARNO to promote ARF6 activation and modulate actin remodeling.
Collapse
Affiliation(s)
- Chun-Chun Li
- *Institute of Molecular Medicine, College of Medicine, National Taiwan University, and Department of Medical Research, National Taiwan University Hospital, Taipei 100, Taiwan; and
| | - Tsai-Chen Chiang
- *Institute of Molecular Medicine, College of Medicine, National Taiwan University, and Department of Medical Research, National Taiwan University Hospital, Taipei 100, Taiwan; and
| | - Tsung-Sheng Wu
- *Institute of Molecular Medicine, College of Medicine, National Taiwan University, and Department of Medical Research, National Taiwan University Hospital, Taipei 100, Taiwan; and
| | - Gustavo Pacheco-Rodriguez
- Pulmonary-Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1434
| | - Joel Moss
- Pulmonary-Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1434
| | - Fang-Jen S. Lee
- *Institute of Molecular Medicine, College of Medicine, National Taiwan University, and Department of Medical Research, National Taiwan University Hospital, Taipei 100, Taiwan; and
| |
Collapse
|
104
|
Famulok M, Hartig JS, Mayer G. Functional aptamers and aptazymes in biotechnology, diagnostics, and therapy. Chem Rev 2007; 107:3715-43. [PMID: 17715981 DOI: 10.1021/cr0306743] [Citation(s) in RCA: 673] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Michael Famulok
- LIMES Institute, Program Unit Chemical Biology and Medicinal Chemistry, c/o Kekulé-Institut für Organische Chemie und Biochemie, Gerhard Domagk-Strasse 1, 53121 Bonn, Germany.
| | | | | |
Collapse
|
105
|
Moore CD, Thacker EE, Larimore J, Gaston D, Underwood A, Kearns B, Patterson SI, Jackson T, Chapleau C, Pozzo-Miller L, Theibert A. The neuronal Arf GAP centaurin alpha1 modulates dendritic differentiation. J Cell Sci 2007; 120:2683-93. [PMID: 17635995 PMCID: PMC2810648 DOI: 10.1242/jcs.006346] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Centaurin alpha1 is an Arf GTPase-activating protein (GAP) that is highly expressed in the nervous system. In the current study, we show that endogenous centaurin alpha1 protein is localized in the synaptosome fraction, with peak expression in early postnatal development. In cultured dissociated hippocampal neurons, centaurin alpha1 localizes to dendrites, dendritic spines and the postsynaptic region. siRNA-mediated knockdown of centaurin alpha1 levels or overexpression of a GAP-inactive mutant of centaurin alpha1 leads to inhibition of dendritic branching, dendritic filopodia and spine-like protrusions in dissociated hippocampal neurons. Overexpression of wild-type centaurin alpha1 in cultured hippocampal neurons in early development enhances dendritic branching, and increases dendritic filopodia and lamellipodia. Both filopodia and lamellipodia have been implicated in dendritic branching and spine formation. Following synaptogenesis in cultured neurons, wild-type centaurin alpha1 expression increases dendritic filopodia and spine-like protrusions. Expression of a GAP-inactive mutant diminishes spine density in CA1 pyramidal neurons within cultured organotypic hippocampal slice cultures. These data support the conclusion that centaurin alpha1 functions through GAP-dependent Arf regulation of dendritic branching and spines that underlie normal dendritic differentiation and development.
Collapse
Affiliation(s)
- Carlene D. Moore
- Department of Neurobiology and Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Erin E. Thacker
- Department of Neurobiology and Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer Larimore
- Department of Neurobiology and Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - David Gaston
- Department of Neurobiology and Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Alison Underwood
- Department of Neurobiology and Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Brian Kearns
- Department of Neurobiology and Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sean I. Patterson
- IHEM-CONICET, Departmento de Morfo-Fisiología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Trevor Jackson
- Departments of Physiology and Dermatology, School of Clinical and Laboratory Sciences, Medical School, University of Newcastle upon Tyne, NE2 4HH, UK
| | - Chris Chapleau
- Department of Neurobiology and Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lucas Pozzo-Miller
- Department of Neurobiology and Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Anne Theibert
- Department of Neurobiology and Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Author for correspondence ()
| |
Collapse
|
106
|
Nishida M, Funahashi T, Shimomura I. Pathophysiological significance of adiponectin. Med Mol Morphol 2007; 40:55-67. [PMID: 17572841 DOI: 10.1007/s00795-007-0366-7] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2007] [Accepted: 03/16/2007] [Indexed: 12/22/2022]
Abstract
Adipose tissue, which classically has been considered as an energy-storing organ, is now viewed as a massive source of bioactive substances such as leptin, tumor necrosis factor (TNF)-alpha, and adiponectin. Adiponectin was discovered to be the most abundant adipose-specific transcript. Its function had been unclear, but epidemiological and clinical studies have demonstrated that serum levels of adiponectin are inversely associated with body weight, especially abdominal visceral fat accumulation. In addition, adiponectin was inversely related to cardiovascular risk factors, such as insulin resistance, blood pressure, and low-density lipoprotein (LDL) cholesterol and triglyceride levels, and was positively related to high-density lipoprotein (HDL) cholesterol levels. Moreover, low adiponectin concentration is associated with a high incidence of cardiovascular disease (CVD), diabetes, some kinds of cancer, and other various diseases. These associations suggest the clinical significance of adiponectin, and a number of investigations are now being conducted to clarify the biological functions of adiponectin. Recent studies have revealed that adiponectin exhibits antiinflammatory, antiatherogenic, and antidiabetic properties. In addition, adiponectin has been thought to be a key molecule in "metabolic syndrome," which is an epidemiological target for preventing cardiovascular disease. Various functions of adiponectin may possibly serve to prevent and treat obesity-related diseases and CVD. Furthermore, enhancement of adiponectin secretion or action may become a promising therapeutic target.
Collapse
Affiliation(s)
- Makoto Nishida
- Health Care Center, Osaka University 1-17 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan.
| | | | | |
Collapse
|
107
|
Heck JN, Mellman DL, Ling K, Sun Y, Wagoner MP, Schill NJ, Anderson RA. A conspicuous connection: structure defines function for the phosphatidylinositol-phosphate kinase family. Crit Rev Biochem Mol Biol 2007; 42:15-39. [PMID: 17364683 DOI: 10.1080/10409230601162752] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The phosphatidylinositol phosphate (PIP) kinases are a unique family of enzymes that generate an assortment of lipid messengers, including the pivotal second messenger phosphatidylinositol 4,5-bisphosphate (PI4,5P2). While members of the PIP kinase family function by catalyzing a similar phosphorylation reaction, the specificity loop of each PIP kinase subfamily determines substrate preference and partially influences distinct subcellular targeting. Specific protein-protein interactions that are unique to particular isoforms or splice variants play a key role in targeting PIP kinases to appropriate subcellular compartments to facilitate the localized generation of PI4,5P2 proximal to effectors, a mechanism key for the function of PI4,5P2 as a second messenger. This review documents the discovery of the PIP kinases and their signaling products, and summarizes our current understanding of the mechanisms underlying the localized generation of PI4,5P2 by PIP kinases for the regulation of cellular events including actin cytoskeleton dynamics, vesicular trafficking, cell migration, and an assortment of nuclear events.
Collapse
Affiliation(s)
- Jessica N Heck
- Program in Molecular and Cellular Pharmacology, Department of Pharmacology, University of Wisconsin-Madison, University of Wisconsin Medical School, Madison, WI 53706, USA
| | | | | | | | | | | | | |
Collapse
|
108
|
Jaworski J. ARF6 in the nervous system. Eur J Cell Biol 2007; 86:513-24. [PMID: 17559968 DOI: 10.1016/j.ejcb.2007.04.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2007] [Revised: 04/11/2007] [Accepted: 04/18/2007] [Indexed: 01/19/2023] Open
Abstract
Actin cytoskeleton dynamics and membrane trafficking are tightly connected and are among the most important driving forces of neuronal development, basic synaptic transmission events, and synaptic plasticity. One group of proteins involved in coordination of these two processes is the family of ADP ribosylation factors (ARFs) regulating actin dynamics, lipid modification and membrane trafficking. ARF6 is the only member of the ARF family that can simultaneously regulate actin cytoskeleton changes and membrane exchange between plasma membrane and endocytic compartments. The presence of ARF6 and its guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs) in the brain, as well as its capability to regulate several aspects of neuronal development and synaptic plasticity, has been recently demonstrated. The main purpose of this review is to present the current knowledge about how ARF6 can influence morphological processes crucial for proper formation of the neuronal circuits in the brain, including dendrite and axon differentiation, development of dendritic arbor complexity and dendritic spine formation. Potential effects of ARF6 on synaptic events resulting from its ability to control exo- and endocytosis will be also discussed.
Collapse
Affiliation(s)
- Jacek Jaworski
- Laboratory of Molecular and Cell Neurobiology, International Institute of Molecular and Cell Biology, Ks. Trojdena St. 4, PL-02-109, Warsaw, Poland.
| |
Collapse
|
109
|
Schindler C, Spang A. Interaction of SNAREs with ArfGAPs precedes recruitment of Sec18p/NSF. Mol Biol Cell 2007; 18:2852-63. [PMID: 17522384 PMCID: PMC1949378 DOI: 10.1091/mbc.e06-08-0756] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins are key components of the fusion machinery in vesicular transport and in homotypic membrane fusion. We previously found that ADP-ribosylation factor GTPase activating proteins (ArfGAPs) promoted a conformational change on SNAREs that allowed recruitment of the small GTPase Arf1p in stoichiometric amounts. Here, we show that the ArfGAP Gcs1p accelerates vesicle (v)-target membrane (t)-SNARE complex formation in vitro, indicating that ArfGAPs may act as folding chaperones. These SNARE complexes were resolved in the presence of ATP by the yeast homologues of alpha-soluble N-ethylmaleimide-sensitive factor attachment protein and N-ethylmaleimide-sensitive factor, Sec17p and Sec18p, respectively. In addition, Sec18p and Sec17p also recognized the "activated" SNAREs even when they were not engaged in v-t-SNARE complexes. Here again, the induction of a conformational change by ArfGAPs was essential. Surprisingly, recruitment of Sec18p to SNAREs did not require Sec17p or ATP hydrolysis. Moreover, Sec18p displaced prebound Arf1p from SNAREs, indicating that Sec18p may have more than one function: first, to ensure that all vesicle coat proteins are removed from the SNAREs before the engagement in a trans-SNARE complex; and second, to resolve cis-SNARE complexes after fusion has occurred.
Collapse
Affiliation(s)
| | - Anne Spang
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| |
Collapse
|
110
|
Wessels E, Duijsings D, Lanke KHW, Melchers WJG, Jackson CL, van Kuppeveld FJM. Molecular determinants of the interaction between coxsackievirus protein 3A and guanine nucleotide exchange factor GBF1. J Virol 2007; 81:5238-45. [PMID: 17329336 PMCID: PMC1900206 DOI: 10.1128/jvi.02680-06] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2006] [Accepted: 02/19/2007] [Indexed: 11/20/2022] Open
Abstract
The 3A protein of coxsackievirus B3 (CVB3), a small membrane protein that forms homodimers, inhibits endoplasmic reticulum-to-Golgi complex transport. Recently, we described the underlying mechanism by showing that the CVB3 3A protein binds to and inhibits the function of GBF1, a guanine nucleotide exchange factor for ADP-ribosylation factor 1 (Arf1), thereby interfering with Arf1-mediated COP-I recruitment. This study was undertaken to gain more insight into the molecular determinants underlying the interaction between 3A and GBF1. Here we show that 3A mutants that have lost the ability to dimerize are no longer able to bind to GBF1 and trap it on membranes. Moreover, we identify a conserved region in the N terminus of 3A that is crucial for GBF1 binding but not for 3A dimerization. Analysis of the binding domain in GBF1 showed that the extreme N terminus, the dimerization/cyclophilin binding domain, and the homology upstream of Sec7 domain are required for the interaction with 3A. In contrast to that of full-length GBF1, overexpression of a GBF1 mutant lacking its extreme N terminus failed to rescue the effects of 3A. Together, these data provide insight into the molecular requirements of the interaction between 3A and GBF1.
Collapse
Affiliation(s)
- Els Wessels
- Department of Medical Microbiology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | | | | | | | | | | |
Collapse
|
111
|
Luo R, Ahvazi B, Amariei D, Shroder D, Burrola B, Losert W, Randazzo P. Kinetic analysis of GTP hydrolysis catalysed by the Arf1-GTP-ASAP1 complex. Biochem J 2007; 402:439-47. [PMID: 17112341 PMCID: PMC1863566 DOI: 10.1042/bj20061217] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2006] [Revised: 11/09/2006] [Accepted: 11/20/2006] [Indexed: 11/17/2022]
Abstract
Arf (ADP-ribosylation factor) GAPs (GTPase-activating proteins) are enzymes that catalyse the hydrolysis of GTP bound to the small GTP-binding protein Arf. They have also been proposed to function as Arf effectors and oncogenes. We have set out to characterize the kinetics of the GAP-induced GTP hydrolysis using a truncated form of ASAP1 [Arf GAP with SH3 (Src homology 3) domain, ankyrin repeats and PH (pleckstrin homology) domains 1] as a model. We found that ASAP1 used Arf1-GTP as a substrate with a k(cat) of 57+/-5 s(-1) and a K(m) of 2.2+/-0.5 microM determined by steady-state kinetics and a kcat of 56+/-7 s(-1) determined by single-turnover kinetics. Tetrafluoroaluminate (AlF4-), which stabilizes complexes of other Ras family members with their cognate GAPs, also stabilized a complex of Arf1-GDP with ASAP1. As anticipated, mutation of Arg-497 to a lysine residue affected kcat to a much greater extent than K(m). Changing Trp-479, Iso-490, Arg-505, Leu-511 or Asp-512 was predicted, based on previous studies, to affect affinity for Arf1-GTP. Instead, these mutations primarily affected the k(cat). Mutants that lacked activity in vitro similarly lacked activity in an in vivo assay of ASAP1 function, the inhibition of dorsal ruffle formation. Our results support the conclusion that the Arf GAP ASAP1 functions in binary complex with Arf1-GTP to induce a transition state towards GTP hydrolysis. The results have led us to speculate that Arf1-GTP-ASAP1 undergoes a significant conformational change when transitioning from the ground to catalytically active state. The ramifications for the putative effector function of ASAP1 are discussed.
Collapse
Key Words
- arf gap with sh3
- ankyrin repeats and ph domains 1 (asap1)
- adp-ribosylation factor (arf)
- gtpase-activating protein (gap)
- gtp-binding protein
- gtp hydrolysis
- kinetics
- ank, ankyrin repeat
- arf, adp-ribosylation factor
- asap, arf gap with sh3 (src homology 3), anks and ph domains
- dtt, dithiothreitol
- gap, gtpase-activating protein
- gst, glutathione s-transferase
- ha, haemagglutinin
- luvs, large unilamellar vesicles
- myrarf1, myristoylated arf1
- pap, phosphatidic acid phosphohydrolase
- pdgf, platelet-derived growth factor
- ph domain, pleckstrin homology domain
Collapse
Affiliation(s)
- Ruibai Luo
- *Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, U.S.A
| | - Bijan Ahvazi
- †National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, MD 20892, U.S.A
| | - Diana Amariei
- *Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, U.S.A
| | - Deborah Shroder
- *Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, U.S.A
| | - Beatriz Burrola
- ‡Department of Physics and IPST (Institute for Physical Sciences and Technology), University of Maryland, College Park, MD 20742, U.S.A
| | - Wolfgang Losert
- ‡Department of Physics and IPST (Institute for Physical Sciences and Technology), University of Maryland, College Park, MD 20742, U.S.A
| | - Paul A. Randazzo
- *Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, U.S.A
| |
Collapse
|
112
|
Sakagami H, Honma T, Sukegawa J, Owada Y, Yanagisawa T, Kondo H. Somatodendritic localization of EFA6A, a guanine nucleotide exchange factor for ADP-ribosylation factor 6, and its possible interaction with α-actinin in dendritic spines. Eur J Neurosci 2007; 25:618-28. [PMID: 17298598 DOI: 10.1111/j.1460-9568.2007.05345.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
EFA6A is a member of the guanine nucleotide exchange factors that can specifically activate ADP ribosylation factor 6 (ARF6). In this study, we identified alpha-actinin-1 as a possible interacting protein with EFA6A by the yeast two-hybrid screening with its C-terminal region as bait. The central region of alpha-actinin-1 containing a part of spectrin repeat 1 and spectrin repeats 2-3 is responsible for this interaction. In the hippocampal formation, EFA6A immunoreactivity occurred at a high level as numerous fine puncta in the strata oriens, radiatum, lacunosum-moleculare of the hippocampal CA1-3 subfields and the dentate molecular layer, whereas the immunoreactivity was faint in the neuronal cell layers and the stratum lucidum, the mossy fiber-recipient layer of the CA3 subfield. Double-immunofluorescent analyses revealed a partial overlapping of EFA6A and alpha-actinin at the dendritic spines of in vivo and cultured hippocampal neurons. Our present findings suggest that EFA6A may form a protein complex with alpha-actinin and activate ARF6 in close proximity of the actin cytoskeleton and membrane proteins in the dendritic spines.
Collapse
Affiliation(s)
- Hiroyuki Sakagami
- Division of Histology, Department of Cell Biology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Sendai 980-8575, Japan.
| | | | | | | | | | | |
Collapse
|
113
|
Kerr ID, Bennett MJ. New insight into the biochemical mechanisms regulating auxin transport in plants. Biochem J 2007; 401:613-22. [PMID: 17209803 PMCID: PMC1770846 DOI: 10.1042/bj20061411] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The transport of the plant hormone auxin has been under intense investigation since its identification 80 years ago. Studies have gradually refined our understanding of the importance of auxin transport in many aspects of plant signalling and development, and the focus has intensified in recent years towards the identification of the proteins involved in auxin transport and their functional mechanism. Within the past 18 months, the field has progressed rapidly, with confirmation that several distinct classes of proteins, previously dubbed as 'putative auxin permeases' or 'auxin transport facilitators', are bona fide transporters of IAA (indol-3-ylacetic acid). In this review we will appraise the recent transport data and highlight likely future research directions, including the characterization of auxiliary proteins necessary for the regulation of auxin transporters.
Collapse
Affiliation(s)
- Ian D Kerr
- School of Biomedical Sciences, University of Nottingham, Queen's Medical Centre, Nottingham NG7 2UH, UK.
| | | |
Collapse
|
114
|
Yu X, Prekeris R, Gould GW. Role of endosomal Rab GTPases in cytokinesis. Eur J Cell Biol 2007; 86:25-35. [PMID: 17157409 DOI: 10.1016/j.ejcb.2006.10.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2006] [Revised: 10/24/2006] [Accepted: 10/26/2006] [Indexed: 01/02/2023] Open
Abstract
Completion of cytokinesis requires Rab 11-dependent membrane trafficking events to deliver new membrane to the furrow and for abscission. Many Rabs have overlapping endosomal distributions, hence, we examined whether these Rabs also function in cytokinesis. Analysis of the distribution of Rabs 4, 5, 7, 8, 9, 11, 21, and 22 revealed that only Rab 11 was enriched within the furrow of cells in telophase or present within the midbody. By contrast, Rabs 4, 5, 7, 8, and 9 were mainly localised within a peri-nuclear compartment facing away from the furrow. Using RNA interference and dominant negative Rab mutants, we evaluated the role of these Rabs in furrowing and abscission. Consistent with previous work, we find that Rab 11 is intimately involved in abscission. However, we further found that depletion of Rab 4 slowed but did not prevent abscission. Depletion of any other Rab species had little effect on furrowing or abscission. These data suggest that the membrane trafficking events required for completion of cytokinesis are largely controlled by Rab 11 and not other endosomal Rab proteins, and further suggest that the relocation of Rab 11-specific cargo is an integral facet of abscission. Arf6 knockdown was without effect on cytokinesis, but when both Rab 11 and Arf6 were knocked-down, we found the furrow rapidly regressed and the cells were unable to form a stable midbody. We suggest that Rab 11 and Arf6 function synergistically in the switch from furrowing to abscission, as well as in the terminal stage of abscission.
Collapse
Affiliation(s)
- Xinzi Yu
- Henry Wellcome Laboratory of Cell Biology, Division of Biochemistry and Molecular Biology, Institute of Biomedical and Life Sciences, University of Glasgow, Davidson Building, Glasgow G12 8QQ, Scotland, UK
| | | | | |
Collapse
|
115
|
Abstract
Cell adhesion, migration and the maintenance of cell polarity are all processes that depend on the correct targeting of integrins and the dynamic remodelling of integrin-containing adhesion sites. The importance of the endo/exocytic cycle of integrins as a key regulator of these functions is increasingly recognized. Several recent publications have provided mechanistic insight into how integrin traffic is regulated in cells. Increasing evidence suggests that small GTPases such as Arf6 and members of the Rab family control integrin internalization and recycling back to the plasma membrane along microtubules. The fine tuning of these trafficking events seems to be mediated by specific guanine-nucleotide-exchange factors (GEFs) and GTPase-activating proteins (GAPs). In addition, several kinases regulate integrin traffic. The identification of their substrates has demonstrated how these kinases regulate integrin traffic by controlling small GTPases or stabilizing cytoskeletal tracks that are crucial for efficient traffic of integrins to the plasma membrane.
Collapse
|
116
|
Lay D, Gorgas K, Just WW. Peroxisome biogenesis: Where Arf and coatomer might be involved. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2006; 1763:1678-87. [PMID: 17023067 DOI: 10.1016/j.bbamcr.2006.08.036] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2006] [Revised: 08/12/2006] [Accepted: 08/23/2006] [Indexed: 11/24/2022]
Abstract
The present review summarizes recent observations on binding of Arf and COPI coat to isolated rat liver peroxisomes. The general structural and functional features of both Arf and coatomer were considered along with the requirements and dependencies of peroxisomal Arf and coatomer recruitment. Studies on the expression of mammalian Pex11 proteins, mainly Pex11alpha and Pex11beta, intimately related to the process of peroxisome proliferation, revealed a sequence of individual steps including organelle elongation/tubulation, formation of membrane and matrix protein patches segregating distinct proteins from each other, development of membrane constrictions and final membrane fission. Based on the similarities of the processes leading to cargo selection and concentration on Golgi membranes on the one hand and to the formation of peroxisomal protein patches on the other hand, an implication of Arf and COPI in distinct processes of peroxisomal proliferation is hypothesized. Alternatively, peroxisomal Arf/COPI might facilitate the formation of COPI-coated peroxisomal vesicles functioning in cargo transport and retrieval from peroxisomes to the ER. Recent observations suggesting transport of Pex3 and Pex19 during early steps of peroxisome biogenesis from the ER to peroxisomes inevitably propose such a retrieval mechanism, provided the ER to peroxisome pathway is based on transporting vesicles.
Collapse
Affiliation(s)
- Dorothee Lay
- Biochemie-Zentrum der Universität Heidelberg (BZH), Im Neuenheimer Feld 328D-69120 Heidelberg, Germany
| | | | | |
Collapse
|
117
|
Shultz T, Nash-Livni N, Shmuel M, Altschuler Y. EFA6 regulates endosomal trafficking and affects early endosomes in polarized MDCK cells. Biochem Biophys Res Commun 2006; 351:106-12. [PMID: 17054918 DOI: 10.1016/j.bbrc.2006.10.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2006] [Accepted: 10/03/2006] [Indexed: 12/21/2022]
Abstract
The small-GTPase family of ADP ribosylation factors (ARFs) recruit coat proteins to promote vesicle budding. ARFs are activated by an association with sec7-containing exchange factors which load them with GTP. In epithelial cells, the small GTPase ARF6 operates within the endocytic system and has been shown to associate with ARNO to promote apical endocytosis and early to late endosomal trafficking. EFA6 has been shown to stimulate tight-junction formation and maintenance. Here, we show that in polarized epithelial MDCK cells, EFA6 is localized to early endosomes, causes their dramatic enlargement, and promotes basolateral targeting of IgA, which is normally targeted to the apical PM. These results suggest that the physiological function of ARF6 within the endocytic system is regulated by the exchange factor it associates with.
Collapse
Affiliation(s)
- Tamar Shultz
- Department of Pharmacology, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | | | | | | |
Collapse
|
118
|
Tse YC, Lo SW, Hillmer S, Dupree P, Jiang L. Dynamic response of prevacuolar compartments to brefeldin a in plant cells. PLANT PHYSIOLOGY 2006; 142:1442-59. [PMID: 17041023 PMCID: PMC1676059 DOI: 10.1104/pp.106.090423] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2006] [Accepted: 10/09/2006] [Indexed: 05/12/2023]
Abstract
Little is known about the dynamics and molecular components of plant prevacuolar compartments (PVCs) in the secretory pathway. Using transgenic tobacco (Nicotiana tabacum) Bright-Yellow-2 (BY-2) cells expressing membrane-anchored yellow fluorescent protein (YFP) reporters marking Golgi or PVCs, we have recently demonstrated that PVCs are mobile multivesicular bodies defined by vacuolar sorting receptor proteins. Here, we demonstrate that Golgi and PVCs have different sensitivity in response to brefeldin A (BFA) treatment in living tobacco BY-2 cells. BFA at low concentrations (5-10 microg mL(-1)) induced YFP-marked Golgi stacks to form both endoplasmic reticulum-Golgi hybrid structures and BFA-induced aggregates, but had little effect on YFP-marked PVCs in transgenic BY-2 cells at both confocal and immunogold electron microscopy levels. However, BFA at high concentrations (50-100 microg mL(-1)) caused both YFP-marked Golgi stacks and PVCs to form aggregates in a dose- and time-dependent manner. Normal Golgi or PVC signals can be recovered upon removal of BFA from the culture media. Confocal immunofluorescence and immunogold electron microscopy studies with specific organelle markers further demonstrate that the PVC aggregates are distinct, but physically associated, with Golgi aggregates in BFA-treated cells and that PVCs might lose their internal vesicle structures at high BFA concentration. In addition, vacuolar sorting receptor-marked PVCs in root-tip cells of tobacco, pea (Pisum sativum), mung bean (Vigna radiata), and Arabidopsis (Arabidopsis thaliana) upon BFA treatment are also induced to form similar aggregates. Thus, we have demonstrated that the effects of BFA are not limited to endoplasmic reticulum and Golgi, but extend to PVC in the endomembrane system, which might provide a quick tool for distinguishing Golgi from PVC for its identification and characterization, as well as a possible new tool in studying PVC-mediated protein traffic in plant cells.
Collapse
Affiliation(s)
- Yu Chung Tse
- Department of Biology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | | | | | | | | |
Collapse
|
119
|
Yoon HY, Miura K, Cuthbert EJ, Davis KK, Ahvazi B, Casanova JE, Randazzo PA. ARAP2 effects on the actin cytoskeleton are dependent on Arf6-specific GTPase-activating-protein activity and binding to RhoA-GTP. J Cell Sci 2006; 119:4650-66. [PMID: 17077126 DOI: 10.1242/jcs.03237] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
ARAP2 is a protein that contains both ArfGAP and RhoGAP domains. We found that it is a phosphatidylinositol (3,4,5)-trisphosphate-dependent Arf6 GAP that binds RhoA-GTP but lacks RhoGAP activity. In agreement with the hypothesis that ARAP2 mediates effects of RhoA, endogenous ARAP2 associated with focal adhesions (FAs) and reduction of ARAP2 expression, by RNAi, resulted in fewer FAs and actin stress fibers (SFs). In cells with reduced levels of endogenous ARAP2, FAs and SFs could be restored with wild-type recombinant ARAP2 but not mutants lacking ArfGAP or Rho-binding activity. Constitutively active Arf6 also caused a loss of SFs. The Rho effector ROKα was ineffective in restoring FAs. Conversely, overexpression of ARAP2 did not restore SFs in cells treated with a ROK inhibitor but induced punctate accumulations of paxillin. We conclude that ARAP2 is an Arf6GAP that functions downstream of RhoA to regulate focal adhesion dynamics.
Collapse
Affiliation(s)
- Hye-Young Yoon
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Department of Health and Human Services, Building 37, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
120
|
Murphy JA, Jensen ON, Walikonis RS. BRAG1, a Sec7 domain-containing protein, is a component of the postsynaptic density of excitatory synapses. Brain Res 2006; 1120:35-45. [PMID: 17045249 DOI: 10.1016/j.brainres.2006.08.096] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2006] [Revised: 08/16/2006] [Accepted: 08/21/2006] [Indexed: 12/13/2022]
Abstract
The postsynaptic density (PSD) at excitatory synapses is a dynamic complex of glutamatergic receptors and associated proteins that governs synaptic structure and coordinates signal transduction. In this study, we report that BRAG1, a putative guanine nucleotide exchange factor for the Arf family of GTP-binding proteins, is a major component of the PSD. BRAG1 was identified in a 190 kDa band in the PSD fraction with the use of mass spectrometry coupled to searching of a protein sequence database. BRAG1 expression is abundant in the adult rat forebrain, and it is strongly enriched in the PSD fraction compared to forebrain homogenate and synaptosomes. Immunocytochemical localization of BRAG1 in dissociated hippocampal neurons shows that it forms discrete clusters that colocalize with the postsynaptic marker PSD-95 at sites along dendrites. BRAG1 contains a Sec7 domain, a domain that catalyzes exchange of GDP for GTP on the Arf family of small GTP-binding proteins. In their GTP-bound active state, Arfs regulate trafficking of vesicles and cytoskeletal structure. We demonstrate that the Sec7 domain of BRAG1 promotes binding of GTP to Arf in vitro. These data suggest that BRAG1 may modulate the functions of Arfs at synaptic sites.
Collapse
Affiliation(s)
- Jessica A Murphy
- Department of Physiology and Neurobiology, University of Connecticut, 75 North Eagleville Road, U-3156, Storrs, CT 06269, USA
| | | | | |
Collapse
|
121
|
Jeynov B, Lay D, Schmidt F, Tahirovic S, Just WW. Phosphoinositide synthesis and degradation in isolated rat liver peroxisomes. FEBS Lett 2006; 580:5917-24. [PMID: 17045591 DOI: 10.1016/j.febslet.2006.09.058] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2006] [Revised: 09/18/2006] [Accepted: 09/19/2006] [Indexed: 11/30/2022]
Abstract
Analyzing peroxisomal phosphoinositide (PId(#)) synthesis in highly purified rat liver peroxisomes we found synthesis of phosphatidylinositol 4-phosphate (PtdIns4P), PtdIns(4,5)P(2) and PtdIns(3,5)P(2). PtdIns3P was hardly detected in vitro, however, was observed in vivo after [(32)P]-phosphate labeling of primary rat hepatocytes. In comparison with other subcellular organelles peroxisomes revealed a unique PId pattern suggesting peroxisomal specificity of the observed synthesis. Use of phosphatase inhibitors enhanced the amount of PtdIns4P. The results obtained provide evidence that isolated rat liver peroxisomes synthesize PIds and suggest the association of PId 4-kinase and PId 5-kinase and PId 4-phosphatase activities with the peroxisomal membrane.
Collapse
Affiliation(s)
- Boyan Jeynov
- Biochemie-Zentrum der Universität Heidelberg (BZH), Im Neuenheimer Feld 328, D-69120 Heidelberg, Germany
| | | | | | | | | |
Collapse
|
122
|
Deretic D. A role for rhodopsin in a signal transduction cascade that regulates membrane trafficking and photoreceptor polarity. Vision Res 2006; 46:4427-33. [PMID: 17010408 DOI: 10.1016/j.visres.2006.07.028] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2006] [Revised: 07/24/2006] [Accepted: 07/26/2006] [Indexed: 11/25/2022]
Abstract
This review summarizes the most recent progress in the understanding of the role of rhodopsin C-terminal domain in the regulation of intracellular trafficking and photoreceptor morphogenesis. A proposed cascade of molecular interactions, initiated by the rhodopsin C-terminal sequence VXPX-COOH during trafficking from the Golgi/TGN in retinal photoreceptors, is relayed by the small GTPase ARF4 to the downstream effectors. One of the candidates for an ARF4 effector is the ARF-GAP ASAP1, which may function as a subunit of, or form a novel protein coat involved in trafficking from the TGN and in cytoskeletal remodeling, whose assembly is regulated by the binding of ARF4 to rhodopsin, and whose function is essential for the polarized trafficking toward the ROS.
Collapse
Affiliation(s)
- Dusanka Deretic
- Department of Surgery, Division of Ophthalmology, University of New Mexico, Albuquerque, NM 87131, USA.
| |
Collapse
|
123
|
Wessels E, Duijsings D, Lanke KHW, van Dooren SHJ, Jackson CL, Melchers WJG, van Kuppeveld FJM. Effects of picornavirus 3A Proteins on Protein Transport and GBF1-dependent COP-I recruitment. J Virol 2006; 80:11852-60. [PMID: 17005635 PMCID: PMC1642585 DOI: 10.1128/jvi.01225-06] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The 3A protein of the coxsackievirus B3 (CVB3), an enterovirus that belongs to the family of the picornaviruses, inhibits endoplasmic reticulum-to-Golgi transport. Recently, we elucidated the underlying mechanism by showing that CVB3 3A interferes with ADP-ribosylation factor 1 (Arf1)-dependent COP-I recruitment to membranes by binding and inhibiting the function of GBF1, a guanine nucleotide exchange factor that is required for the activation of Arf1 (E. Wessels et al., Dev. Cell 11:191-201, 2006). Here, we show that the 3A protein of poliovirus, another enterovirus, is also able to interfere with COP-I recruitment through the same mechanism. No interference with protein transport or COP-I recruitment was observed for the 3A proteins of any of the other picornaviruses tested here (human rhinovirus [HRV], encephalomyocarditis virus, foot-and-mouth disease virus, and hepatitis A virus). We show that the 3A proteins of HRV, which are the most closely related to the enteroviruses, are unable to inhibit COP-I recruitment, due to a reduced ability to bind GBF1. When the N-terminal residues of the HRV 3A proteins are replaced by those of CVB3 3A, chimeric proteins are produced that have gained the ability to bind GBF1 and, by consequence, to inhibit protein transport. These results show that the N terminus of the CVB3 3A protein is important for binding of GBF1 and its transport-inhibiting function. Taken together, our data demonstrate that the activity of the enterovirus 3A protein to inhibit GBF1-dependent COP-I recruitment is unique among the picornaviruses.
Collapse
Affiliation(s)
- Els Wessels
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
124
|
Chen S, Wang J, Muthusamy BP, Liu K, Zare S, Andersen RJ, Graham TR. Roles for the Drs2p-Cdc50p Complex in Protein Transport and Phosphatidylserine Asymmetry of the Yeast Plasma Membrane. Traffic 2006; 7:1503-17. [PMID: 16956384 DOI: 10.1111/j.1600-0854.2006.00485.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Drs2p, a P-type adenosine triphosphatase required for a phosphatidylserine (PS) flippase activity in the yeast trans Golgi network (TGN), was first implicated in protein trafficking by a screen for mutations synthetically lethal with arf1 (swa). Here, we show that SWA4 is allelic to CDC50, encoding a membrane protein previously shown to chaperone Drs2p from the endoplasmic reticulum to the Golgi complex. We find that cdc50Delta exhibits the same clathrin-deficient phenotypes as drs2Delta, including delayed transport of carboxypeptidase Y to the vacuole, mislocalization of resident TGN enzymes and the accumulation of aberrant membrane structures. These trafficking defects precede appearance of cell polarity defects in cdc50Delta, suggesting that the latter are a secondary consequence of disrupting Golgi function. Involvement of Drs2p-Cdc50p in PS translocation suggests a role in restricting PS to the cytosolic leaflet of the Golgi and plasma membrane. Annexin V binding and papuamide B hypersensitivity indicate that drs2Delta or cdc50Delta causes a loss of plasma membrane PS asymmetry. However, clathrin and other endocytosis null mutants also exhibit a comparable loss of PS asymmetry, and studies with drs2-ts and clathrin (chc1-ts) conditional mutants suggest that loss of plasma membrane asymmetry is a secondary consequence of disrupting protein trafficking.
Collapse
Affiliation(s)
- Sophie Chen
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA
| | | | | | | | | | | | | |
Collapse
|
125
|
Santarius M, Lee C, Anderson R. Supervised membrane swimming: small G-protein lifeguards regulate PIPK signalling and monitor intracellular PtdIns(4,5)P2 pools. Biochem J 2006; 398:1-13. [PMID: 16856876 PMCID: PMC1525017 DOI: 10.1042/bj20060565] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Regulation of PIPK (phosphatidylinositol phosphate kinase) and PtdIns(4,5)P2 signalling by small G-proteins and their effectors is key to many biological functions. Through selective recruitment and activation of different PIPK isoforms, small G-proteins such as Rho, Rac and Cdc42 modulate actin dynamics and cytoskeleton-dependent cellular events in response to extracellular signalling. These activities affect a number of processes, including endocytosis, bacterial penetration into host cells and cytolytic granule-mediated targeted cell killing. Small G-proteins and their modulators are also regulated by phosphoinositides through translocation and conformational changes. Arf family small G-proteins act at multiple sites as regulators of membrane trafficking and actin cytoskeletal remodelling, and regulate a feedback loop comprising phospholipase D, phosphatidic acid, PIPKs and PtdIns(4,5)P2, contributing to enhancement of PtdIns(4,5)P2-mediated cellular events and receptor signalling. Na+, Kir (inwardly rectifying K+), Ca2+ and TRP (transient receptor potential) ion channels are regulated by small G-proteins and membrane pools of PtdIns(4,5)P2. Yeast phosphatidylinositol 4-phosphate 5-kinases Mss4 and Its3 are involved in resistance against disturbance of sphingolipid biosynthesis and maintenance of cell integrity through the synthesis of PtdIns(4,5)P2 and downstream signalling through the Rom2/Rho2 and Rgf1/Rho pathways. Here, we review models for regulated intracellular targeting of PIPKs by small G-proteins and other modulators in response to extracellular signalling. We also describe the spatial and temporal cross-regulation of PIPKs and small G-proteins that is critical for a number of cellular functions.
Collapse
Affiliation(s)
- Megan Santarius
- *Program in Molecular and Cellular Pharmacology, University of Wisconsin-Madison, 1300 University Avenue, Madison, WI 53706, U.S.A
| | - Chang Ho Lee
- †Department of Pharmacology, College of Medicine, Hanyang University, 17 Hengdang-dong, Seongdong-ku, Seoul, 133-791, South Korea
- To whom correspondence should be addressed (email )
| | - Richard A. Anderson
- *Program in Molecular and Cellular Pharmacology, University of Wisconsin-Madison, 1300 University Avenue, Madison, WI 53706, U.S.A
- ‡Department of Pharmacology, University of Wisconsin Medical School, 1300 University Avenue, Madison, WI 53706, U.S.A
| |
Collapse
|
126
|
Ishizaki R, Shin HW, Iguchi-Ariga SMM, Ariga H, Nakayama K. AMY-1 (associate of Myc-1) localization to the trans-Golgi network through interacting with BIG2, a guanine-nucleotide exchange factor for ADP-ribosylation factors. Genes Cells 2006; 11:949-59. [PMID: 16866877 DOI: 10.1111/j.1365-2443.2006.00991.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: 11/27/2022]
Abstract
AMY-1 (associate of Myc-1) was originally identified as a c-Myc-binding protein that enhances the c-Myc transcription activity, and subsequently found to interact with A-kinase-anchoring proteins (AKAPs), including AKAP149, S-AKAP84 and AKAP95. We show here that, using anti-AMY-1 antibodies we raised, AMY-1 localizes to the trans-Golgi network (TGN) and the nucleus. To explore the possible function of AMY-1, we have undertaken a search for interacting partners by co-immunoprecipitation experiments using cells stably expressing FLAG-tagged AMY-1. Interestingly, we have found that AMY-1 interacts with BIG2 and BIG1, both of which are high molecular weight guanine-nucleotide exchange factors for ADP-ribosylation factors (ARFs) and mainly localize to the TGN. Furthermore, we have demonstrated that AMY-1 is associated with the TGN through interacting with BIG2 but not with BIG1 using an RNA interference approach, although AMY-1 can interact with both BIG1 and BIG2 in vitro. Taken together with the facts that BIG2 contains domains that bind to regulatory subunits of protein kinase A and that recruitment of ARF1 onto Golgi membranes is mediated, at least in part, by activation of protein kinase A, these results suggest that BIG2 alone or in concert with recruited AMY-1 coordinates ARF-mediated membrane trafficking and signaling pathways.
Collapse
Affiliation(s)
- Ray Ishizaki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | | | | | | | | |
Collapse
|
127
|
Someya A, Moss J, Nagaoka I. Involvement of a guanine nucleotide-exchange protein, ARF-GEP100/BRAG2a, in the apoptotic cell death of monocytic phagocytes. J Leukoc Biol 2006; 80:915-21. [PMID: 16877676 DOI: 10.1189/jlb.0106059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We previous identified adenosine 5'-diphosphate-ribosylation factor (ARF)-guanine nucleotide-exchange protein, 100 kDa (GEP(100)), as a novel GEP with a molecular size of approximately 100 kDa, which preferentially activates ARF6. In this study, we examined the effect of ARF-GEP(100) on monocytic cell apoptosis. Overexpression of ARF-GEP(100) in PMA-differentiated human monocyte-macrophage-like U937 cells and mouse macrophage RAW264.7 cells induced apoptotic cell death, which was detected by morphological changes (chromatin condensation, nucleus fragmentation, and shrinking of cytoplasm), annexin V-staining, and TUNEL assay. It is interesting that a mutant lacking the Sec7 domain, which is responsible for ARF activation, was able to induce apoptosis of the target cells to the level of that of a wild-type ARF-GEP(100). Furthermore, ARF-GEP(100)-silencing experiments indicated that the TNF-alpha-induced apoptosis was significantly suppressed among ARF-GEP(100)-depressed cells. These observations apparently suggest that ARF-GEP(100) is involved in the induction of apoptosis in monocytic phagocytes, possibly independent of ARF activation.
Collapse
Affiliation(s)
- Akimasa Someya
- Department of Host Defense and Biochemical Research, Juntendo University School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan.
| | | | | |
Collapse
|
128
|
Walker DH, Yu XJ. Progress in rickettsial genome analysis from pioneering of Rickettsia prowazekii to the recent Rickettsia typhi. Ann N Y Acad Sci 2006; 1063:13-25. [PMID: 16481486 DOI: 10.1196/annals.1355.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Three rickettsial genomes have been sequenced and annotated. Rickettsia prowazekii and R. typhi have similar gene order and content. The few differences between R. prowazekii and R. typhi include a 12-kb insertion in R. prowazekii, a large inversion close to the origin of replication in R. typhi, and loss of the complete cytochrome c oxidase system by R. typhi. R. prowazekii, R. typhi, and R. conorii have 13, 24, and 560 unique genes, respectively, and share 775 genes, most likely their essential genes. The small genomes contain many pseudogenes and much noncoding DNA, reflecting the process of genome decay. R. typhi contains the largest number of pseudogenes (41), and R. conorii the fewest, in accordance with its larger number of genes and smaller proportion of noncoding DNA. Conversely, typhus rickettsiae contain fewer repetitive sequences. These genomes portray the key themes of rickettsial intracellular survival: lack of enzymes for sugar metabolism, lipid biosynthesis, nucleotide synthesis, and amino acid metabolism, suggesting that rickettsiae depend on the host for nutrition and building blocks; enzymes for the complete TCA cycle and several copies of ATP/ADP translocase genes, suggesting independent synthesis of ATP and acquisition of host ATP; and type IV secretion system. All rickettsiae share two outer membrane proteins (OmpB and Sca 4) and LPS biosynthesis machinery. RickA, unique to spotted fever rickettsiae, plays a role in induction of actin polymerization in R. conorii, but not in R. prowazekii or R. typhi. The genome of R. typhi contains four potentially membranolytic genes (tlyA, tlyC, pldA, and pat-1) and five autotransporter genes, sca 1, sca 2, sca 3, ompA, and ompB. The presence of six 50-amino acid repeat units in Sca 2 suggests function as an adhesin. The high laboratory passage of the sequenced strains raises the issue of the occurrence of laboratory mutations in genes not required for growth in cell culture or eggs. Resequencing revealed that eight annotated pseudogenes of E strain are actually intact genes. Comparative genomics of virulent and avirulent strains of rickettsial species may reveal their virulence factors.
Collapse
Affiliation(s)
- David H Walker
- Department of Pathology, University of Texas Medical Branch-Galveston, TX 77555-0609, USA.
| | | |
Collapse
|
129
|
Anadu NO, Davisson VJ, Cushman M. Synthesis and Anticancer Activity of Brefeldin A Ester Derivatives. J Med Chem 2006; 49:3897-905. [PMID: 16789745 DOI: 10.1021/jm0602817] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Ester derivatives of brefeldin A (BFA) were synthesized to determine which of its two hydroxyl groups could be modified while still maintaining biological activity. The compounds were tested for antiproliferative activity in the National Cancer Institute's 60 cancer cell line screen. Monoderivatization at the C4 and C7 alcohols was tolerated, yielding biologically active compounds, whereas the analogues derivatized at both positions were the least active in the series. Molecular modeling of the analogues revealed that both the C4 and C7 derivatives were well tolerated at the interface between ARF1 and its guanine nucleotide exchange factor ARNO. The Golgi-disruptive properties of the analogues were determined using fluorescence imaging assays. The BFA ester conjugates synthesized in this study were cytotoxic to cancer cells, and we have shown that the disruption of the Golgi complex is not necessary for cytotoxicity. The brefeldin A ester derivatives are potential anticancer agents.
Collapse
Affiliation(s)
- Nwanne O Anadu
- Department of Medicinal Chemistry and Molecular Pharmacology, School of Pharmacy and Pharmaceutical Sciences, and the Purdue Cancer Center, Purdue University, West Lafayette, Indiana 47907, USA
| | | | | |
Collapse
|
130
|
Yahara N, Sato K, Nakano A. The Arf1p GTPase-activating protein Glo3p executes its regulatory function through a conserved repeat motif at its C-terminus. J Cell Sci 2006; 119:2604-12. [PMID: 16735437 DOI: 10.1242/jcs.02997] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
ADP-ribosylation factors (Arfs), key regulators of intracellular membrane traffic, are known to exert multiple roles in vesicular transport. We previously isolated eight temperature-sensitive (ts) mutants of the yeast ARF1 gene, which showed allele-specific defects in protein transport, and classified them into three groups of intragenic complementation. In this study, we show that the overexpression of Glo3p, one of the GTPase-activating proteins of Arf1p (ArfGAP), suppresses the ts growth of a particular group of the arf1 mutants (arf1-16 and arf1-17). Other ArfGAPs do not show such a suppression activity. All these ArfGAPs show sequence similarity in the ArfGAP catalytic domain, but are divergent in the rest of molecules. By domain swapping analysis of Glo3p and another ArfGAP, Gcs1p, we have shown that the non-catalytic C-terminal region of Glo3p is required for the suppression of the growth defect in the arf1 ts mutants. Interestingly, Glo3p and its homologues from other eukaryotes harbor a well-conserved repeated ISSxxxFG sequence near the C-terminus, which is not found in Gcs1p and its homologues. We name this region the Glo3 motif and present evidence that the motif is required for the function of Glo3p in vivo.
Collapse
Affiliation(s)
- Natsuko Yahara
- Molecular Membrane Biology Laboratory, RIKEN Discovery Research Institute, Hirosawa, Wako, Saitama 351-0198, Japan
| | | | | |
Collapse
|
131
|
Sakagami H, Suzuki H, Kamata A, Owada Y, Fukunaga K, Mayanagi H, Kondo H. Distinct spatiotemporal expression of EFA6D, a guanine nucleotide exchange factor for ARF6, among the EFA6 family in mouse brain. Brain Res 2006; 1093:1-11. [PMID: 16707115 DOI: 10.1016/j.brainres.2006.02.058] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2005] [Revised: 01/30/2006] [Accepted: 02/13/2006] [Indexed: 11/29/2022]
Abstract
The EFA6 family is a member of guanine nucleotide exchange factors (GEFs) that can activate ARF6 specifically in vitro. In this study, we determined the complete primary sequence of mouse EFA6D encoding a protein of 1004 amino acids with a calculated molecular weight of 111,207 Da. In ARF pull-down assay, EFA6D showed a preferential GEF activity toward ARF6. RT-PCR analysis revealed the widespread tissue distribution of EFA6D and the high expression of EFA6A, C and D in the brain. In situ hybridization analysis demonstrated a distinct spatiotemporal expression pattern of EFA6D from those of EFA6A and C in mouse brain. Furthermore, immunoblot analysis revealed that EFA6D was highly concentrated in the postsynaptic density fraction. These findings suggest differential spatiotemporal regulation of ARF6 by three members of the EFA6 family in the brain.
Collapse
Affiliation(s)
- Hiroyuki Sakagami
- Division of Histology, Department of Cell Biology, Graduate School of Medicine, Tohoku University, Sendai 980-8574, Japan.
| | | | | | | | | | | | | |
Collapse
|
132
|
Ye K. PIKE GTPase-mediated nuclear signalings promote cell survival. Biochim Biophys Acta Mol Cell Biol Lipids 2006; 1761:570-6. [PMID: 16567124 DOI: 10.1016/j.bbalip.2006.02.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2005] [Revised: 02/19/2006] [Accepted: 02/20/2006] [Indexed: 10/24/2022]
Abstract
The nuclear GTPase PIKE (PI 3-kinase Enhancer) binds PI 3-kinase and enhances it lipid kinase activity. PIKE predominantly distributes in the brain, and nerve growth factor stimulation triggers PIKE activation by provoking nuclear translocation of PLC-gamma1, which acts as a physiologic guanine nucleotide exchange factor (GEF) for PIKE through its SH3 domain. PIKE contains GTPase and ArfGAP domains, which are separated by a PH domain. C-terminal ArfGAP domain activates its internal GTPase activity, and this process is regulated by the interaction between phosphatidylinositols and PH domain. PI 3-kinase occurs in the nuclei of a broad range of cell types, and various stimuli elicit its nuclear translocation. The nuclei from NGF-treated PC12 cells are resistant to DNA fragmentation initiated by activated cell-free apoptosome, for which PIKE/nuclear PI 3-kinase signaling through nuclear PI(3,4,5)P(3) and Akt plays an essential role. As a nuclear receptor for PI(3,4,5)P(3,) B23 binds to PI(3,4,5)P(3) in an NGF-dependent way. The PI(3,4,5)P(3)/B23 complex inhibits DNA fragmentation activity of CAD. Nuclear Akt regulation of apoptosis is dependent on its phosphorylation of key substrates in the nucleus, but the identities of these substrates are unknown. Identification of its nuclear substrates will further our understanding of the physiological roles of nuclear PI 3-kinase/Akt signaling.
Collapse
Affiliation(s)
- Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
133
|
Madziva MT, Birnbaumer M. A Role for ADP-ribosylation Factor 6 in the Processing of G-protein-coupled Receptors. J Biol Chem 2006; 281:12178-86. [PMID: 16497672 DOI: 10.1074/jbc.m601357200] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
After agonist-induced internalization, the vasopressin V2 receptor (V2R) does not recycle to the plasma membrane. The ADP-ribosylation factor (ARF) proteins initiate vesicular intracellular traffic by promoting the recruitment of adaptor proteins; thus, we sought to determine whether ARF6 could promote V2R recycling. Neither the agonist-induced internalization nor the recycling of the V2R was regulated by ARF6, but a constitutively active mutant of ARF6 reduced cell-surface V2Rs 10-fold in the absence of agonist treatment. Visualization of the ARF6 mutant-expressing cells revealed a vacuolar-staining pattern of the V2R instead of the normal plasma membrane expression. Analysis of V2R maturation revealed that reduced cell-surface expression was due to the diminished ability of the newly synthesized receptor to migrate from the endoplasmic reticulum to the Golgi network. The same mechanism affected processing of the V1aR and acetylcholine M2 receptors. Therefore, ARF6 controls the exit of the V2 and other receptors from the endoplasmic reticulum in addition to its established role in the trafficking of plasma-membrane-derived vesicles.
Collapse
Affiliation(s)
- Michael T Madziva
- Laboratory of Signal Transduction, NIEHS, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | |
Collapse
|
134
|
Krauss M, Haucke V. Functional assay of effectors of ADP ribosylation factor 6 during clathrin/AP-2 coat recruitment to membranes. Methods Enzymol 2006; 404:388-98. [PMID: 16413285 DOI: 10.1016/s0076-6879(05)04034-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
Abstract
Arf proteins play pivotal roles in membrane traffic, cell signaling, and actin cytoskeletal rearrangements. We describe here methods to functionally analyze interacting partner proteins of recombinantly produced N-myristoylated Arf6. Combined evidence from affinity purification and chemical crosslinking experiments, in vitro recruitment assays, and the analysis of lipid kinase activities indicates that Arf6-GTP facilitates clathrin/AP-2 recruitment to synaptic membranes by direct binding and activation of the brain-specific phosphatidylinositol 4-phosphate 5-kinase type Igamma (PIPK Igamma). These methods shall help to mechanistically dissect the role of Arf6 in regulating exo-endocytic vesicle cycling at synapses and in related membrane trafficking events.
Collapse
Affiliation(s)
- Michael Krauss
- Department of Biochemistry II, Center for Biochemistry and Molecular Cell Biology, University of Göttingen, Germany
| | | |
Collapse
|
135
|
Aikawa Y, Martin TFJ. ADP-ribosylation factor 6 regulation of phosphatidylinositol-4,5-bisphosphate synthesis, endocytosis, and exocytosis. Methods Enzymol 2006; 404:422-31. [PMID: 16413288 DOI: 10.1016/s0076-6879(05)04037-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Unlike other members of the ADP-ribosylation factor (ARF) family, Arf6 is localized to the plasma membrane and endosomes, and regulates membrane traffic from and into the plasma membrane. Arf6 regulates a clathrin-independent endocytic membrane recycling pathway in nonpolarized cells and clathrin-dependent endocytosis in polarized cells. It also regulates recycling endosome traffic back to the plasma membrane as well as dense-core vesicle exocytosis in neuroendocrine cells. A key effector for Arf6 is phosphatidylinositol 4-monophosphate 5-kinase, which catalyzes plasma membrane synthesis of phosphatidylinositol-4,5-bisphosphate (PIP2), a common required cofactor for several endocytic and exocytic membrane trafficking pathways. Long-term expression of a constitutively active Arf6 mutant in cells can lead to the depletion of PIP2 from the plasma membrane, its accumulation in intracellular vacuoles, and the inhibition of PIP2-dependent membrane trafficking at the plasma membrane.
Collapse
Affiliation(s)
- Yoshikatsu Aikawa
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences at Kagawa, Tokushima-bunri University, Japan
| | | |
Collapse
|
136
|
Clarke M, Ewart MA, Santy LC, Prekeris R, Gould GW. ACRP30 is secreted from 3T3-L1 adipocytes via a Rab11-dependent pathway. Biochem Biophys Res Commun 2006; 342:1361-7. [PMID: 16516854 DOI: 10.1016/j.bbrc.2006.02.102] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2006] [Accepted: 02/12/2006] [Indexed: 01/06/2023]
Abstract
Adipocytes are now known to secrete a range of adipokines that exhibit distinct biological functions. Here, we sought to understand the secretory pathways utilised by ACRP30 to the surface of adipocytes. We find that ACRP30 overlaps with adipsin in intracellular compartments distinct from Glut4, but nonetheless exhibits insulin-stimulated secretion from cells. Both adipsin and ACRP30 overlap with transferrin receptor-positive membranes, implying that the pathway of secretion involves the transferrin receptor-positive endosomal system. Consistent with this, we show that ablation of endosomes significantly inhibited the secretion of ACRP30, as did treatment of cells with Brefeldin A. In order to further probe the role of recycling endosomes on the secretion of ACRP30, we over-expressed a mutant form of Rab11, Rab11-S25N, in 3T3-L1 adipocytes and found that expression of this mutant significantly reduced basal and insulin-stimulated secretion. We also demonstrate that Arf6 also plays a role in the secretion of ACRP30. Collectively, these data implicate both Arf6 and Rab11 as crucial mediators of constitutive and insulin-stimulated secretion of ACRP30 and further suggest that recycling endosomes may play a central role in this process.
Collapse
Affiliation(s)
- Mairi Clarke
- Henry Wellcome Laboratory of Cell Biology, Division of Biochemistry and Molecular Biology, Faculty of Biomedical and Life Sciences, University of Glasgow, Davidson Building, Glasgow G12 8QQ, Scotland, UK
| | | | | | | | | |
Collapse
|
137
|
Citterio C, Jones HD, Pacheco-Rodriguez G, Islam A, Moss J, Vaughan M. Effect of protein kinase A on accumulation of brefeldin A-inhibited guanine nucleotide-exchange protein 1 (BIG1) in HepG2 cell nuclei. Proc Natl Acad Sci U S A 2006; 103:2683-8. [PMID: 16467138 PMCID: PMC1413798 DOI: 10.1073/pnas.0510571103] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Brefeldin A-inhibited guanine nucleotide-exchange proteins, BIG1 and BIG2, are activators of ADP-ribosylation factor GTPases that are essential for regulating vesicular traffic among intracellular organelles. Biochemical analyses and immunofluorescence microscopy demonstrated BIG1 in nuclei as well as membranes and cytosol of serum-starved HepG2 cells. Within 20 min after addition of 8-Br-cAMP, BIG1 accumulated in nuclei, and this effect was blocked by protein kinase A (PKA) inhibitors H-89 and PKI, suggesting a dependence on PKA-catalyzed phosphorylation. BIG2 localization was not altered by cAMP, nor did BIG2 small interfering RNA influence nuclear accumulation of BIG1 induced by cAMP. Mutant BIG1 (S883A) in which Ala replaced Ser-883, a putative PKA phosphorylation site, did not move to the nucleus with cAMP addition, whereas replacement with Asp (S883D) resulted in nuclear accumulation of BIG1 without or with cAMP exposure, consistent with the mechanistic importance of a negative charge at that site. Mutation (712KPK714) of the nuclear localization signal inhibited BIG1 accumulation in nuclei, and PKA-catalyzed phosphorylation of S883, although necessary, was not sufficient for nuclear accumulation, as shown by the double mutation S883D/nuclear localization signal. A role for microtubules in cAMP-induced translocation of BIG1 is inferred from its inhibition by nocodazole. Thus, two more critical elements of BIG1 molecular structure were identified, as well as the potential function of microtubules in a novel PKA effect on BIG1 translocation.
Collapse
Affiliation(s)
- Carmen Citterio
- Pulmonary–Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Heather D. Jones
- Pulmonary–Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Gustavo Pacheco-Rodriguez
- Pulmonary–Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Aminul Islam
- Pulmonary–Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Joel Moss
- Pulmonary–Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Martha Vaughan
- Pulmonary–Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
138
|
Shmuel M, Santy LC, Frank S, Avrahami D, Casanova JE, Altschuler Y. ARNO through its coiled-coil domain regulates endocytosis at the apical surface of polarized epithelial cells. J Biol Chem 2006; 281:13300-13308. [PMID: 16484220 DOI: 10.1074/jbc.m513723200] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
ARNO is a guanine-nucleotide exchange protein for the ARF family of GTPases. Here we show that in polarized epithelial cells, ARNO is localized exclusively to the apical plasma membrane, where it regulates endocytosis. Expression of ARNO stimulates apical endocytosis of the polymeric immunoglobulin receptor, and coexpression of ARF6 with ARNO leads to a synergistic stimulation of apical endocytosis. Expression of a dominant negative ARF6 mutant, ARF6-T27N, antagonizes this stimulatory effect. Deletion of the N-terminal coiled-coil (CC) domain of ARNO causes the mutant ARNO to localize to both the apical and basolateral plasma membranes. Expression of the CC domain alone abolishes ARNO-induced apical endocytosis as well as co-localization of IgA-receptor complexes with ARNO and clathrin. These results suggest that the CC domain contributes to the specificity of apical localization of ARNO through association with components of the apical plasma membrane. We conclude that ARNO acts together with ARF6 to regulate apical endocytosis.
Collapse
Affiliation(s)
- Miriam Shmuel
- Department of Pharmacology, School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Lorraine C Santy
- Department of Pharmacology, School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Scott Frank
- Department of Pharmacology, School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Dana Avrahami
- Department of Pharmacology, School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - James E Casanova
- Department of Cell Biology, University of Virginia Health Sciences Center, Charlottesville, Virginia 22908
| | - Yoram Altschuler
- Department of Pharmacology, School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel.
| |
Collapse
|
139
|
Meyer MZ, Déliot N, Chasserot-Golaz S, Premont RT, Bader MF, Vitale N. Regulation of neuroendocrine exocytosis by the ARF6 GTPase-activating protein GIT1. J Biol Chem 2006; 281:7919-26. [PMID: 16439353 DOI: 10.1074/jbc.m600081200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neuroendocrine cells release hormones and neuropeptides by exocytosis, a highly regulated process in which secretory granules fuse with the plasma membrane to release their contents in response to a calcium trigger. Using chromaffin and PC12 cells, we have recently described that the granule-associated GTPase ARF6 plays a crucial role in exocytosis by activating phospholipase D1 at the plasma membrane and, presumably, promoting the fusion reaction between the two membrane bilayers. ARF6 is activated by the nucleotide exchange factor ARNO following docking of granules to the plasma membrane. We show here that GIT1, a GTPase-activating protein stimulating GTP hydrolysis on ARF6, is the second molecular partner that turns over the GDP/GTP cycle of ARF6 during cell stimulation. Western blot and immunofluorescence experiments indicated that GIT1 is cytosolic in resting cells but is recruited to the plasma membrane in stimulated cells, where it co-localizes with ARF6 at the granule docking sites. Over-expression of wild-type GIT1 inhibits growth hormone secretion from PC12 cells; this inhibitory effect was not observed in cells expressing a GIT1 mutant impaired in its ARF-GTPase-activating protein (GAP) activity or in cells expressing other ARF6-GAPs. Conversely reduction of GIT1 by RNA interference increased the exocytotic activity. Using a real time assay for individual chromaffin cells, we found that microinjection of GIT1 strongly reduced the number of exocytotic events. These results provide the first evidence that GIT1 plays a function in calcium-regulated exocytosis in neuroendocrine cells. We propose that GIT1 represents part of the pathway that inactivates ARF6-dependent reactions and thereby negatively regulates and/or terminates exocytotic release.
Collapse
Affiliation(s)
- Maria Zeniou Meyer
- Department of Neurotransmission and Neuroendocrine Secretion, Institut des Neurosciences Cellulaires et Intégratives UMR-7168 CNRS/Université Louis Pasteur, 5 rue Blaise Pascal, 67084 Strasbourg, France
| | | | | | | | | | | |
Collapse
|
140
|
Kellersch B, Kolanus W. Membrane-proximal signaling events in beta-2 integrin activation. Results Probl Cell Differ 2006; 43:245-57. [PMID: 17068975 DOI: 10.1007/400_024] [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: 05/12/2023]
Abstract
In the immune system, integrins have essential roles in leukocyte trafficking and function. These include immune cell attachment to endothelial and antigen-presenting cells, cytotoxicity, and extravasation into tissues. The integrin leukocyte function-associated antigen-1 (LFA-1), which is exclusively expressed on hematopoietic cells, has been intensely studied since this receptor is important for many functions of the immune system. LFA-1 is involved in a) the interaction between T-cells and antigen presenting cells, b) the adhesion of cells to post-capillary high endothelial venules or to activated endothelium at sites of inflammation (extravasation), c) the control of cell differentiation and proliferation, and d) the regulation of T-cell effector functions. Therefore, a precise understanding of the spatial and temporal control of LFA-1 interaction with its cellular counter-receptors, the intercellular adhesion molecules (ICAM) -1, -2 and -3, in the various contexts, is of high interest. LFA-1 mediated adhesion is induced by several extracellular stimuli in different cell types. In T-cells, LFA-1 becomes activated upon signaling from the T-cell receptor (TCR), and upon cytokine and chemokine sensing. Adhesion of monocytes to ICAM-1 is induced by lipopolysaccharide (LPS), a component of the bacterial cell wall. To investigate the regulation of LFA-1 adhesiveness, research has focused on the identification of interaction partners of the intracellular portions of the integrin alpha and beta subunits. This review will highlight recent developments on transmembrane and intracellular signaling proteins, which have been implicated in beta-2 integrin activation.
Collapse
Affiliation(s)
- Bettina Kellersch
- Life and Medical Sciences Institute (LIMES), Molecular Immune and Cell Biology Program Unit, Laboratory of Molecular Immunology, University of Bonn, Germany
| | | |
Collapse
|
141
|
Nie Z, Hirsch DS, Luo R, Jian X, Stauffer S, Cremesti A, Andrade J, Lebowitz J, Marino M, Ahvazi B, Hinshaw JE, Randazzo PA. A BAR Domain in the N Terminus of the Arf GAP ASAP1 Affects Membrane Structure and Trafficking of Epidermal Growth Factor Receptor. Curr Biol 2006; 16:130-9. [PMID: 16431365 DOI: 10.1016/j.cub.2005.11.069] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2005] [Revised: 11/21/2005] [Accepted: 11/23/2005] [Indexed: 11/24/2022]
Abstract
BACKGROUND Arf GAPs are multidomain proteins that function in membrane traffic by inactivating the GTP binding protein Arf1. Numerous Arf GAPs contain a BAR domain, a protein structural element that contributes to membrane traffic by either inducing or sensing membrane curvature. We have examined the role of a putative BAR domain in the function of the Arf GAP ASAP1. RESULTS ASAP1's N terminus, containing the putative BAR domain together with a PH domain, dimerized to form an extended structure that bound to large unilamellar vesicles containing acidic phospholipids, properties that define a BAR domain. A recombinant protein containing the BAR domain of ASAP1, together with the PH and Arf GAP domains, efficiently bent the surface of large unilamellar vesicles, resulting in the formation of tubular structures. This activity was regulated by Arf1*GTP binding to the Arf GAP domain. In vivo, the tubular structures induced by ASAP1 mutants contained epidermal growth factor receptor (EGFR) and Rab11, and ASAP1 colocalized in tubular structures with EGFR during recycling of receptor. Expression of ASAP1 accelerated EGFR trafficking and slowed cell spreading. An ASAP1 mutant lacking the BAR domain had no effect. CONCLUSIONS The N-terminal BAR domain of ASAP1 mediates membrane bending and is necessary for ASAP1 function. The Arf dependence of the bending activity is consistent with ASAP1 functioning as an Arf effector.
Collapse
Affiliation(s)
- Zhongzhen Nie
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Someya A, Moss J, Vaughan M, Nagaoka I. ARF-GEP100, a guanine nucleotide-exchange protein for ADP-ribosylation factor 6, involved in the apoptotic cell death of phagocytes. Inflamm Regen 2006. [DOI: 10.2492/inflammregen.26.107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
143
|
Sakane H, Yamamoto T, Tanaka K. The Functional Relationship between the Cdc50p-Drs2p Putative Aminophospholipid Translocase and the Arf GAP Gcs1p in Vesicle Formation in the Retrieval Pathway from Yeast Early Endosomes to the TGN. Cell Struct Funct 2006; 31:87-108. [PMID: 17062999 DOI: 10.1247/csf.06021] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Drs2p, the catalytic subunit of the Cdc50p-Drs2p putative aminophospholipid translocase, has been implicated in conjunction with the Arf1 signaling pathway in the formation of clathrin-coated vesicles (CCVs) from the TGN. Herein, we searched for Arf regulator genes whose mutations were synthetically lethal with cdc50Delta, and identified the Arf GAP gene GCS1. Most of the examined transport pathways in the Cdc50p-depleted gcs1Delta mutant were nearly normal, including endocytic transport to vacuoles, carboxypeptidase Y sorting, and the processing and secretion of invertase. In contrast, this mutant exhibited severe defects in the early endosome-to-TGN transport pathway; proteins that are transported via this pathway, such as the v-SNARE Snc1p, the t-SNARE Tlg1p, and the chitin synthase III subunit Chs3p, accumulated in TGN-independent aberrant membrane structures. We extended our analyses to clathrin adaptors, and found that Gga1p/Gga2p and AP-1 were also involved in this pathway. The Cdc50p-depleted gga1Delta gga2Delta mutant and the gcs1Delta apl2Delta (the beta1 subunit of AP-1) mutant exhibited growth defects and intracellular Snc1p-containing membranes accumulated in these cells. These results suggest that Cdc50p-Drs2p plays an important role in the Arf1p-mediated formation of CCVs for the retrieval pathway from early endosomes to the TGN.
Collapse
Affiliation(s)
- Hiroshi Sakane
- Division of Molecular Interaction, Institute for Genetic Medicine, Hokkaido University Graduate School of Medicine, Kita-ku, Sapporo 060-0815, Japan
| | | | | |
Collapse
|
144
|
Abstract
Eukaryotic cells have systems of internal organelles to synthesize lipids and membrane proteins, to release secreted proteins, to take up nutrients and to degrade membrane-bound and internalized molecules. Proteins and lipids move from organelle to organelle using transport vesicles. The accuracy of this traffic depends upon organelles being correctly recognized. In general, organelles are identified by the activated GTPases and specific lipid species that they display. These short-lived determinants provide organelles with an identity that is both unique and flexible. Recent studies have helped to establish how cells maintain and restrict these determinants and explain how this system is exploited by invading pathogens.
Collapse
Affiliation(s)
- Rudy Behnia
- MRC Laboratory of Molecular Biology, Hills Road, Cambridge CB2 2QH, UK
| | | |
Collapse
|
145
|
Nie Z, Fei J, Premont RT, Randazzo PA. The Arf GAPs AGAP1 and AGAP2 distinguish between the adaptor protein complexes AP-1 and AP-3. J Cell Sci 2005; 118:3555-66. [PMID: 16079295 DOI: 10.1242/jcs.02486] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
ADP ribosylation factors (Arf) regulate membrane trafficking at multiple intracellular sites by recruiting coat proteins to membranes. The site-specific regulation of Arf is thought to be mediated by regulatory proteins including the guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). Here, we test this hypothesis by comparing the site of action of the Arf GAP AGAP2 to the closely related AGAP1. AGAP1 has previously been found to associate with the adaptor protein complex AP-3 and regulate the function of AP-3 endosomes. We found that AGAP2 directly interacted with AP-1. AGAP2 colocalized with AP-1, transferrin receptor and Rab4 on endosomes. Overexpression of AGAP2 changed the intracellular distribution of AP-1 and promoted Rab4-dependent fast recycling of transferrin. Based on these results, we concluded that the closely related Arf GAPs, AGAP1 and AGAP2, distinguish between these related heterotetrameric adaptor protein complexes to specifically regulate AP-3 endosomes and AP-1 recycling endosomes.
Collapse
Affiliation(s)
- Zhongzhen Nie
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
146
|
Choi W, Karim ZA, Whiteheart SW. Arf6 plays an early role in platelet activation by collagen and convulxin. Blood 2005; 107:3145-52. [PMID: 16352809 PMCID: PMC1895749 DOI: 10.1182/blood-2005-09-3563] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Small GTPases play critical roles in hemostasis, though the roster of such molecules in platelets is not complete. In this study, we report the presence of Ras-related GTPases of the ADP-ribosylation factor (Arf) family. Platelets contain Arf1 or 3 and Arf6, with the latter being predominantly membrane associated. Using effector domain pull-down assays, we show, counter to other GTPases, that Arf6-GTP is present in resting platelets and decreases rapidly upon activation with collagen or convulxin. This decrease does not completely rely on secondary agonists (ADP and thromboxane A2) or require integrin signaling. The decrease in free Arf6-GTP temporally precedes activation of Rho family GTPases (RhoA, Cdc42, and Rac1). Using a membrane-permeant, myristoylated peptide, which mimics the N-terminus of Arf6, we show that the Arf6-GTP decrease is essential for collagen- and convulxin-induced aggregation, platelet adherence, and spreading on collagen-coated glass. Treatment with this peptide also affects the activation of Rho family GTPases, but has little effect on RalA and Rap1 or on agonist-induced calcium mobilization. These data show that Arf6 is a key element in activation through GPVI, and is required for activation of the Rho family GTPases and the subsequent cytoskeletal rearrangements needed for full platelet function.
Collapse
Affiliation(s)
- Wangsun Choi
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40536-0509, USA
| | | | | |
Collapse
|
147
|
Bradley RR, Terajima M. Vaccinia virus K1L protein mediates host-range function in RK-13 cells via ankyrin repeat and may interact with a cellular GTPase-activating protein. Virus Res 2005; 114:104-12. [PMID: 16039000 DOI: 10.1016/j.virusres.2005.06.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2004] [Revised: 06/01/2005] [Accepted: 06/01/2005] [Indexed: 11/20/2022]
Abstract
The K1L protein of vaccinia virus is required for its growth in certain cell lines (RK-13 and human). The cowpox host-range protein CP77 has been shown to complement K1L function in RK-13 cells, despite a lack of homology between the two proteins except for ankyrin repeats. We investigated the role of ankyrin repeats of K1L protein in RK-13 cells. The growth of a recombinant vaccinia virus, with K1L gene mutated in the most conserved ankyrin repeat, was severely impaired. Infection with the mutant virus caused shutdown of cellular and viral protein synthesis early in infection. We also investigated the interaction of K1L protein with cellular proteins and found that K1L interacts with the rabbit homologue of human ACAP2, a GTPase-activating protein with ankyrin repeats. Our result suggests the importance of ankyrin repeat for host-range function of K1L in RK-13 cells and identifies ACAP2 as a cellular protein, which may be interacting with K1L.
Collapse
Affiliation(s)
- Ritu R Bradley
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, USA
| | | |
Collapse
|
148
|
Botrugno OA, Paris S, Za L, Gualdoni S, Cattaneo A, Bachi A, de Curtis I. Characterization of the endogenous GIT1-betaPIX complex, and identification of its association to membranes. Eur J Cell Biol 2005; 85:35-46. [PMID: 16373173 DOI: 10.1016/j.ejcb.2005.09.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2005] [Revised: 09/21/2005] [Accepted: 09/22/2005] [Indexed: 12/25/2022] Open
Abstract
G protein-coupled receptor kinase interactors (GITs) are adaptor proteins with ADP-ribosylating factor--GTPase-activating protein (ARF-GAP) activity that form complexes with the p21-activated kinase-interacting exchange factor (PIX) guanine nucleotide exchanging factors for Rac and Cdc42. In this study we have characterized the endogenous GIT1/p95-APP1/Cat1 (GIT1)- PIX complexes in neuronal and non-neuronal cells. In COS7 cells, immunocytochemical analysis shows the localization of endogenous GIT1 in the perinuclear region of the cell, as well as at the cell periphery, where GIT1 co-localizes with filamentous actin. The perinuclear localization of endogenous GIT1 was confirmed in avian fibroblasts. In COS7 cells, immunoprecipitation and microsequencing experiments with either anti-GIT1 or anti-betaPIX antibodies unequivocally show that betaPIX is uniquely associated with GIT1 in lysates from these cells, while GIT2/PKL/p95-APP2/Cat2 (GIT2) is undetectable in the endogenous complexes. Moreover, this analysis demonstrates that betaPIX is the limiting factor for the formation of the endogenous complexes, since a small fraction of GIT1 can be co-immunoprecipitated with most betaPIX from these cells. Saponin treatment of unfixed cells indicates that betaPIX-bound GIT1 is preferentially retained in the saponin-resistant fraction when compared to betaPIX-free GIT1. Moreover, analysis by tissue fractionation shows that a significant fraction of the endogenous GIT1-betaPIX complex is firmly associated to membranes from brain homogenates. Our findings show the specific localization of the complex at intracellular membranes, and indicate a correlation between the association of GIT1 to betaPIX, and the localization of the endogenous complex at membranes.
Collapse
Affiliation(s)
- Oronza A Botrugno
- Cell Adhesion Unit, Department of Molecular Biology and Functional Genomics, San Raffaele Scientific Institute, Via Olgettina 58, I-20132 Milano, Italy
| | | | | | | | | | | | | |
Collapse
|
149
|
Zeniou-Meyer M, Borg JP, Vitale N. Le complexe GIT-PIX : Une plate-forme de régulation des GTPases ARF et Rac/Cdc42. Med Sci (Paris) 2005; 21:849-53. [PMID: 16197902 DOI: 10.1051/medsci/20052110849] [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: 11/14/2022] Open
Abstract
We recently described that the tumor suppressor factor Scribble anchors the PIX exchange factor for Rac/Cdc42 and the ARF-GAP GIT proteins at the plasma membrane. Because it has been postulated that the GIT-PIX proteins dimerize and tightly self-assemble to form a high molecular weight complex, this nexus may be capable of linking together important signalling molecules to control cytosqueleton polymerization and membrane dynamics. To date, most studies that have tempted to unravel the function of these proteins have found their implication in a great variety of cellular functions (receptor recycling, endo-exocytosis, cell migration, synapse formation...) but have mostly neglected to consider the multimeric organization of this hub. There is no doubt that our comprehension of physiopathological disorders such as cancers will be improved when the nature of the complex pathways integrated by the GIT-PIX nodule will be understood.
Collapse
Affiliation(s)
- Maria Zeniou-Meyer
- CNRS UPR-2356, Neurotransmission et sécrétion neuroendocrine, Centre de neurochimie, Strasbourg, France
| | | | | |
Collapse
|
150
|
Dhonukshe P, Kleine-Vehn J, Friml J. Cell polarity, auxin transport, and cytoskeleton-mediated division planes: who comes first? PROTOPLASMA 2005; 226:67-73. [PMID: 16231102 DOI: 10.1007/s00709-005-0104-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2005] [Accepted: 03/30/2005] [Indexed: 05/04/2023]
Abstract
In plants, cell polarity is an issue more recurring than in other systems, because plants, due to their adaptive and flexible development, often change cell polarity postembryonically according to intrinsic cues and demands of the environment. Recent findings on the directional movement of the plant signalling molecule auxin provide a unique connection between individual cell polarity and the establishment of polarity at the tissue, organ, and whole-plant levels. Decisions about the subcellular polar targeting of PIN auxin transport components determine the direction of auxin flow between cells and consequently mediate multiple developmental events. In addition, mutations or chemical interference with PIN-based auxin transport result in abnormal cell divisions. Thus, the complicated links between cell polarity establishment, auxin transport, cytoskeleton, and oriented cell divisions now begin to emerge. Here we review the available literature on the issues of cell polarity in both plants and animals to extend our understanding on the generation, maintenance, and transmission of cell polarity in plants.
Collapse
Affiliation(s)
- Pankaj Dhonukshe
- Zentrum für Molekularbiologie der Pflanzen, Universität Tübingen, Tübingen, Federal Republic of Germany.
| | | | | |
Collapse
|